US20060234376A1 - Repair and regeneration of ocular tissue using postpartum-derived cells - Google Patents

Repair and regeneration of ocular tissue using postpartum-derived cells Download PDF

Info

Publication number
US20060234376A1
US20060234376A1 US11/317,574 US31757405A US2006234376A1 US 20060234376 A1 US20060234376 A1 US 20060234376A1 US 31757405 A US31757405 A US 31757405A US 2006234376 A1 US2006234376 A1 US 2006234376A1
Authority
US
United States
Prior art keywords
cells
cell
protein
derived
postpartum
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/317,574
Inventor
Sanjay Mistry
Darin Messina
Ian Harris
Alexander Harmon
Anthony Kihm
Agnieszka Seyda
Chin-Feng Yi
Anna Gosiewska
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ethicon Inc
Original Assignee
Ethicon Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ethicon Inc filed Critical Ethicon Inc
Priority to US11/317,574 priority Critical patent/US20060234376A1/en
Publication of US20060234376A1 publication Critical patent/US20060234376A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/51Umbilical cord; Umbilical cord blood; Umbilical stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/48Reproductive organs
    • A61K35/50Placenta; Placental stem cells; Amniotic fluid; Amnion; Amniotic stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1808Epidermal growth factor [EGF] urogastrone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1833Hepatocyte growth factor; Scatter factor; Tumor cytotoxic factor II
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1841Transforming growth factor [TGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1858Platelet-derived growth factor [PDGF]
    • A61K38/1866Vascular endothelial growth factor [VEGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1891Angiogenesic factors; Angiogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/204IL-6
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2053IL-8
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/27Growth hormone [GH] (Somatotropin)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/06Free radical scavengers or antioxidants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0605Cells from extra-embryonic tissues, e.g. placenta, amnion, yolk sac, Wharton's jelly
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/32Amino acids
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/34Sugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/44Thiols, e.g. mercaptoethanol
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components
    • C12N2500/95Protein-free medium and culture conditions
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/12Hepatocyte growth factor [HGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/21Chemokines, e.g. MIP-1, MIP-2, RANTES, MCP, PF-4
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/02Coculture with; Conditioned medium produced by embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/03Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from non-embryonic pluripotent stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2509/00Methods for the dissociation of cells, e.g. specific use of enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins

Definitions

  • This invention relates to the field of cell-based or regenerative therapy for ophthalmic diseases and disorders.
  • the invention provides pharmaceutical compositions, devices and methods for the regeneration or repair of cells and tissues of the eye, using postpartum derived cells.
  • the eye can experience numerous diseases and other deleterious conditions that affect its ability to function normally. Many of these conditions are associated with damage or degeneration of specific ocular cells, and tissues made up of those cells. As one example, diseases and degenerative conditions of the optic nerve and retina are the leading causes of blindness throughout the world. Damage or degeneration of the cornea, lens and associated ocular tissues represent another significant cause of vision loss worldwide.
  • the retina contains seven layers of alternating cells and processes that convert a light signal into a neural signal.
  • the retinal photoreceptors and adjacent retinal pigment epithelium (RPE) form a functional unit that, in many disorders, becomes unbalanced due to genetic mutations or environmental conditions (including age). This results in loss of photoreceptors through apoptosis or secondary degeneration, which leads to progressive deterioration of vision and, in some instances, to blindness (for a review, see, e.g., Lund, R. D. et al., 2001, Progress in Retinal and Eye Research 20: 415-449).
  • Two classes of ocular disorders that fall into this pattern are age-related macular degeneration (AMD) and retinitis pigmentosa (RP).
  • AMD age-related macular degeneration
  • RP retinitis pigmentosa
  • AMD AMD is the most common cause of vision loss in the United States in those 50 or older, and its prevalence increases with age.
  • the primary disorder in AMD appears to be due to RPE dysfunction and changes in Bruch's membranes, e.g., lipid deposition, protein cross-linking and decreased permeability to nutrients (see Lund et al., 2001 supra).
  • a variety of elements may contribute to macular degeneration, including genetic makeup, age, nutrition, smoking and exposure to sunlight.
  • RP is mainly considered an inherited disease—over 100 mutations have been associated with photoreceptor loss (see Lund et al., 2001, supra). Though the majority of mutations target photoreceptors, some affect RPE cells directly. Together, these mutations affect such processes as molecular trafficking between photoreceptors and RPE cells and phototransduction, for example.
  • Diabetic retinopathy may be classified as (1) non-proliferative or background retinopathy, characterized by increased capillary permeability, edema, hemorrhage, microaneurysms, and exudates, or 2) proliferative retinopathy, characterized by neovascularization extending from the retina to the vitreous, scarring, fibrous tissue formation, and potential for retinal detachment.
  • CNVM choroidal neovascular membrane
  • Glaucoma a condition of the eye that is made up of a collection of eye diseases that cause vision loss by damage to the optic nerve. Elevated intraocular pressure (IOP) due to inadequate ocular drainage is a primary cause of glaucoma, but it can also develop in the absence of elevated IOP. Glaucoma can develop as the eye ages, or it can occur as the result of an eye injury, inflammation, tumor, or in advanced cases of cataract or diabetes, or it can be caused by certain drugs, such as steroids.
  • IOP intraocular pressure
  • the primary features of the optic neuropathy in glaucoma include characteristic changes in the optic nerve head, a decrease in number of surviving retinal ganglion cells, and loss of vision. It has been proposed that a cascade of events links degeneration of the optic nerve head with the slow death of retinal ganglion cells observed in the disease, and that this cascade of events can be slowed or prevented through the use of neuroprotective agents (Osborne et al., 2003, Eur. J. Ophthalmol. 13 (Supp 3): S19-S26).
  • LEF Limbal epithelial cell deficiency
  • LECD often leads to an imbalance between the corneal epithelium and the conjunctival epithelium in which the cornea is covered by invading conjunctival epithelial cells, which severely compromises the corneal surface and affects visual acuity (Nakamura, T. & Kinoshita, S., 2003. Cornea 22 (Supp. 1): S75-S80).
  • Stem cells are capable of self-renewal and differentiation to generate a variety of mature cell lineages. Transplantation of such cells can be utilized as a clinical tool for reconstituting a target tissue, thereby restoring physiologic and anatomic functionality.
  • the application of stem cell technology is wide-ranging, including tissue engineering, gene therapy delivery, and cell therapeutics, i.e., delivery of biotherapeutic agents to a target location via exogenously supplied living cells or cellular components that produce or contain those agents (For a review, see Tresco, P. A. et al., 2000, Advanced Drug Delivery Reviews 42: 2-37).
  • stem cell technology An obstacle to realization of the therapeutic potential of stem cell technology has been difficulty in obtaining sufficient numbers of stem cells.
  • One source of stem cells is embryonic or fetal tissue. Embryonic stem and progenitor cells have been isolated from a number of mammalian species, including humans, and several such cell types have been shown capable of self-renewal and expansion, as well differentiation into a variety of cell lineages. In animal model systems, embryonic stem cells have been reported to differentiate into a RPE cell phenotype, as well as to enhance the survival of host photoreceptors following transplantation (Haruta, M. et al., 2004, Investig. Ophthalmol. Visual Sci. 45: 1020-1025; Schraermeyer, U. et al., 2001, Cell Transplantation 10: 673-680). But the derivation of stem cells from embryonic or fetal sources has raised many ethical issues that are desirable to avoid by identifying other sources of multipotent or pluripotent cells.
  • Adult tissue also can yield stem cells useful for cell-based ocular therapy.
  • retinal and corneal stem cells themselves may be utilized for cell replacement therapy in the eye.
  • neural stem cells from the hippocampus have been reported to integrate with the host retina, adopting certain neural and glial characteristics (see review of Lund, R. L. et al., 2003, J. Leukocyte Biol. 74: 151-160).
  • Neural stem cells prepared from fetal rat cortex were shown to differentiate along an RPE cell pathway following transplantation into the adult rat subretinal space (Enzmann, V. et al., 2003, Investig. Ophthalmol. Visual Sci. 44: 5417-5422).
  • Bone marrow stem cells have been reported to differentiate into retinal neural cells and photoreceptors following transplantation into host retinas (Tomita, M. et al., 2002, Stem Cells 20: 279-283; Kicic, A. et al., 2003, J. Neurosci. 23: 7742-7749).
  • An ocular surface reconstruction in a rabbit model system, utilizing cultured mucosal epithelial stem cells has also been reported (Nakamura, T. & Kinoshita, S., 2003, supra). While these reports show promise for the use of adult progenitor and stem cells in cell-based therapy for the eye, it must be noted that adult stem cell populations are comparatively rare and are often obtainable only by invasive procedures. Further, adult stem cells may have a more limited ability to expand in culture than do embryonic stem cells.
  • a reliable, well-characterized and plentiful supply of substantially homogeneous populations of such cells would be an advantage in a variety of diagnostic and therapeutic applications in ocular repair and regeneration, including drug screening assays, ex vivo or in vitro trophic support of ocular and other useful cell types, and in vivo cell-based therapy.
  • compositions and methods applicable to cell-based or regenerative therapy for ophthalmic diseases and disorders provides compositions and methods applicable to cell-based or regenerative therapy for ophthalmic diseases and disorders.
  • the invention features pharmaceutical compositions, devices and methods for the regeneration or repair of ocular tissue using postpartum-derived cells.
  • One aspect of the invention features an isolated postpartum-derived cell, derived from human placental or umbilical cord tissue substantially free of blood, wherein the cell is capable of self-renewal and expansion in culture and has the potential to differentiate into a cell of a neural phenotype; wherein the cell requires L-valine for growth and is capable of growth in at least about 5% oxygen.
  • This cell further comprises one or more of the following characteristics: (a) potential for at least about 40 doublings in culture; (b) attachment and expansion on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin, or fibronectin; (c) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; (d) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C; (e) lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; (f) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell
  • the postpartum-derived cell is an umbilicus-derived cell. In other embodiments it is a placenta-derived cell. In specific embodiments, the cell has all identifying features of any one of: cell type PLA 071003 (P8) (ATCC Accession No. PTA-6074); cell type PLA 071003 (P11) (ATCC Accession No. PTA-6075); cell type PLA 071003 (P16) (ATCC Accession No. PTA-6079); cell type UMB 022803 (P7) (ATCC Accession No. PTA-6067); or cell type UMB 022803 (P17) (ATCC Accession No. PTA-6068).
  • postpartum-derived cells are isolated in the presence of one or more enzyme activities comprising metalloprotease activity, mucolytic activity and neutral protease activity.
  • the cells Preferably, the cells have a normal karyotype, which is maintained as the cells are passaged in culture.
  • the postpartum-derived cells comprise each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C and does not comprise any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
  • the population is a substantially homogeneous population of the postpartum-derived cells.
  • the population comprises a clonal cell line of the postpartum-derived cells.
  • the population is a heterogeneous population comprising the postpartum-derived cells and at least one other cell type.
  • the other cell type is an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell or other multipotent or pluripotent stem cell.
  • the cell population is cultured in contact with one or more factors that stimulate stem cell differentiation toward a neural or epithelial lineage.
  • a cell lysate prepared from postpartum-derived cells.
  • the cell lysate may be separated into a membrane enriched fraction and a soluble cell fraction.
  • the invention also features an extracellular matrix produced by the postpartum-derived cells, as well as a conditioned medium in which the cells have been grown.
  • Another aspect of the invention features a method of treating a patient having an ocular degenerative condition, which comprises administering to the patient multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord, in an amount effective to treat the ocular degenerative condition.
  • the cells are postpartum-derived cells, as described above.
  • the ocular degenerative condition is an acute ocular degenerative condition such as brain trauma, optic nerve trauma or ocular lesion. In other embodiments, it is a chronic or progressive degenerative condition, such as macular degeneration, retinitis pigmentosa, diabetic retinopathy, glaucoma or limbal epithelial cell deficiency.
  • the cells are induced in vitro to differentiate into a neural or epithelial lineage cells prior to administration.
  • the cells are administered with at least one other cell type, such as an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell.
  • the other cell type can be administered simultaneously with, or before, or after, the postpartum-derived cells.
  • the cells are administered with at least one other agent, such as a drug for ocular therapy, or another beneficial adjunctive agent such as an anti-inflammatory agent, anti-apoptotic agents, antioxidants or growth factors.
  • the other agent can be administered simultaneously with, or before, or after, the postpartum cells.
  • the cells are administered to the surface of an eye, or they are administered to the interior of an eye or to a location in proximity to the eye, e.g., behind the eye.
  • the cells can be administered through a cannula or from a device implanted in the patient's body within or in proximity to the eye, or they may be administered by implantation of a matrix or scaffold containing the cells.
  • compositions for treating a patient having an ocular degenerative condition comprising a pharmaceutically acceptable carrier and multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord in an amount effective to treat the ocular degenerative condition.
  • the postpartum cells are postpartum-derived cells as described above.
  • the ocular degenerative condition may be an acute, chronic or progressive condition.
  • the composition comprises cells that have been induced in vitro to differentiate into a neural or epithelial lineage cells prior to formulation of the composition.
  • the pharmaceutical composition comprises at least one other cell type, such as an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell.
  • the pharmaceutical composition comprises at least one other agent, such as a drug for treating the ocular degenerative disorder or other beneficial adjunctive agents, e.g., anti-inflammatory agents, anti-apoptotic agents, antioxidants or growth factors.
  • the pharmaceutical compositions are formulated for administration to the surface of an eye. Alternatively, they can be formulated for administration to the interior of an eye or in proximity to the eye (e.g., behind the eye).
  • the compositions also can be formulated as a matrix or scaffold containing the cells.
  • kits for treating a patient having an ocular degenerative condition.
  • the kit comprises a pharmaceutically acceptable carrier, a population of multipotent or pluripotent cells isolated from postpartum placenta or umbilicus, preferably the postpartum-derived cells described above, and instructions for using the kit in a method of treating the patient.
  • the kit may also contain one or more additional components, such as reagents and instructions for culturing the cells, or a population of at least one other cell type, or one or more agents useful in the treatment of an ocular degenerative condition.
  • a method for treating a patient having an ocular degenerative condition comprises administering to the patient a preparation made from multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord, in an amount effective to treat the ocular degenerative condition, wherein the preparation comprises a cell lysate of the cells (or fraction thereof), or a conditioned medium in which the cells were grown, or an extracellular matrix of the cells.
  • the cells are the postpartum-derived cells described above.
  • the invention features a pharmaceutical composition
  • a pharmaceutically acceptable carrier and a preparation made from the postpartum cells, which may be a cell lysate (or fraction thereof) of the postpartum cells, an extracellular matrix of the postpartum cells or a conditioned medium in which the postpartum cells were grown.
  • Kits for practicing this aspect of the invention are also provided. These may include the one or more of a pharmaceutically acceptable carrier or other agent or reagent, one or more of a cell lysate or fraction thereof, an extracellular matrix or a conditioned medium from the postpartum cells, and instructions for use of the kit components.
  • Another aspect of the invention features a method for increasing the survival, growth or activity of cells for transplantation to treat an ocular degenerative disorder.
  • the method comprises co-culturing the cells for transplantation with cultured cells derived from postpartum placental or umbilical tissue, under conditions effective to increase the survival, growth or activity of the cells for transplantation.
  • the postpartum cells are the postpartum-derived cells described above.
  • a kit for practicing the method is also provided.
  • the kit comprises the cultured postpartum cells and instructions for co-culturing the cells for transplantation with the postpartum cells under conditions effective to increase the survival, growth or activity of the cells for transplantation.
  • Stem cells are undifferentiated cells defined by the ability of a single cell both to self-renew, and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute substantially to most, if not all, tissues following injection into blastocysts.
  • Stem cells are classified according to their developmental potential as: (1) totipotent; (2) pluripotent; (3) multipotent; (4) oligopotent; and (5) unipotent.
  • Totipotent cells are able to give rise to all embryonic and extraembryonic cell types.
  • Pluripotent cells are able to give rise to all embryonic cell types.
  • Multipotent cells include those able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem cells (HSC) can produce progeny that include HSC (self-renewal), blood cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood).
  • HSC hematopoietic stem cells
  • Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells; and cells that are unipotent are able to give rise to a single cell lineage (e.g., spermatogenic stem cells).
  • Stem cells are also categorized on the basis of the source from which they may be obtained.
  • An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types.
  • An embryonic stem cell is a pluripotent cell from the inner cell mass of a blastocyst-stage embryo.
  • a fetal stem cell is one that originates from fetal tissues or membranes.
  • a postpartum stem cell is a multipotent or pluripotent cell that originates substantially from extraembryonic tissue available after birth, namely, the placenta and the umbilical cord.
  • Postpartum stem cells may be blood-derived (e.g., as are those obtained from umbilical cord blood) or non-blood-derived (e.g., as obtained from the non-blood tissues of the umbilical cord and placenta).
  • Embryonic tissue is typically defined as tissue originating from the embryo (which in humans refers to the period from fertilization to about six weeks of development. Fetal tissue refers to tissue originating from the fetus, which in humans refers to the period from about six weeks of development to parturition. Extraembryonic tissue is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryonic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta (which itself forms from the chorion and the maternal decidua basalis).
  • Differentiation is the process by which an unspecialized (“uncommitted”) or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example.
  • a differentiated cell is one that has taken on a more specialized (“committed”) position within the lineage of a cell.
  • the term committed, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type.
  • De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell.
  • the lineage of a cell defines the heredity of the cell, i.e. which cells it came from and what cells it can give rise to.
  • the lineage of a cell places the cell within a hereditary scheme of development and differentiation.
  • a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors.
  • stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells.
  • this broad definition of progenitor cell may be used.
  • a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types. This type of progenitor cell is generally not able to self-renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a non-renewing progenitor cell or as an intermediate progenitor or precursor cell.
  • the phrase differentiates into an ocular lineage or phenotype refers to a cell that becomes partially or fully committed to a specific ocular phenotype, including without limitation, retinal and corneal stem cells, pigment epithelial cells of the retina and iris, photoreceptors, retinal ganglia and other optic neural lineages (e.g., retinal glia, microglia, astrocytes, Mueller cells), cells forming the crystalline lens, and epithelial cells of the sclera, cornea, limbus and conjunctiva.
  • retinal and corneal stem cells pigment epithelial cells of the retina and iris, photoreceptors, retinal ganglia and other optic neural lineages (e.g., retinal glia, microglia, astrocytes, Mueller cells), cells forming the crystalline lens, and epithelial cells of the sclera, cornea, limbus and conjunctiva.
  • the phrase differentiates into a neural lineage or phenotype refers to a cell that becomes partially or fully committed to a specific neural phenotype of the CNS or PNS, i.e., a neuron or a glial cell, the latter category including without limitation astrocytes, oligodendrocytes, Schwann cells and microglia.
  • the cells exemplified herein and preferred for use in present invention are generally referred to as postpartum-derived cells (or PPDCs). They also may sometimes be referred to more specifically as umbilicus-derived cells or placenta-derived cells (UDCs or PDCs).
  • the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense.
  • the term derived is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a Growth Medium to expand the population and/or to produce a cell line).
  • the in vitro manipulations of umbilical stem cells and the unique features of the umbilicus-derived cells of the present invention are described in detail below.
  • Cells isolated from postpartum placenta and umbilicus by other means are also considered suitable for use in the present invention. These other cells are referred to herein as postpartum cells (rather than postpartum-derived cells).
  • Cell culture refers generally to cells taken from a living organism and grown under controlled condition (“in culture” or “cultured”).
  • a primary cell culture is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture.
  • Cells are expanded in culture when they are placed in a Growth Medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells.
  • the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as doubling time.
  • a cell line is a population of cells formed by one or more subcultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been passaged. A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture.
  • the primary culture i.e., the first culture following the isolation of cells from tissue, is designated P0. Following the first subculture, the cells are described as a secondary culture (P1 or passage 1).
  • the cells After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number.
  • the expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and time between passaging.
  • a conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide trophic support to other cells. Such trophic factors include, but are not limited to hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, and granules.
  • the medium containing the cellular factors is the conditioned medium.
  • a trophic factor is defined as a substance that promotes survival, growth, differentiation, proliferation and/or maturation of a cell, or stimulates increased activity of a cell.
  • the interaction between cells via trophic factors may occur between cells of different types. Cell interaction by way of trophic factors is found in essentially all cell types, and is a particularly significant means of communication among neural cell types.
  • Trophic factors also can function in an autocrine fashion, i.e., a cell may produce trophic factors that affect its own survival, growth, differentiation, proliferation and/or maturation.
  • senescence also replicative senescence or cellular senescence refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's limit).
  • cellular senescence was first described using fibroblast-like cells, most normal human cell types that can be grown successfully in culture undergo cellular senescence.
  • the in vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide).
  • Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone. Thus, cells made quiescent by removing essential growth factors are able to resume growth and division when the growth factors are re-introduced, and thereafter carry out the same number of doublings as equivalent cells grown continuously. Similarly, when cells are frozen in liquid nitrogen after various numbers of population doublings and then thawed and cultured, they undergo substantially the same number of doublings as cells maintained unfrozen in culture. Senescent cells are not dead or dying cells; they are actually resistant to programmed cell death (apoptosis), and have been maintained in their nondividing state for as long as three years. These cells are very much alive and metabolically active, but they do not divide. The nondividing state of senescent cells has not yet been found to be reversible by any biological, chemical, or viral agent.
  • ocular, ophthalmic and optic are used interchangeably herein to define “of, or about, or related to the eye.”
  • ocular degenerative condition is an inclusive term encompassing acute and chronic conditions, disorders or diseases of the eye, inclusive of the neural connection between the eye and the brain, involving cell damage, degeneration or loss.
  • An ocular degenerative condition may be age-related, or it may result from injury or trauma, or it may be related to a specific disease or disorder.
  • Acute ocular degenerative conditions include, but are not limited to, conditions associated with cell death or compromise affecting the eye including conditions arising from cerebrovascular insufficiency, focal or diffuse brain trauma, diffuse brain damage, infection or inflammatory conditions of the eye, retinal tearing or detachment, intra-ocular lesions (contusion penetration, compression, laceration) or other physical injury (e.g., physical or chemical burns).
  • Chronic ocular degenerative conditions include, but are not limited to, retinopathies and other retinal/macular disorders such as retinitis pigmentosa (RP), age-related macular degeneration (AMD), choroidal neovascular membrane (CNVM); retinopathies such as diabetic retinopathy, occlusive retinopathy, sickle cell retinopathy and hypertensive retinopathy, central retinal vein occlusion, stenosis of the carotid artery, optic neuropathies such as glaucoma and related syndromes; disorders of the lens and outer eye, e.g., limbal stem cell deficiency (LSCD), also referred to as limbal epithelial cell deficiency (LECD), such as occurs in chemical or thermal injury, Steven-Johnson syndrome, contact lens-induced keratopathy, ocular cicatricial pemphigoid, congenital diseases of aniridia or ectodermal dysplasia
  • RP
  • treating (or treatment of) an ocular degenerative condition refers to ameliorating the effects of, or delaying, halting or reversing the progress of, or delaying or preventing the onset of, an ocular degenerative condition as defined herein.
  • an effective amount refers to a concentration or amount of a reagent or pharmaceutical composition, such as a growth factor, differentiation agent, trophic factor, cell population or other agent, that is effective for producing an intended result, including cell growth and/or differentiation in vitro or in vivo, or treatment of ocular degenerative conditions, as described herein.
  • a reagent or pharmaceutical composition such as a growth factor, differentiation agent, trophic factor, cell population or other agent, that is effective for producing an intended result, including cell growth and/or differentiation in vitro or in vivo, or treatment of ocular degenerative conditions, as described herein.
  • growth factors an effective amount may range from about 1 nanogram/milliliter to about 1 microgram/milliliter.
  • PPDCs as administered to a patient in vivo, an effective amount may range from as few as several hundred or fewer to as many as several million or more. In specific embodiments, an effective amount may range from 10 3 -10 11 , more specifically at least about 10 4 cells.
  • the number of cells to be administered will vary depending on the specifics of the disorder to be treated, including but not limited to size or total volume/surface area to be treated, as well as proximity of the site of administration to the location of the region to be treated, among other factors familiar to the medicinal biologist.
  • effective period and effective conditions refer to a period of time or other controllable conditions (e.g., temperature, humidity for in vitro methods), necessary or preferred for an agent or pharmaceutical composition to achieve its intended result.
  • controllable conditions e.g., temperature, humidity for in vitro methods
  • patient or subject refers to animals, including mammals, preferably humans, who are treated with the pharmaceutical compositions or in accordance with the methods described herein.
  • pharmaceutically acceptable carrier refers to reagents, cells, compounds, materials, compositions, and/or dosage forms that are not only compatible with the cells and other agents to be administered therapeutically, but also are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other complication commensurate with a reasonable benefit/risk ratio.
  • autologous transfer, autologous transplantation, autograft and the like refer to treatments wherein the cell donor is also the recipient of the cell replacement therapy.
  • allogeneic transfer, allogeneic transplantation, allograft and the like refer to treatments wherein the cell donor is of the same species as the recipient of the cell replacement therapy, but is not the same individual.
  • a cell transfer in which the donor's cells and have been histocompatibly matched with a recipient is sometimes referred to as a syngeneic transfer.
  • xenogeneic transfer, xenogeneic transplantation, xenograft and the like refer to treatments wherein the cell donor is of a different species than the recipient of the cell replacement therapy.
  • Ocular degenerative conditions which encompass acute, chronic and progressive disorders and diseases having divergent causes, have as a common feature the dysfunction or loss of a specific or vulnerable group of ocular cells. This commonality enables development of similar therapeutic approaches for the repair or regeneration of vulnerable, damaged or lost ocular tissue, one of which is cell-based therapy.
  • Development of cell therapy for ocular degenerative conditions heretofore has been limited to a comparatively few types of stem or progenitor cells, including ocular-derived stem cells themselves (e.g., retinal and corneal stem cells), embryonic stem cells and a few types of adult stem or progenitor cells (e.g., neural, mucosal epithelial and bone marrow stem cells).
  • the present inventors have identified a significant new source of stem cells for this purpose, namely, cells isolated from the postpartum placenta and umbilical cord. Accordingly, in its various embodiments described herein, the present invention features methods and pharmaceutical compositions for repair and regeneration of ocular tissues, which utilize progenitor cells and cell populations isolated from postpartum tissues.
  • the invention is applicable to any ocular degenerative condition, but is expected to be particularly suitable for a number of ocular disorders for which treatment or cure heretofore has been difficult or unavailable. These include, without limitation, age-related macular degeneration, retinitis pigmentosa, diabetic and other retinopathies, glaucoma and other optic neuropathies, and disorders associated with limbal stem cell deficiency.
  • the invention utilizes postpartum-derived cells (PPDCs) as defined above, which are derived from placental or umbilical cord tissue that has been rendered substantially free of blood, preferably in accordance with the method set forth below.
  • PPDCs postpartum-derived cells
  • the PPDCs are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes.
  • Certain embodiments features populations comprising such cells, pharmaceutical compositions comprising the cells or components or products thereof, and methods of using the pharmaceutical compositions for treatment of patients with acute or chronic ocular degenerative conditions.
  • the postpartum-derived cells have been characterized by their growth properties in culture, by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
  • a mammalian placenta and umbilical cord are recovered upon or shortly after termination of either a full-term or pre-term pregnancy, for example, after expulsion after birth.
  • the postpartum tissue may be transported from the birth site to a laboratory in a sterile container such as a flask, beaker, culture dish, or bag.
  • the container may have a solution or medium, including but not limited to a salt solution, such as, for example, Dulbecco's Modified Eagle's Medium (DMEM) or phosphate buffered saline (PBS), or any solution used for transportation of organs used for transplantation, such as University of Wisconsin solution or perfluorochemical solution.
  • DMEM Dulbecco's Modified Eagle's Medium
  • PBS phosphate buffered saline
  • antibiotic and/or antimycotic agents such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin, may be added to the medium or buffer.
  • the postpartum tissue may be rinsed with an anticoagulant solution such as heparin-containing solution. It is preferable to keep the tissue at about 4-10° C. prior to extraction of PPDCs. It is even more preferable that the tissue not be frozen prior to extraction of PPDCs.
  • Isolation of PPDCs preferably occurs in an aseptic environment.
  • the umbilical cord may be separated from the placenta by means known in the art. Alternatively, the umbilical cord and placenta are used without separation. Blood and debris are preferably removed from the postpartum tissue prior to isolation of PPDCs.
  • the postpartum tissue may be washed with buffer solution, such as but not limited to phosphate buffered saline.
  • the wash buffer also may comprise one or more antimycotic and/or antibiotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin.
  • Postpartum tissue comprising a whole placenta or a fragment or section thereof is disaggregated by mechanical force (mincing or shear forces).
  • the isolation procedure also utilizes an enzymatic digestion process.
  • Many enzymes are known in the art to be useful for the isolation of individual cells from complex tissue matrices to facilitate growth in culture. Ranging from weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin), such enzymes are available commercially.
  • a nonexhaustive list of enzymes compatible herewith includes mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and deoxyribonucleases.
  • enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities.
  • collagenases are known to be useful for isolating various cells from tissues.
  • Deoxyribonucleases can digest single-stranded DNA and can minimize cell-clumping during isolation.
  • Preferred methods involve enzymatic treatment with for example collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral protease dispase are used in the dissociating step. More preferred are those methods which employ digestion in the presence of at least one collagenase from Clostridium histolyticum , and either of the protease activities, dispase and thermolysin. Still more preferred are methods employing digestion with both collagenase and dispase enzyme activities. Also preferred are methods which include digestion with a hyaluronidase activity in addition to collagenase and dispase activities.
  • enzyme treatments are known in the art for isolating cells from various tissue sources.
  • the LIBERASE Blendzyme (Roche) series of enzyme combinations are suitable for use in the instant methods.
  • Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources.
  • the skilled artisan is also well-equipped to assess new, or additional enzymes or enzyme combinations for their utility in isolating the cells of the invention.
  • Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer.
  • the tissue is incubated at 37° C. during the enzyme treatment of the dissociation step.
  • postpartum tissue is separated into sections comprising various aspects of the tissue, such as neonatal, neonatal/maternal, and maternal aspects of the placenta, for instance.
  • the separated sections then are dissociated by mechanical and/or enzymatic dissociation according to the methods described herein.
  • Cells of neonatal or maternal lineage may be identified by any means known in the art, for example, by karyotype analysis or in situ hybridization for a Y chromosome.
  • Isolated cells or postpartum tissue from which PPDCs grow out may be used to initiate, or seed, cell cultures. Isolated cells are transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen (native, denatured or crosslinked), gelatin, fibronectin, and other extracellular matrix proteins.
  • extracellular matrix or ligands such as laminin, collagen (native, denatured or crosslinked), gelatin, fibronectin, and other extracellular matrix proteins.
  • PPDCs are cultured in any culture medium capable of sustaining growth of the cells such as, but not limited to, DMEM (high or low glucose), advanced DMEM, DMEM/MCDB 201, Eagle's basal medium, Ham's F10 medium (F10), Ham's F-12 medium (F12), Iscove's modified Dulbecco's medium, Mesenchymal Stem Cell Growth Medium (MSCGM), DMEM/F12, RPMI 1640, and CELL-GRO-FREE.
  • DMEM high or low glucose
  • advanced DMEM high or low glucose
  • DMEM/MCDB 201 Eagle's basal medium
  • Ham's F10 medium (F10) Ham's F-12 medium (F12)
  • Iscove's modified Dulbecco's medium Iscove's modified Dulbecco's medium
  • MSCGM Mesenchymal Stem Cell Growth Medium
  • DMEM/F12 RPMI 1640
  • CELL-GRO-FREE CELL
  • the culture medium may be supplemented with one or more components including, for example, fetal bovine serum (FBS), preferably about 2-15% (v/v); equine serum (ES); human serum (HS); beta-mercaptoethanol (BME or 2-ME), preferably about 0.001% (v/v); one or more growth factors, for example, platelet-derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), leukocyte inhibitory factor (LIF) and erythropoietin; amino acids, including L-valine; and one or more antibiotic and/or antimycotic agents to control microbial contamination, such as, for example, penicillin G, streptomycin sulfate, amphotericin B, gentamicin, and nystatin, either alone or in combination.
  • the culture medium preferably comprises Growth Medium (DMEM-low glucose, serum, BME
  • the cells are seeded in culture vessels at a density to allow cell growth.
  • the cells are cultured at about 0 to about 5 percent by volume CO 2 in air.
  • the cells are cultured at about 2 to about 25 percent O 2 in air, preferably about 5 to about 20 percent O 2 in air.
  • the cells preferably are cultured at about 25 to about 40° C. and more preferably are cultured at 37° C.
  • the cells are preferably cultured in an incubator.
  • the medium in the culture vessel can be static or agitated, for example, using a bioreactor.
  • PPDCs preferably are grown under low oxidative stress (e.g., with addition of glutathione, Vitamin C, Catalase, Vitamin E, N-Acetylcysteine). “Low oxidative stress”, as used herein, refers to conditions of no or minimal free radical damage to the cultured cells.
  • PPDCs After culturing the isolated cells or tissue fragments for a sufficient period of time, PPDCs will have grown out, either as a result of migration from the postpartum tissue or cell division, or both.
  • PPDCs are passaged, or removed to a separate culture vessel containing fresh medium of the same or a different type as that used initially, where the population of cells can be mitotically expanded.
  • the cells of the invention may be used at any point between passage 0 and senescence.
  • the cells preferably are passaged between about 3 and about 25 times, more preferably are passaged about 4 to about 12 times, and preferably are passaged 10 or 11 times. Cloning and/or subcloning may be performed to confirm that a clonal population of cells has been isolated.
  • the different cell types present in postpartum tissue are fractionated into subpopulations from which the PPDCs can be isolated.
  • This may be accomplished using standard techniques for cell separation including, but not limited to, enzymatic treatment to dissociate postpartum tissue into its component cells, followed by cloning and selection of specific cell types, for example but not limited to selection based on morphological and/or biochemical markers; selective growth of desired cells (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; differential adherence properties of the cells in the mixed population; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation); unit gravity separation; countercurrent distribution; electrophoresis; and fluorescence activated cell sorting (FACS).
  • FACS fluorescence activated cell sorting
  • the culture medium is changed as necessary, for example, by carefully aspirating the medium from the dish, for example, with a pipette, and replenishing with fresh medium. Incubation is continued until a sufficient number or density of cells accumulate in the dish.
  • the original explanted tissue sections may be removed and the remaining cells trypsinized using standard techniques or using a cell scraper. After trypsinization, the cells are collected, removed to fresh medium and incubated as above.
  • the medium is changed at least once at approximately 24 hours post-trypsinization to remove any floating cells. The cells remaining in culture are considered to be PPDCs.
  • PPDCs may be cryopreserved. Accordingly, in a preferred embodiment described in greater detail below, PPDCs for autologous transfer (for either the mother or child) may be derived from appropriate postpartum tissues following the birth of a child, then cryopreserved so as to be available in the event they are later needed for transplantation.
  • PPDCs may be characterized, for example, by growth characteristics (e.g., population doubling capability, doubling time, passages to senescence), karyotype analysis (e.g., normal karyotype; maternal or neonatal lineage), flow cytometry (e.g., FACS analysis), immunohistochemistry and/or immunocytochemistry (e.g., for detection of epitopes), gene expression profiling (e.g., gene chip arrays; polymerase chain reaction (for example, reverse transcriptase PCR, real time PCR, and conventional PCR)), protein arrays, protein secretion (e.g., by plasma clotting assay or analysis of PDC-conditioned medium, for example, by Enzyme Linked ImmunoSorbent Assay (ELISA)), mixed lymphocyte reaction (e.g., as measure of stimulation of PBMCs), and/or other methods known in the art.
  • growth characteristics e.g., population doubling capability, doubling time, passages to senescence
  • Examples of PPDCs derived from placental tissue were deposited with the American Type Culture Collection (ATCC, Manassas, Va.) and assigned ATCC Accession Numbers as follows: (1) strain designation PLA 071003 (P8) was deposited Jun. 15, 2004 and assigned Accession No. PTA-6074; (2) strain designation PLA 071003 (P111) was deposited Jun. 15, 2004 and assigned Accession No. PTA-6075; and (3) strain designation PLA 071003 (P16) was deposited Jun. 16, 2004 and assigned Accession No. PTA-6079.
  • Examples of PPDCs derived from umbilicus tissue were deposited with the American Type Culture Collection on Jun. 10, 2004, and assigned ATCC Accession Numbers as follows: (1) strain designation UMB 022803 (P7) was assigned Accession No. PTA-6067; and (2) strain designation UMB 022803 (P17) was assigned Accession No. PTA-6068.
  • the PPDCs possess one or more of the following growth features (1) they require L-valine for growth in culture; (2) they are capable of growth in atmospheres containing oxygen from about 5% to at least about 20% (3) they have the potential for at least about 40 doublings in culture before reaching senescence; and (4) they attach and expand on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin or fibronectin.
  • the PPDCs possess a normal karyotype, which is maintained as the cells are passaged.
  • Karyotyping is particularly useful for identifying and distinguishing neonatal from maternal cells derived from placenta. Methods for karyotyping are available and known to those of skill in the art.
  • the PPDCs may be characterized by production of certain proteins, including (1) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; and (2) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C cell surface markers, as detected by flow cytometry.
  • the PPDCs may be characterized by lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ cell surface markers, as detected by flow cytometry.
  • Particularly preferred are cells that produce at least two of tissue factor, vimentin, and alpha-smooth muscle actin. More preferred are those cells producing all three of the proteins tissue factor, vimentin, and alpha-smooth muscle actin.
  • the PPDCs may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for a gene encoding at least one of interleukin 8; reticulon 1; chemokine (C—X—C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C—X—C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C—X—C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp56
  • the PPDCs may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for a gene encoding at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 ( Drosophila ); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neural
  • the PPDCs may be characterized by secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1.
  • the PPDCs may be characterized by lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
  • the cell comprises two or more of the above-listed growth, protein/surface marker production, gene expression or substance-secretion characteristics. More preferred are those cells comprising, three, four, or five or more of the characteristics. Still more preferred are PPDCs comprising six, seven, or eight or more of the characteristics. Still more preferred presently are those cells comprising all of above characteristics.
  • Certain cells having the potential to differentiate along lines leading to various phenotypes are unstable and thus can spontaneously differentiate.
  • Presently preferred for use with the invention are cells that do not spontaneously differentiate, for example along neural lines.
  • Preferred cells, when grown in Growth Medium, are substantially stable with respect to the cell markers produced on their surface, and with respect to the expression pattern of various genes, for example as determined using an Affymetrix GENECHIP. The cells remain substantially constant, for example in their surface marker characterisitics over passaging, through multiple population doublings.
  • PPDCs may be deliberately induced to differentiate into various lineage phenotypes by subjecting them to differentiation-inducing cell culture conditions.
  • the PPDCs may be induced to differentiate into neural phenotypes using one or more methods known in the art.
  • PPDCs may be plated on flasks coated with laminin in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine and Penicillin/Streptomycin, the combination of which is referred to herein as Neural Progenitor Expansion (NPE) medium.
  • NPE Neural Progenitor Expansion
  • NPE media may be further supplemented with bFGF and/or EGF.
  • PPDCs may be induced to differentiate in vitro by (1) co-culturing the PPDCs with neural progenitor cells, or (2) growing the PPDCs in neural progenitor cell-conditioned medium.
  • PPDCs Differentiation of the PPDCs into neural phenotypes may be demonstrated by a bipolar cell morphology with extended processes.
  • the induced cell populations may stain positive for the presence of nestin.
  • Differentiated PPDCs may be assessed by detection of nestin, TuJ1 (BIII tubulin), GFAP, tyrosine hydroxylase, GABA, 04 and/or MBP.
  • PPDCs have exhibited the ability to form three-dimensional bodies characteristic of neuronal stem cell formation of neurospheres.
  • the populations comprise the PPDCs described above, and these cell populations are described in the section below.
  • the cell population is heterogeneous.
  • a heterogeneous cell population of the invention may comprise at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% PPDCs.
  • the heterogeneous cell populations of the invention may further comprise stem cells or other progenitor cells, such as epithelial or neural progenitor cells, or it may further comprise fully differentiated cells.
  • the population is substantially homogeneous, i.e., comprises substantially only PPDCs (preferably at least about 96%, 97%, 98%, 99% or more PPDCs).
  • the homogeneous cell population of the invention may comprise umbilicus- or placenta-derived cells. Homogeneous populations of umbilicus-derived cells are preferably free of cells of maternal lineage. Homogeneous populations of placenta-derived cells may be of neonatal or maternal lineage. Homogeneity of a cell population may be achieved by any method known in the art, for example, by cell sorting (e.g., flow cytometry) or by clonal expansion in accordance with known methods. Thus, preferred homogeneous PPDC populations may comprise a clonal cell line of postpartum-derived cells. Such populations are particularly useful when a cell clone with highly desirable functionality has been isolated.
  • populations of cells incubated in the presence of one or more factors, or under conditions, that stimulate stem cell differentiation along a desired pathway e.g., neural, epithelial.
  • a desired pathway e.g., neural, epithelial
  • factors are known in the art and the skilled artisan will appreciate that determination of suitable conditions for differentiation can be accomplished with routine experimentation. Optimization of such conditions can be accomplished by statistical experimental design and analysis, for example response surface methodology allows simultaneous optimization of multiple variables, for example in a biological culture.
  • Presently preferred factors include, but are not limited to factors, such as growth or trophic factors, demethylating agents, co-culture with neural or epithelial lineage cells or culture in neural or epithelial lineage cell-conditioned medium, as well other conditions known in the art to stimulate stem cell differentiation along these pathways (for factors useful in neural differentiation, see, e.g., Lang, K. J. D. et al., 2004, J. Neurosci. Res. 76: 184-192; Johe, K. K. et al., 1996, Genes Devel. 10: 3129-3140; Gottleib, D., 2002, Ann. Rev. Neurosci. 25: 381-407).
  • factors such as growth or trophic factors, demethylating agents, co-culture with neural or epithelial lineage cells or culture in neural or epithelial lineage cell-conditioned medium, as well other conditions known in the art to stimulate stem cell differentiation along these pathways (for factors useful in neural differentiation, see, e.g., Lang, K. J
  • Postpartum cells may also be genetically modified to produce therapeutically useful gene products, or to produce antineoplastic agents for treatment of tumors, for example. Genetic modification may be accomplished using any of a variety of vectors including, but not limited to, integrating viral vectors, e.g., retrovirus vector or adeno-associated viral vectors; non-integrating replicating vectors, e.g., papilloma virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors. Other methods of introducing DNA into cells include the use of liposomes, electroporation, a particle gun, or by direct DNA injection.
  • integrating viral vectors e.g., retrovirus vector or adeno-associated viral vectors
  • non-integrating replicating vectors e.g., papilloma virus vectors, SV40 vectors, adenoviral vectors
  • replication-defective viral vectors e.g., papilloma virus vectors,
  • Hosts cells are preferably transformed or transfected with DNA controlled by or in operative association with, one or more appropriate expression control elements such as promoter or enhancer sequences, transcription terminators, polyadenylation sites, among others, and a selectable marker.
  • Any promoter may be used to drive the expression of the inserted gene.
  • viral promoters include, but are not limited to, the CMV promoter/enhancer, SV 40, papillomavirus, Epstein-Barr virus or elastin gene promoter.
  • the control elements used to control expression of the gene of interest can allow for the regulated expression of the gene so that the product is synthesized only when needed in vivo.
  • constitutive promoters are preferably used in a non-integrating and/or replication-defective vector.
  • inducible promoters could be used to drive the expression of the inserted gene when necessary.
  • Inducible promoters include, but are not limited to, those associated with metallothionein and heat shock proteins.
  • engineered cells may be allowed to grow in enriched media and then switched to selective media.
  • the selectable marker in the foreign DNA confers resistance to the selection and allows cells to stably integrate the foreign DNA as, for example, on a plasmid, into their chromosomes and grow to form foci which, in turn, can be cloned and expanded into cell lines. This method can be advantageously used to engineer cell lines that express the gene product.
  • Cells may be genetically engineered to “knock out” or “knock down” expression of factors that promote inflammation or rejection at the implant site. Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are discussed below. “Negative modulation,” as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level and/or activity of the target gene product in the absence of the modulatory treatment.
  • the expression of a gene native to a neuron or glial cell can be reduced or knocked out using a number of techniques including, for example, inhibition of expression by inactivating the gene using the homologous recombination technique.
  • an exon encoding an important region of the protein is interrupted by a positive selectable marker, e.g., neo, preventing the production of normal mRNA from the target gene and resulting in inactivation of the gene.
  • a gene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the sequences intervening the two regions can be deleted (Mombaerts et al., 1991, Proc. Nat. Acad. Sci. U.S.A. 88:3084).
  • Antisense, DNAzymes, ribozymes, small interfering RNA (siRNA) and other such molecules that inhibit expression of the target gene can also be used to reduce the level of target gene activity.
  • antisense RNA molecules that inhibit the expression of major histocompatibility gene complexes (HLA) have been shown to be most versatile with respect to immune responses.
  • triple helix molecules can be utilized in reducing the level of target gene activity.
  • the invention provides cell lysates and cell soluble fractions prepared from postpartum stem cells, preferably PPDCs, or heterogeneous or homogeneous cell populations comprising PPDCs, as well as PPDCs or populations thereof that have been genetically modified or that have been stimulated to differentiate along a neurogenic pathway.
  • PPDCs postpartum stem cells
  • PPDCs preferably PPDCs
  • PPDCs or populations thereof that have been genetically modified or that have been stimulated to differentiate along a neurogenic pathway.
  • Such lysates and fractions thereof have many utilities.
  • Use of the cell lysate soluble fraction i.e., substantially free of membranes
  • Methods of lysing cells are well-known in the art and include various means of mechanical disruption, enzymatic disruption, or chemical disruption, or combinations thereof.
  • Such cell lysates may be prepared from cells directly in their Growth Medium and thus containing secreted growth factors and the like, or may be prepared from cells washed free of medium in, for example, PBS or other solution. Washed cells may be resuspended at concentrations greater than the original population density if preferred.
  • whole cell lysates are prepared, e.g., by disrupting cells without subsequent separation of cell fractions.
  • a cell membrane fraction is separated from a soluble fraction of the cells by routine methods known in the art, e.g., centrifugation, filtration, or similar methods.
  • Cell lysates or cell soluble fractions prepared from populations of postpartum-derived cells may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, partially purified, combined with pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions.
  • Cell lysates or fractions thereof may be used in vitro or in vivo, alone or for example, with autologous or syngeneic live cells.
  • the lysates, if introduced in vivo may be introduced locally at a site of treatment, or remotely to provide, for example needed cellular growth factors to a patient.
  • postpartum cells can be cultured in vitro to produce biological products in high yield.
  • such cells which either naturally produce a particular biological product of interest (e.g., a trophic factor), or have been genetically engineered to produce a biological product, can be clonally expanded using the culture techniques described herein.
  • cells may be expanded in a medium that induces differentiation to a desired lineage.
  • biological products produced by the cell and secreted into the medium can be readily isolated from the conditioned medium using standard separation techniques, e.g., such as differential protein precipitation, ion-exchange chromatography, gel filtration chromatography, electrophoresis, and HPLC, to name a few.
  • a “bioreactor” may be used to take advantage of the flow method for feeding, for example, a three-dimensional culture in vitro. Essentially, as fresh media is passed through the three-dimensional culture, the biological product is washed out of the culture and may then be isolated from the outflow, as above.
  • a biological product of interest may remain within the cell and, thus, its collection may require that the cells be lysed, as described above.
  • the biological product may then be purified using any one or more of the above-listed techniques.
  • the invention provides conditioned medium from cultured postpartum cells, preferably PPDCs, for use in vitro and in vivo as described below.
  • conditioned medium allows the beneficial trophic factors secreted by the postpartum cells to be used allogeneically in a patient without introducing intact cells that could trigger rejection, or other adverse immunological responses.
  • Conditioned medium is prepared by culturing cells in a culture medium, then removing the cells from the medium.
  • Conditioned medium prepared from populations of postpartum-derived cells may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, partially purified, combined with pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions.
  • Conditioned medium may be used in vitro or in vivo, alone or for example, with autologous or syngeneic live cells.
  • the conditioned medium, if introduced in vivo may be introduced locally at a site of treatment, or remotely to provide, for example needed cellular growth or trophic factors to a patient.
  • an extracellular matrix (ECM) produced by culturing postpartum cells (preferably PPDCs) on liquid, solid or semi-solid substrates is prepared, collected and utilized as an alternative to implanting live cells into a subject in need of tissue repair or replacement.
  • the cells are cultured in vitro, on a three dimensional framework as described elsewhere herein, under conditions such that a desired amount of ECM is secreted onto the framework.
  • the comprising the new tissue are removed, and the ECM processed for further use, for example, as an injectable preparation.
  • cells on the framework are killed and any cellular debris removed from the framework.
  • This process may be carried out in a number of different ways.
  • the living tissue can be flash-frozen in liquid nitrogen without a cryopreservative, or the tissue can be immersed in sterile distilled water so that the cells burst in response to osmotic pressure.
  • the cellular membranes may be disrupted and cellular debris removed by treatment with a mild detergent rinse, such as EDTA, CHAPS or a zwitterionic detergent.
  • a mild detergent rinse such as EDTA, CHAPS or a zwitterionic detergent.
  • the tissue can be enzymatically digested and/or extracted with reagents that break down cellular membranes and allow removal of cell contents.
  • enzymes include, but are not limited to, hyaluronidase, dispase, proteases, and nucleases.
  • detergents include non-ionic detergents such as, for example, alkylaryl polyether alcohol (TRITON X-100), octylphenoxy polyethoxy-ethanol (Rohm and Haas Philadelphia, Pa.), BRIJ-35, a polyethoxyethanol lauryl ether (Atlas Chemical Co., San Diego, Calif.), polysorbate 20 (TWEEN 20), a polyethoxyethanol sorbitan monolaureate (Rohm and Haas), polyethylene lauryl ether (Rohm and Haas); and ionic detergents such as, for example, sodium dodecyl sulphate, sulfated higher aliphatic alcohols, sulfonated alkanes and sulfonated alkylarenes containing 7 to 22 carbon atoms in a branched or unbranched chain.
  • non-ionic detergents such as, for example, alkylaryl polyether alcohol (TRITON X-100), oc
  • the collection of the ECM can be accomplished in a variety of ways, depending, for example, on whether the new tissue has been formed on a three-dimensional framework that is biodegradable or non-biodegradable.
  • the framework is non-biodegradable
  • the ECM can be removed by subjecting the framework to sonication, high pressure water jets, mechanical scraping, or mild treatment with detergents or enzymes, or any combination of the above.
  • the ECM can be collected, for example, by allowing the framework to degrade or dissolve in solution.
  • the biodegradable framework is composed of a material that can itself be injected along with the ECM, the framework and the ECM can be processed in toto for subsequent injection.
  • the ECM can be removed from the biodegradable framework by any of the methods described above for collection of ECM from a non-biodegradable framework. All collection processes are preferably designed so as not to denature the ECM.
  • the ECM may be processed further.
  • the ECM can be homogenized to fine particles using techniques well known in the art such as by sonication, so that it can pass through a surgical needle.
  • the components of the ECM can be crosslinked, if desired, by gamma irradiation.
  • the ECM can be irradiated between 0.25 to 2 mega rads to sterilize and crosslink the ECM.
  • the amounts and/or ratios of proteins may be adjusted by mixing the ECM produced by the cells of the invention with ECM of one or more other cell types.
  • biologically active substances such as proteins, growth factors and/or drugs, can be incorporated into the ECM.
  • tissue growth factors such as TGF-beta, and the like, which promote healing and tissue repair at the site of the injection.
  • additional agents may be utilized in any of the embodiments described herein above, e.g., with whole cell lysates, soluble cell fractions, or further purified components and products produced by the cells.
  • the invention provides pharmaceutical compositions that utilize postpartum cells, preferably PPDCs, cell populations and cell components and products in various methods for treatment of ocular degenerative conditions.
  • Certain embodiments encompass pharmaceutical compositions comprising live cells (e.g., PPDCs alone or admixed with other cell types).
  • Other embodiments encompass pharmaceutical compositions comprising PPDC cellular components (e.g., cell lysates, soluble cell fractions, conditioned medium, ECM, or components of any of the foregoing) or products (e.g., trophic and other biological factors produced naturally by PPDCs or through genetic modification, conditioned medium from PPDC culture).
  • the pharmaceutical composition may further comprise other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative, neuroprotective or ophthalmic drugs as known in the art.
  • PPDC pharmaceutical compositions examples include, but are not limited to: (1) other neuroprotective or neurobeneficial drugs; (2) selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs (alternatively, PPDCs may be genetically engineered to express and produce growth factors); (3) anti-apoptotic agents (e.g., erythropoietin (EPO), EPO mimetibody, thrombopoietin, insulin-like growth factor (IGF)-I, IGF-II, hepatocyte growth factor, caspase inhibitors); (4) anti-inflammatory compounds (e.g., p38 MAP kinase inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and non-steroidal anti-inflammatory drugs (NSAIDS) (such as TEPOX), TEPOX,
  • compositions of the invention comprise postpartum cells (preferably PPDCs), or components or products thereof, formulated with a pharmaceutically acceptable carrier or medium.
  • suitable pharmaceutically acceptable carriers include water, salt solution (such as Ringer's solution), alcohols, oils, gelatins, and carbohydrates, such as lactose, amylose, or starch, fatty acid esters, hydroxymethylcellulose, and polyvinyl pyrolidine.
  • Such preparations can be sterilized, and if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, and coloring.
  • compositions comprising cellular components or products, but not live cells are formulated as liquids.
  • Pharmaceutical compositions comprising PPDC live cells are typically formulated as liquids, semisolids (e.g., gels) or solids (e.g., matrices, scaffolds and the like, as appropriate for ophthalmic tissue engineering).
  • compositions may comprise auxiliary components as would be familiar to medicinal chemists or biologists.
  • they may contain antioxidants in ranges that vary depending on the kind of antioxidant used.
  • Reasonable ranges for commonly used antioxidants are about 0.01% to about 0.15% weight by volume of EDTA, about 0.01% to about 2.0% weight volume of sodium sulfite, and about 0.01% to about 2.0% weight by volume of sodium metabisulfite.
  • One skilled in the art may use a concentration of about 0.1% weight by volume for each of the above.
  • Other representative compounds include mercaptopropionyl glycine, N-acetyl cysteine, beta-mercaptoethylamine, glutathione and similar species, although other anti-oxidant agents suitable for ocular administration, e.g. ascorbic acid and its salts or sulfite or sodium metabisulfite may also be employed.
  • a buffering agent may be used to maintain the pH of eye drop formulations in the range of about 4.0 to about 8.0; so as to minimize irritation of the eye.
  • formulations should be at pH 7.2 to 7.5, preferably at pH 7.3-7.4.
  • the ophthalmologic compositions may also include tonicity agents suitable for administration to the eye. Among those suitable is sodium chloride to make formulations approximately isotonic with 0.9% saline solution.
  • compositions are formulated with viscosity enhancing agents.
  • agents are hydroxyethylcellulose, hydroxypropylcellulose, methylcellulose, and polyvinylpyrrolidone.
  • the pharmaceutical compositions may have cosolvents added if needed. Suitable cosolvents may include glycerin, polyethylene glycol (PEG), polysorbate, propylene glycol, and polyvinyl alcohol. Preservatives may also be included, e.g., benzalkonium chloride, benzethonium chloride, chlorobutanol, phenylmercuric acetate or nitrate, thimerosal, or methyl or propylparabens.
  • Formulations for injection are preferably designed for single-use administration and do not contain preservatives.
  • Injectable solutions should have isotonicity equivalent to 0.9% sodium chloride solution (osmolality of 290-300 milliosmoles). This may be attained by addition of sodium chloride or other co-solvents as listed above, or excipients such as buffering agents and antioxidants, as listed above.
  • Suitable reducing agents include N-acetylcysteine, ascorbic acid or a salt form, and sodium sulfite or metabisulfite, with ascorbic acid and/or N-acetylcysteine or glutathione being particularly suitable for injectable solutions.
  • compositions comprising cells, cell components or cell products may be delivered to the eye of a patient in one or more of several delivery modes known in the art.
  • the compositions are topically delivered to the eye in eye drops or washes.
  • the compositions may be delivered to various locations within the eye via periodic intraocular injection or by infusion in an irrigating solution such as BSS or BSS PLUS (Alcon USA, Fort Worth, Tex.).
  • the compositions may be applied in other ophthalmologic dosage forms known to those skilled in the art, such as pre-formed or in situ-formed gels or liposomes, for example as disclosed in U.S. Pat. No. 5,718,922 to Herrero-Vanrell.
  • the composition may be delivered to or through the lens of an eye in need of treatment via a contact lens (e.g. Lidofilcon B, Bausch & Lomb CW79 or DELTACON (Deltafilcon A) or other object temporarily resident upon the surface of the eye.
  • a contact lens e.g. Lidofilcon B, Bausch & Lomb CW79 or DELTACON (Deltafilcon A) or other object temporarily resident upon the surface of the eye.
  • supports such as a collagen corneal shield (e.g. BIO-COR dissolvable corneal shields, Summit Technology, Watertown, Mass.) can be employed.
  • compositions can also be administered by infusion into the eyeball, either through a cannula from an osmotic pump (ALZET, Alza Corp., Palo Alto, Calif.) or by implantation of timed-release capsules (OCCUSENT) or biodegradable disks (OCULEX, OCUSERT).
  • AZAT osmotic pump
  • OCUSENT timed-release capsules
  • OCULEX biodegradable disks
  • compositions comprising live cells in a semi-solid or solid carrier are typically formulated for surgical implantation at the site of ocular damage or distress. It will be appreciated that liquid compositions also may be administered by surgical procedures.
  • semi-solid or solid pharmaceutical compositions may comprise semi-permeable gels, lattices, cellular scaffolds and the like, which may be non-biodegradable or biodegradable. For example, in certain embodiments, it may be desirable or appropriate to sequester the exogenous cells from their surroundings, yet enable the cells to secrete and deliver biological molecules to surrounding cells.
  • cells may be formulated as autonomous implants comprising living PPDCs or cell population comprising PPDCs surrounded by a non-degradable, selectively permeable barrier that physically separates the transplanted cells from host tissue.
  • Such implants are sometimes referred to as “immunoprotective,” as they have the capacity to prevent immune cells and macromolecules from killing the transplanted cells in the absence of pharmacologically induced immunosuppression (for a review of such devices and methods, see, e.g., P. A. Tresco et al., 2000, Adv. Drug Delivery Rev. 42: 3-27).
  • degradable materials particularly suitable for sustained release formulations include biocompatible polymers, such as poly(lactic acid), poly (lactic-co-glycolic acid), methylcellulose, hyaluronic acid, collagen, and the like.
  • biocompatible polymers such as poly(lactic acid), poly (lactic-co-glycolic acid), methylcellulose, hyaluronic acid, collagen, and the like.
  • the structure, selection and use of degradable polymers in drug delivery vehicles have been reviewed in several publications, including, A. Domb et al., 1992, Polymers for Advanced Technologies 3:279.
  • U.S. Pat. No. 5,869,079 to Wong et al. discloses combinations of hydrophilic and hydrophobic entities in a biodegradable sustained release ocular implant.
  • a biodegradable, preferably bioresorbable or bioabsorbable, scaffold or matrix typically three-dimensional biomaterials contain the living cells attached to the scaffold, dispersed within the scaffold, or incorporated in an extracellular matrix entrapped in the scaffold. Once implanted into the target region of the body, these implants become integrated with the host tissue, wherein the transplanted cells gradually become established (see, e.g., P. A. Tresco et al., 2000, supra; see also D. W. Hutraum, 2001, J. Biomater. Sci. Polymer Edn. 12: 107-174).
  • scaffold or matrix (sometimes referred to collectively as “framework”) material examples include nonwoven mats, porous foams, or self assembling peptides.
  • Nonwoven mats may, for example, be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the tradename VICRYL (Ethicon, Inc., Somerville, NJ), Foams, composed of, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, formed by processes such as freeze-drying, or lyophilized, as discussed in U.S. Pat. No. 6,355,699, also may be utilized.
  • PGA/PLA synthetic absorbable copolymer of glycolic and lactic acids
  • VICRYL Ethicon, Inc., Somerville, NJ
  • Foams composed of, for example, poly(epsilon-caprolactone)/poly(glycolic
  • Hydrogels such as self-assembling peptides (e.g., RAD16) may also be used.
  • In situ-forming degradable networks are also suitable for use in the invention (see, e.g., Anseth, K. S. et al., 2002, J. Controlled Release 78: 199-209; Wang, D. et al., 2003, Biomaterials 24: 3969-3980; U.S. Patent Publication 2002/0022676 to He et al.). These materials are formulated as fluids suitable for injection, then may be induced by a variety of means (e.g., change in temperature, pH, exposure to light) to form degradable hydrogel networks in situ or in vivo.
  • means e.g., change in temperature, pH, exposure to light
  • the framework is a felt, which can be composed of a multifilament yarn made from a bioabsorbable material, e.g., PGA, PLA, PCL copolymers or blends, or hyaluronic acid.
  • the yarn is made into a felt using standard textile processing techniques consisting of crimping, cutting, carding and needling.
  • cells are seeded onto foam scaffolds that may be composite structures.
  • the framework may be molded into a useful shape.
  • PPDCs may be cultured on pre-formed, non-degradable surgical or implantable devices, e.g., in a manner corresponding to that used for preparing fibroblast-containing GDC endovascular coils, for instance (Marx, W. F. et al., 2001, Am. J. Neuroradiol. 22: 323-333).
  • the matrix, scaffold or device may be treated prior to inoculation of cells in order to enhance cell attachment.
  • nylon matrices can be treated with 0.1 molar acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon.
  • Polystyrene can be similarly treated using sulfuric acid.
  • the external surfaces of a framework may also be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma coating the framework or addition of one or more proteins (e.g., collagens, elastic fibers, reticular fibers), glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, among others.
  • proteins e.g., collagens, elastic fibers, reticular fibers
  • glycoproteins e.g., glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermat
  • Frameworks containing living cells are prepared according to methods known in the art. For example, cells can be grown freely in a culture vessel to sub-confluency or confluency, lifted from the culture and inoculated onto the framework. Growth factors may be added to the culture medium prior to, during, or subsequent to inoculation of the cells to trigger differentiation and tissue formation, if desired. Alternatively, the frameworks themselves may be modified so that the growth of cells thereon is enhanced, or so that the risk of rejection of the implant is reduced. Thus, one or more biologically active compounds, including, but not limited to, anti-inflammatories, immunosuppressants or growth factors, may be added to the framework for local release.
  • biologically active compounds including, but not limited to, anti-inflammatories, immunosuppressants or growth factors
  • Postpartum cells preferably PPDCs, or cell populations, components of or products produced by such cells, may be used in a variety of ways to support and facilitate repair and regeneration of ocular cells and tissues. Such utilities encompass in vitro, ex vivo and in vivo methods. The methods set forth below are directed to PPDCs, but other postpartum cells may also be suitable for use in those methods.
  • PPDCs may be used in vitro to screen a wide variety of compounds for effectiveness and cytotoxicity of pharmaceutical agents, growth factors, regulatory factors, and the like. For example, such screening may be performed on substantially homogeneous populations of PPDCs to assess the efficacy or toxicity of candidate compounds to be formulated with, or co-administered with, the PPDCs, for treatment of a an ocular condition. Alternatively, such screening may be performed on PPDCs that have been stimulated to differentiate into a cell type found in the eye, or progenitor thereof, for the purpose of evaluating the efficacy of new pharmaceutical drug candidates. In this embodiment, the PPDCs are maintained in vitro and exposed to the compound to be tested.
  • the activity of a potentially cytotoxic compound can be measured by its ability to damage or kill cells in culture. This may readily be assessed by vital staining techniques.
  • the effect of growth or regulatory factors may be assessed by analyzing the number or robustness of the cultured cells, as compared with cells not exposed to the factors. This may be accomplished using standard cytological and/or histological techniques, including the use of immunocytochemical techniques employing antibodies that define type-specific cellular antigens.
  • PPDCs can be cultured in vitro to produce biological products that are either naturally produced by the cells, or produced by the cells when induced to differentiate into other lineages, or produced by the cells via genetic modification.
  • TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1b, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were found to be secreted from umbilicus-derived cells grown in Growth Medium.
  • TIMP1, TPO, KGF, HGF, HBEGF, BDNF, MIP1a, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were found to be secreted from placenta-derived PPDCs cultured in Growth Medium (see Examples). Some of these trophic factors, such as BDNF and IL-6, have important roles in neural regeneration. Other trophic factors, as yet undetected or unexamined, of use in repair and regeneration of ocular tissues, are likely to be produced by PPDCs and possibly secreted into the medium.
  • another embodiment of the invention features use of PPDCs for production of conditioned medium, either from undifferentiated PPDCs or from PPDCs incubated under conditions that stimulate differentiation.
  • conditioned media are contemplated for use in in vitro or ex vivo culture of epithelial or neural precursor cells, for example, or in vivo to support transplanted cells comprising homogeneous populations of PPDCs or heterogeneous populations comprising PPDCs and other progenitors, for example.
  • Yet another embodiment comprises the use of PPCD cell lysates, soluble cell fractions or components thereof, or ECM or components thereof, for a variety of purposes. As mentioned above, some of these components may be used in pharmaceutical compositions. In other embodiments, a cell lysate or ECM is used to coat or otherwise treat substances or devices to be used surgically, or for implantation, or for ex vivo purposes, to promote healing or survival of cells or tissues contacted in the course of such treatments.
  • PPDCs have demonstrated the ability to support survival, growth and differentiation of adult neural progenitor cells when grown in co-culture with those cells.
  • the experimental results set forth in Example 18 indicates that PPDCs may function to support cells of the retina via trophic mechanisms.
  • PPDCs are used advantageously in co-cultures in vitro to provide trophic support to other cells, in particular neural cells and neural and ocular progenitors (e.g., neural stem cells and retinal or corneal epithelial stem cells).
  • neural cells and ocular progenitors e.g., neural stem cells and retinal or corneal epithelial stem cells.
  • the PPDCs can first be grown to confluence, and then will serve as a substrate for the second desired cell type in culture.
  • the cells may further be physically separated, e.g., by a membrane or similar device, such that the other cell type may be removed and used separately, following the co-culture period.
  • Use of PPDCs in co-culture to promote expansion and differentiation of neural or ocular cell types may find applicability in research and in clinical/therapeutic areas.
  • PPDC co-culture may be utilized to facilitate growth and differentiation of such cells in culture, for basic research purposes or for use in drug screening assays, for example.
  • PPDC co-culture may also be utilized for ex vivo expansion of neural or ocular progenitors for later administration for therapeutic purposes.
  • neural or ocular progenitor cells may be harvested from an individual, expanded ex vivo in co-culture with PPDCs, then returned to that individual (autologous transfer) or another individual (syngeneic or allogeneic transfer).
  • autologous transfer or another individual (syngeneic or allogeneic transfer).
  • the mixed population of cells comprising the PPDCs and progenitors could be administered to a patient in need of treatment.
  • the co-cultured cell populations may be physically separated in culture, enabling removal of the autologous progenitors for administration to the patient.
  • PPDCs have been shown to be effectively transplanted into the body, and to supply lost neural or retinal function in animal models accepted for their predictability of efficacy in humans. These results support a preferred embodiment of the invention, wherein PPDCs are used in cell therapy for treating an ocular degenerative condition.
  • PPDCs Once transplanted into a target location in the eye, PPDCs may themselves differentiate into one or more phenotypes, or they may provide trophic support for ocular cells in situ, or they may exert a beneficial effect in both of those fashions, among others.
  • PPDCs may be administered alone (e.g., as substantially homogeneous populations) or as admixtures with other cells.
  • PPDCs may be administered as formulated in a pharmaceutical preparation with a matrix or scaffold, or with conventional pharmaceutically acceptable carriers.
  • PPDCs may be administered simultaneously or sequentially with the other cells (either before or after the other cells).
  • Cells that may be administered in conjunction with PPDCs include, but are not limited to, neurons, astrocytes, oligodendrocytes, neural progenitor cells, neural stem cells, ocular progenitor cells, retinal or corneal epithelial stem cells and/or other multipotent or pluripotent stem cells.
  • the cells of different types may be admixed with the PPDCs immediately or shortly prior to administration, or they may be co-cultured together for a period of time prior to administration.
  • the PPDCs may be administered with other beneficial drugs, biological molecules, such as growth factors, trophic factors, conditioned medium (from postpartum cells or from progenitor or differentiated cell cultures), or other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art.
  • beneficial drugs biological molecules, such as growth factors, trophic factors, conditioned medium (from postpartum cells or from progenitor or differentiated cell cultures), or other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art.
  • biological molecules such as growth factors, trophic factors, conditioned medium (from postpartum cells or from progenitor or differentiated cell cultures)
  • active agents such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art.
  • Examples of other components that may be administered with postpartum cells include, but are not limited to: (1) other neuroprotective or neurobeneficial drugs; (2) selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs (alternatively, the cells may be genetically engineered to express and produce growth factors); (3) anti-apoptotic agents (e.g., erythropoietin (EPO), EPO mimetibody, thrombopoietin, insulin-like growth factor (IGF)-I, IGF-II, hepatocyte growth factor, caspase inhibitors); (4) anti-inflammatory compounds (e.g., p38 MAP kinase inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and non-steroidal anti-inflammatory drugs (NSAIDS) (such as TEPOXALIN,
  • PPDCs are administered as undifferentiated cells, i.e., as cultured in Growth Medium.
  • PPDCs may be administered following exposure in culture to conditions that stimulate differentiation toward a desired phenotype.
  • the cells may be surgically implanted, injected or otherwise administered directly or indirectly to the site of ocular damage or distress.
  • surgical implantation into a precise location in the body is typically a suitable means of administration.
  • Liquid or fluid pharmaceutical compositions may be administered to a more general location in the eye (e.g., topically or intra-ocularly).
  • compositions comprising PPDC cellular components (e.g., cell lysates or components thereof) or products (e.g., trophic and other biological factors produced naturally by PPDCs or through genetic modification, conditioned medium from PPDC culture).
  • these methods may further comprise administering other active agents, such as growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art.
  • Dosage forms and regimes for administering PPDCs or any of the other pharmaceutical compositions described herein are developed in accordance with good medical practice, taking into account the condition of the individual patient, e.g., nature and extent of the ocular degenerative condition, age, sex, body weight and general medical condition, and other factors known to medical practitioners.
  • the effective amount of a pharmaceutical composition to be administered to a patient is determined by these considerations as known in the art.
  • PPDCs may be genetically modified to reduce their immunogenicity, as mentioned above.
  • Survival of transplanted cells in a living patient can be determined through the use of a variety of scanning techniques, e.g., computerized axial tomography (CAT or CT) scan, magnetic resonance imaging (MRI) or positron emission tomography (PET) scans. Determination of transplant survival can also be done post mortem by removing the tissue and examining it visually or through a microscope. Alternatively, cells can be treated with stains that are specific for neural or ocular cells or products thereof, e.g., neurotransmitters.
  • CAT or CT computerized axial tomography
  • MRI magnetic resonance imaging
  • PET positron emission tomography
  • Transplanted cells can also be identified by prior incorporation of tracer dyes such as rhodamine- or fluorescein-labeled microspheres, fast blue, ferric microparticles, bisbenzamide or genetically introduced reporter gene products, such as beta-galactosidase or beta-glucuronidase.
  • tracer dyes such as rhodamine- or fluorescein-labeled microspheres, fast blue, ferric microparticles, bisbenzamide or genetically introduced reporter gene products, such as beta-galactosidase or beta-glucuronidase.
  • Functional integration of transplanted cells into ocular tissue of a subject can be assessed by examining restoration of the ocular function that was damaged or diseased.
  • effectiveness in the treatment of macular degeneration or other retinopathies may be determined by improvement of visual acuity and evaluation for abnormalities and grading of stereoscopic color fundus photographs. (Age-Related Eye Disease Study Research Group, NEI, NIH, AREDS Report No. 8, 2001, Arch. Ophthalmol. 119: 1417-1436).
  • kits that utilize postpartum cells, preferably PPDCs, cell populations, components and products thereof in various methods for ocular regeneration and repair as described above.
  • the kits may include one or more cell populations, including at least postpartum cells and a pharmaceutically acceptable carrier (liquid, semi-solid or solid).
  • the kits also optionally may include a means of administering the cells, for example by injection.
  • the kits further may include instructions for use of the cells. Kits prepared for field hospital use, such as for military use may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute injuries.
  • Kits for assays and in vitro methods as described herein may contain, for example, one or more of (1) PPDCs or components or products of PPDCs, (2) reagents for practicing the in vitro method, (3) other cells or cell populations, as appropriate, and (4) instructions for conducting the in vitro method.
  • the invention also provides for banking of tissues, cells, cellular components and cell populations of the invention.
  • the cells are readily cryopreserved.
  • the invention therefore provides methods of cryopreserving the cells in a bank, wherein the cells are stored frozen and associated with a complete characterization of the cells based on immunological, biochemical and genetic properties of the cells.
  • the frozen cells can be thawed and expanded or used directly for autologous, syngeneic, or allogeneic therapy, depending on the requirements of the procedure and the needs of the patient.
  • the information on each cryopreserved sample is stored in a computer, which is searchable based on the requirements of the surgeon, procedure and patient with suitable matches being made based on the characterization of the cells or populations.
  • the cells of the invention are grown and expanded to the desired quantity of cells and therapeutic cell compositions are prepared either separately or as co-cultures, in the presence or absence of a matrix or support. While for some applications it may be preferable to use cells freshly prepared, the remainder can be cryopreserved and banked by freezing the cells and entering the information in the computer to associate the computer entry with the samples. Even where it is not necessary to match a source or donor with a recipient of such cells, for immunological purposes, the bank system makes it easy to match, for example, desirable biochemical or genetic properties of the banked cells to the therapeutic needs. Upon matching of the desired properties with a banked sample, the sample is retrieved and prepared for therapeutic use. Cell lysates, ECM or cellular components prepared as described herein may also be cryopreserved or otherwise preserved (e.g., by lyophilization) and banked in accordance with the present invention.
  • the term Growth Medium generally refers to a medium sufficient for the culturing of PPDCs.
  • one presently preferred medium for the culturing of the cells of the invention in comprises Dulbecco's Modified Essential Media (also abbreviated DMEM herein).
  • DMEM-low glucose also DMEM-LG herein
  • the DMEM-low glucose is preferably supplemented with 15% (v/v) fetal bovine serum (e.g.
  • Growth Medium refers to DMEM-low glucose with 15% fetal bovine serum and antibiotics/antimycotics (when penicillin/streptomycin are included, it is preferably at 50 U/ml and 50 microgram/ml respectively; when penicillin/streptomycin/amphotericin B are use, it is preferably at 100 U/ml, 100 microgram/ml and 0.25 microgram/ml, respectively). In some cases different growth media are used, or different supplementations are provided, and these are normally indicated in the text as supplementations to Growth Medium.
  • ANG2 (or Ang2) for angiopoietin 2; APC for antigen-presenting cells; BDNF for brain-derived neurotrophic factor; bFGF for basic fibroblast growth factor; bid (BID) for “bis in die” (twice per day); CK18 for cytokeratin 18; CNS for central nervous system; CXC ligand 3 for chemokine receptor ligand 3; DMEM for Dulbecco's Minimal Essential Medium; DMEM:1 g (or DMEM:Lg, DMEM:LG) for DMEM with low glucose; EDTA for ethylene diamine tetraacetic acid; EGF (or E) for epidermal growth factor; FACS for fluorescent activated cell sorting; FBS for fetal bovine serum; FGF (or F) for fibroblast growth factor; GCP-2 for granulocyte chemotactic protein-2; GFAP for gli
  • This example describes the preparation of postpartum-derived cells from placental and umbilical cord tissues.
  • Postpartum umbilical cords and placentae were obtained upon birth of either a full term or pre-term pregnancy.
  • Cells were harvested from 5 separate donors of umbilicus and placental tissue.
  • Different methods of cell isolation were tested for their ability to yield cells with: 1) the potential to differentiate into cells with different phenotypes, a characteristic common to stem cells, or 2) the potential to provide trophic factors useful for other cells and tissues.
  • Umbilical cell isolation Umbilical cords were obtained from National Disease Research Interchange (NDR1, Philadelphia, Pa.). The tissues were obtained following normal deliveries. The cell isolation protocol was performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of antimycotic and antibiotic (100 units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B).
  • PBS phosphate buffered saline
  • antibiotic 100 units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B).
  • the tissues were then mechanically dissociated in 150 cm 2 tissue culture plates in the presence of 50 milliliters of medium (DMEM-Low glucose or DMEM-High glucose; Invitrogen), until the tissue was minced into a fine pulp.
  • the chopped tissues were transferred to 50 milliliter conical tubes (approximately 5 grams of tissue per tube).
  • the tissue was then digested in either DMEM-Low glucose medium or DMEM-High glucose medium, each containing antimycotic and antibiotic as described above.
  • C:D collagenase (Sigma, St Louis, Mo.), 500 Units/milliliter; and dispase (Invitrogen), 50 Units/milliliter in DMEM:-Low glucose medium).
  • C:D:H collagenase, 500 Units/milliliter; dispase, 50 Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter, in DMEM:-Low glucose).
  • the conical tubes containing the tissue, medium and digestion enzymes were incubated at 37° C. in an orbital shaker (Environ, Brooklyn, NY) at 225 rpm for 2 hrs.
  • the cells were centrifuged at 150 ⁇ g for 5 minutes, the supernatant was aspirated.
  • the pellet was resuspended in 20 milliliters of Growth Medium (DMEM:Low glucose (Invitrogen), 15 percent (v/v) fetal bovine serum (FBS; defined bovine serum; Lot#AND18475; Hyclone, Logan, Utah), 0.001% (v/v) 2-mercaptoethanol (Sigma), 1 milliliter per 100 milliliters of antibiotic/antimycotic as described above.
  • the cell suspension was filtered through a 70-micrometer nylon cell strainer (BD Biosciences). An additional 5 milliliters rinse comprising Growth Medium was passed through the strainer.
  • the cell suspension was then passed through a 40-micrometer nylon cell strainer (BD Biosciences) and chased with a rinse of an additional 5 milliliters of Growth Medium.
  • the filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 ⁇ g for 5 minutes. The supernatant was aspirated and the cells were resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more.
  • the cells isolated from umbilical cords were seeded at 5,000 cells/cm 2 onto gelatin-coated T-75 cm 2 flasks (Corning Inc., Corning, N.Y.) in Growth Medium with antibiotics/antimycotics as described above. After 2 days (in various experiments, cells were incubated from 2-4 days), spent medium was aspirated from the flasks. Cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with Growth Medium and allowed to grow to confluence (about 10 days from passage 0) to passage 1. On subsequent passages (from passage 1 to 2 and so on), cells reached sub-confluence (75-85 percent confluence) in 4-5 days. For these subsequent passages, cells were seeded at 5000 cells/cm 2 . Cells were grown in a humidified incubator with 5 percent carbon dioxide and atmospheric oxygen, at 37° C.
  • Placental Cell Isolation Placental tissue was obtained from NDR1 (Philadelphia, Pa.). The tissues were from a pregnancy and were obtained at the time of a normal surgical delivery. Placental cells were isolated as described for umbilical cell isolation.
  • the following example applies to the isolation of separate populations of maternal-derived and neonatal-derived cells from placental tissue.
  • the cell isolation protocol was performed aseptically in a laminar flow hood.
  • the placental tissue was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of antimycotic and antibiotic (as described above) to remove blood and debris.
  • PBS phosphate buffered saline
  • the placental tissue was then dissected into three sections: top-line (neonatal side or aspect), mid-line (mixed cell isolation neonatal and maternal) and bottom line (maternal side or aspect).
  • the separated sections were individually washed several times in PBS with antibiotic/antimycotic to further remove blood and debris. Each section was then mechanically dissociated in 150 cm 2 tissue culture plates in the presence of 50 milliliters of DMEM/Low glucose, to a fine pulp. The pulp was transferred to 50 milliliter conical tubes. Each tube contained approximately 5 grams of tissue. The tissue was digested in either DMEM-Low glucose or DMEM-High glucose medium containing antimycotic and antibiotic (100 U/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B) and digestion enzymes.
  • C:D collagenase and dispase
  • collagenase Sigma, St Louis, Mo.
  • dispase Invitrogen
  • C:D:H a mixture of collagenase, dispase and hyaluronidase
  • the conical tubes containing the tissue, medium, and digestion enzymes were incubated for 2 h at 37° C. in an orbital shaker (Environ, Brooklyn, N.Y.) at 225 rpm.
  • the tissues were centrifuged at 150 ⁇ g for 5 minutes, the resultant supernatant was aspirated off.
  • the pellet was resuspended in 20 milliliters of Growth Medium with penicillin/streptomycin/amphotericin B.
  • the cell suspension was filtered through a 70 micometer nylon cell strainer (BD Biosciences), chased by a rinse with an additional 5 milliliters of Growth Medium.
  • the total cell suspension was passed through a 40 micometer nylon cell strainer (BD Biosciences) followed with an additional 5 milliliters of Growth Medium as a rinse.
  • the filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150 ⁇ g for 5 minutes. The supernatant was aspirated and the cell pellet was resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more. After the final centrifugation, supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. A cell count was determined using the Trypan Blue Exclusion test. Cells were then cultured at standard conditions.
  • LIBERASE Cell Isolation Cells were isolated from umbilicus tissues in DMEM-Low glucose medium with LIBERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) (2.5 milligrams per milliliter, Blendzyme 3; Roche Applied Sciences, Indianapolis, Ind.) and hyaluronidase (5 Units/milliliter, Sigma). Digestion of the tissue and isolation of the cells was as described for other protease digestions above, using the LIBERASE/hyaluronidase mixture in place of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE resulted in the isolation of cell populations from postpartum tissues that expanded readily.
  • the resulting cell suspension was vortexed for 5 seconds, and incubated for 2 minutes at ambient temperature.
  • the lysate was centrifuged (10 minutes at 200 ⁇ g).
  • the cell pellet was resuspended in complete minimal essential medium (Gibco, Carlsbad Calif.) containing 10 percent fetal bovine serum (Hyclone, Logan Utah), 4 millimolar glutamine (Mediatech Hemdon, VA), 100 Units penicillin per 100 milliliters and 100 micrograms streptomycin per 100 milliliters (Gibco, Carlsbad, Calif.).
  • the resuspended cells were centrifuged (10 minutes at 200 ⁇ g), the supernatant was aspirated, and the cell pellet was washed in complete medium.
  • T75 flasks Corning, N.Y.
  • T75 laminin-coated flasks T75 laminin-coated flasks
  • T175 fibronectin-coated flasks both Becton Dickinson, Bedford, Mass.
  • the preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.
  • Populations of cells can be derived from umbilical cord and placental tissue efficiently using the enzyme combination collagenase (a matrix metalloprotease), dispase (a neutral protease) and hyaluronidase (a mucolytic enzyme that breaks down hyaluronic acid).
  • LIBERASE which is a Blendzyme, may also be used.
  • Blendzyme 3 which is collagenase (4 Wunsch units/g) and thermolysin (1714 casein Units/g) was also used together with hyaluronidase to isolate cells.
  • Cells were also isolated from residual blood in the cords, but not cord blood. The presence of cells in blood clots washed from the tissue, that adhere and grow under the conditions used, may be due to cells being released during the dissection process.
  • PPDCs postpartum-derived cells
  • the process of cell expansion to senescence is referred to as Hayflick's limit (Hayflick L. 1974a, 1974b).
  • Postpartum-derived cells are highly suited for therapeutic use because they can be readily expanded to sufficient cell numbers.
  • Tissue culture plastic flasks were coated by adding 20 milliliters 2% (w/v) porcine gelatin (Type B: 225 Bloom; Sigma, St Louis, Mo.) to a T75 flask (Corning, Corning, N.Y.) for 20 minutes at room temperature. After removing the gelatin solution, 10 milliliters phosphate-buffered saline (PBS) (Invitrogen, Carlsbad, Calif.) was added and then aspirated.
  • PBS phosphate-buffered saline
  • Cells were initially seeded at 5,000 cells/cm 2 on gelatin-coated T75 flasks in Growth Medium with penicillin/streptomycin/amphotericin B. For subsequent passages, cell cultures were treated as follows. After trypsinization, viable cells were counted after Trypan Blue staining. Cell suspension (50 microliters) was combined with Trypan Blue (50 milliliters, Sigma, St. Louis Mo.). Viable cell numbers were estimated using a hemocytometer.
  • cells were seeded at 5,000 cells/cm 2 onto gelatin-coated T 75 flasks in 25 milliliters of fresh Growth Medium. Cells were grown under standard conditions at 37° C. The Growth Medium was changed twice per week. When cells reached about 85 percent confluence they were passaged; this process was repeated until the cells reached senescence.
  • Expansion potential of cell banks at low density The expansion potential of cells banked at passage 10 was also tested, using a different set of conditions. Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, MD), umbilicus-derived cells, and placenta-derived cells were tested. These cell populations had been banked at passage 10 previously, having been cultured at 5,000 cells/cm 2 and grown to confluence at each passage to that point. The effect of cell density on the cell populations following cell thaw at passage 10 was determined. Cells were thawed under standard conditions and counted using Trypan Blue staining.
  • DMEM:Low glucose Growth Medium with antibiotic/antimycotic as described above.
  • Cloning was used in order to expand a population of neonatal cells from placental tissue. Following isolation of three differential cell populations from the placenta (as described herein), these cell populations were expanded under standard growth conditions and then karyotyped to reveal the identity of the isolated cell populations. Because the cells were isolated from a mother who delivered a boy, it was straightforward to distinguish between the male and female chromosomes by performing metaphase spreads.
  • fetal-aspect cells were karyotype positive for neonatal phenotpye
  • mid-layer cells were karyotype positive for both neonatal and maternal phenotypes
  • maternal-aspect cells were karyotype positive for maternal cells.
  • Umbilicus-derived, placenta-derived and fibroblast cells expanded for greater than 10 passages generating cell yields of >1E11 cells in 60 days (Table 2-2). After 60 days under these conditions the fibroblasts became senescent whereas the umbilicus-derived and placenta-derived cell populations senesced after 80 days, completing >50 and >40 population doublings respectively.
  • PPDCs Expansion of PPDCs at low density from initial cell seeding.
  • PPDCs were expanded at low density (1,000 cells/cm 2 ) on gelatin-coated and uncoated plates or flasks. Growth potential of these cells under these conditions was good. The cells expanded readily in a log phase growth. The rate of cell expansion was similar to that observed when placenta-derived cells were seeded at 5000 cells/cm 2 on gelatin-coated flasks in Growth Medium. No differences were observed in cell expansion potential between culturing on either uncoated flasks or gelatin-coated flasks. However, cells appeared phenotypically much smaller on gelatin-coated flasks and more larger cell phenotypes were observed on uncoated flasks.
  • a clonal neonatal or maternal cell population can be expanded from placenta-derived cells isolated from the neonatal aspect or the maternal aspect, respectively, of the placenta.
  • Cells are serially diluted and then seeded onto gelatin-coated plates in Growth medium for expansion at 1 cell/well in 96-well gelatin coated plates. From this initial cloning, expansive clones are identified, trypsinized, and reseeded in 12-well gelatin-coated plates in Growth medium and then subsequently passaged into T25 gelatin-coated flasks at 5,000 cells/cm 2 in Growth medium.
  • Subcloning is performed to ensure that a clonal population of cells has been identified.
  • cells are trypsinized and reseeded at 0.5 cells/well.
  • the subclones that grow well are expanded in gelatin-coated T25 flasks at 5,000 cells cm 2 /flask. Cells are passaged at 5,000 cells cm 2 /T75 flask.
  • the growth characteristics of a clone may be plotted to demonstrate cell expansion.
  • Karyotyping analysis can confirm that the clone is either neonatal or maternal.
  • Cell expansion conditions comprising growing isolated postpartum-derived cells at densities of about 5000 cells/cm 2 , in Growth Medium on gelatin-coated or uncoated flasks, under standard atmospheric oxygen, are sufficient to generate large numbers of cells at passage 11. Furthermore, the data suggests that the cells can be readily expanded using lower density culture conditions (e.g. 1000 cells/cm 2 ). Postpartum-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for growth. Presently, culturing postpartum-derived cells under standard atmospheric conditions is preferred for generating large pools of cells. However, when the culture conditions are altered, postpartum-derived cell expansion can likewise be altered. This strategy may be used to enhance the proliferative and differentiative capacity of these cell populations.
  • lower density culture conditions e.g. 1000 cells/cm 2
  • Postpartum-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for
  • placenta-derived cells were evaluated for their ability to support the growth of placenta-derived cells.
  • Placenta-derived cells at passage 8 were seeded at 1 ⁇ 10 3 cells/well in 96 well plates in Growth Medium with penicillin/streptomycin. After 8 hours the medium was changed as described below and cells were incubated in normal (atmospheric) or low (5%, v/v) oxygen at 37° C., 5% CO 2 for 48 hours. MTS was added to the culture medium (CELLTITER 96 AQueous One Solution Cell Proliferation Assay, Promega, Madison, Wis.) for 3 hours and the absorbance measured at 490 nanometers (Molecular Devices, Sunnyvale Calif.).
  • Standard curves for the MTS assay established a linear correlation between an increase in absorbance and an increase in cell number.
  • the absorbance values obtained were converted into estimated cell numbers and the change (%) relative to the initial seeding was calculated.
  • Placenta-derived cells may be grown in a variety of culture media in normal or low oxygen. Short term growth of placenta-derived cells was determined in twelve basal media with 0, 2 and 10% (v/v) serum in 5% or atmospheric oxygen. In general, placenta-derived cells did not grow as well in serum-free conditions with the exception of Ham's F10 and CELLGRO-Free, which are also protein-free. Growth in these serum-free media was about 25-33% of the maximal growth observed with media containing 15% serum.
  • Placenta-derived cells (P3), fibroblasts (P9) and umbilical-derived cells (P5) were seeded at 5 ⁇ 10 3 cells/cm 2 in gelatin-coated T75 flasks (Corning, Corning, N.Y.). After 24 hours the medium was removed and the cells were washed with phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) to remove residual medium.
  • PBS phosphate buffered saline
  • the medium was replaced with a Modified Growth Medium (DMEM with D-valine (special order Gibco), 15% (v/v) dialyzed fetal bovine serum (Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma), penicillin/streptomycin (Gibco)).
  • DMEM with D-valine special order Gibco
  • 15% (v/v) dialyzed fetal bovine serum Hyclone, Logan, Utah
  • betamercaptoethanol Sigma
  • penicillin/streptomycin Gabco
  • Placenta-derived, umbilical-derived, and fibroblast cells seeded in the D-valine-containing medium did not proliferate, unlike cells seeded in Growth Medium containing dialyzed serum. Fibroblasts cells changed morphologically, increasing in size and changing shape. All of the cells died and eventually detached from the flask surface after 4 weeks. These results indicate that medium containing D-valine is not suitable for selectively growing postpartum-derived cells.
  • Cryopreservation media for the cryopreservation of placenta-derived cells were evaluated.
  • Placenta-derived cells grown in Growth Medium in a gelatin-coated T75 flask were washed with PBS and trypsinized using 1 milliliter Trypsin/EDTA (Gibco). The trypsinization was stopped by adding 10 milliliters Growth Medium. The cells were centrifuged at 150 ⁇ g, supernatant removed, and the cell pellet was resuspended in 1 milliliter Growth Medium. An aliquot of cell suspension, 60 microliters, was removed and added to 60 microliters trypan blue (Sigma). The viable cell number was estimated using a hemocytometer. The cell suspension was divided into four equal aliquots each containing 88 ⁇ 10 4 cells each. The cell suspension was centrifuged and resuspended in 1 milliliter of each media below and transferred into Cryovials (Nalgene).
  • the cells were cooled at approximately ⁇ 1° C./min overnight in a ⁇ 80° C. freezer using a “Mr Frosty” freezing container according to the manufacturer's instructions (Nalgene, Rochester, N.Y.). Vials of cells were transferred into liquid nitrogen for 2 days before thawing rapidly in a 37° C. water bath. The cells were added to 10 milliliters Growth Medium and centrifuged before the cell number and viability was estimated. Cells were seeded onto gelatin-coated flasks at 5,000 cells/cm 2 to determine whether the cells would attach and proliferate.
  • the initial viability of the cells to be cryopreserved was assessed by trypan blue staining to be 100%.
  • the initial viability of the cells to be cryopreserved was assessed by trypan blue staining to be 100%.
  • cryopreservation of cells is one procedure available for preparation of a cell bank or a cell product.
  • Four cryopreservation mixtures were compared for their ability to protect human placenta-derived cells from freezing damage.
  • Dulbecco's modified Eagle's medium (DMEM) and 10% (v/v) dimethylsulfoxide (DMSO) is the preferred medium of those compared for cryopreservation of placenta-derived cells.
  • Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type.
  • Cells used in cell therapy should have a normal chromosome number (46) and structure.
  • placenta- and umbilicus-derived cell lines that are homogeneous and free from cells of non-postpartum tissue origin karyotypes of cell samples were analyzed.
  • PPDCs from postpartum tissue of a male neonate were cultured in Growth Medium containing penicillin/streptomycin.
  • Postpartum tissue from a male neonate (X,Y) was selected to allow distinction between neonatal-derived cells and maternal derived cells (X,X).
  • Cells were seeded at 5,000 cells per square centimeter in Growth Medium in a T25 flask (Corning, Corning, N.Y.) and expanded to 80% confluence. A T25 flask containing cells was filled to the neck with Growth Medium. Samples were delivered to a clinical cytogenetics laboratory by courier (estimated lab to lab transport time is one hour). Cells were analyzed during metaphase when the chromosomes are best visualized.
  • Chromosome analysis identified placenta- and umbilicus-derived cells whose karyotypes appeared normal as interpreted by a clinical cytogenetic laboratory.
  • Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.
  • Characterization of cell surface proteins or “markers” by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions. Postpartum-derived cell (PPDC) lines isolated from the placenta and umbilicus were characterized (by flow cytometry), providing a profile for the identification of these cell lines.
  • PPDC Postpartum-derived cell
  • Placenta and umbilicus comparison were compared to umbilicus-derive cells at passage 8.
  • Placenta- and umbilicus-derived cells were analyzed at passages 8, 15, and 20.
  • Donor to donor comparison To compare differences among donors, placenta-derived cells from different donors were compared to each other, and umbilicus-derived cells from different donors were compared to each other.
  • Placenta-derived cells cultured on gelatin-coated flasks was compared to placenta-derived cells cultured on uncoated flasks.
  • Umbilicus-derived cells cultured on gelatin-coated flasks was compared to umbilicus-derived cells cultured on uncoated flasks.
  • Digestion enzyme comparison Four treatments used for isolation and preparation of cells were compared. Cells isolated from placenta by treatment with 1) collagenase; 2) collagenase/dispase; 3) collagenase/hyaluronidase; and 4) collagenase/hyaluronidase/dispase were compared.
  • Placental layer comparison Cells derived from the maternal aspect of placental tissue were compared to cells derived from the villous region of placental tissue and cells derived from the neonatal fetal aspect of placenta.
  • Placenta vs. umbilicus comparison Placenta- and umbilicus-derived cells analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for detectable expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values comparable to the IgG control. Variations in fluorescence values of positive curves were accounted for. The mean (i.e. CD 13) and range (i.e. CD90) of the positive curves showed some variation, but the curves appeared normal, confirming a homogenous population. Both curves individually exhibited values greater than the IgG control.
  • Placenta-derived cells Passage to passage comparison—placenta-derived cells. Placenta-derived cells at passages 8, 15, and 20 analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, as reflected in the increased value of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ having fluorescence values consistent with the IgG control.
  • Umbilicus-derived cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control.
  • Placenta-derived cells Placenta-derived cells isolated from separate donors analyzed by flow cytometry each expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence value consistent with the IgG control.
  • Donor to donor comparison umbilicus derived cells.
  • Umbilicus-derived cells isolated from separate donors analyzed by flow cytometry each showed positive expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control.
  • Placenta-derived cells expanded on either gelatin-coated or uncoated flasks analyzed by flow cytometry all expressed of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ indicated by fluorescence values consistent with the IgG control.
  • Umbilicus-derived cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
  • Placenta-derived cells isolated using various digestion enzymes analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased values of fluorescence relative to the IgG control.
  • These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control
  • Placental layer comparison Cells isolated from the maternal, villous, and neonatal layers of the placenta, respectively, analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased value of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control.
  • Placenta- and umbilicus-derived cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A,B,C and negative for CD31, CD34, CD45, CD117, CD141 and HLA-DR, DP, DQ. This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, digestion enzymes, and placental layer.
  • anti-human GROalpha—PE (1:100; Becton Dickinson, Franklin Lakes, N.J.
  • anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA)
  • anti-human oxidized LDL receptor 1 ox-LDL R1; 1:100; Santa Cruz Biotech
  • anti-human NOGO-A (1:100; Santa Cruz Biotech).
  • Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, Calif.) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 ⁇ m thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
  • Immunohistochemistry was performed similar to previous studies (e.g., Messina, et al., 2003, Exper. Neurol. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 1 hour to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (CD34, ox-LDL R1), Triton was omitted in all steps of the procedure in order to prevent epitope loss.
  • PBS phosphate-buffered saline
  • Triton Triton X-100
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). Positive staining was represented by fluorescence signal above control staining. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord. In particular, vWF and CD34 expression were restricted to blood vessels contained within the cord. CD34+cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery & vein, but not contained with the vessels themselves. CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.
  • Placenta characterization Vimentin, desmin, SMA, CK18, vWF, and CD34 were all observed within the placenta and regionally specific.
  • GROalpha, GCP-2, ox-LDL R1, and NOGO-A Tissue Expression None of these markers were observed within umbilical cord or placental tissue.
  • Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD 34 are expressed in cells within human umbilical cord and placenta.
  • Affymetrix GENECHIP arrays were used to compare gene expression profiles of umbilicus- and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.
  • Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, MD; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines were cultured in DMEM/F 12 medium (Invitrogen, Carlsbad, Calif.) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen). The cells were grown on standard tissue-treated plastic.
  • hMSC Human mesenchymal stem cells
  • Human ileac crest bone marrow was received from NDR1 with patient consent.
  • the marrow was processed according to the method outlined by Ho, et al. (WO03/025149).
  • the marrow was mixed with lysis buffer (155 mM NH 4 Cl, 10 mM KHCO 3 , and 0.1 mM EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer.
  • the cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500 ⁇ g.
  • the supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 mM glutamine.
  • the cells were centrifuged again and the cell pellet was resuspended in fresh medium.
  • the viable mononuclear cells were counted using trypan-blue exclusion (Sigma, St. Louis, Mo.).
  • the mononuclear cells were seeded in tissue-cultured plastic flasks at 5 ⁇ 104 cells/cm 2 .
  • the cells were incubated at 37° C. with 5% CO 2 at either standard atmospheric O 2 or at 5% O 2 .
  • Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture. The adherent cells were maintained in culture.
  • ICBM (062703) 5 Plastic MEM, 10% FBS (5% 02) hMSC (Lot 2F1655) 3 Plastic MSCGM hMSC (Lot 2F1656) 3 Plastic MSCGM hMSC (Lot 2F 1657) 3 Plastic MSCGM hFibroblast (9F0844) 9 Plastic DMEM-F12, 10% FBS hFibroblast 4 Plastic DMEM-F12, 10% FBS (CCD39SK)
  • Table 9-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs.
  • the Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types.
  • the Euclidean distance is inversely proportional to similarity between the expression of the 290 genes (i.e., the greater the distance, the less similarity exists).
  • TABLE 9-2 The Euclidean Distances for the Cell Pairs.
  • Tables 9-3, 9-4, and 9-5 show the expression of genes increased in placenta-derived cells (Table 9-3), increased in umbilicus-derived cells (Table 9-4), and reduced in umbilicus- and placenta-derived cells (Table 9-5).
  • the column entitled “Probe Set ID” refers to the manufacturer's identification code for the sets of several oligonucleotide probes located on a particular site on the chip, which hybridize to the named gene (column “Gene Name”), comprising a sequence that can be found within the NCBI (GenBank) database at the specified accession number (column “NCBI Accession Number”).
  • Tables 9-6, 9-7, and 9-8 show the expression of genes increased in human fibroblasts (Table 9-6), ICBM cells (Table 9-7), and MSCs (Table 9-8). TABLE 9-6 Genes that were shown to have increased expression in fibroblasts as compared to the other cell lines assayed.
  • the present examination was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas. The examination also included two different lines of dermal fibroblasts, three lines of mesenchymal stem cells, and three lines of ileac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed using an oligonucleotide array that contained probes for 22,000 genes. Results showed that 290 genes are differentially expressed in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells.
  • Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord, as compared with the other cell types. The expression of selected genes has been confirmed by PCR (see the example that follows). These results demonstrate that the postpartum-derived cells have a distinct gene expression profile, for example, as compared to bone marrow-derived cells and fibroblasts.
  • similarities and differences in cells derived from the human placenta and the human umbilical cord were assessed by comparing their gene expression profiles with those of cells derived from other sources (using an oligonucleotide array).
  • Six “signature” genes were identified: oxidized LDL receptor 1, interleukin-8, rennin, reticulon, chemokine receptor ligand 3 (CXC ligand 3), and granulocyte chemotactic protein 2 (GCP-2). These “signature” genes were expressed at relatively high levels in postpartum-derived cells.
  • Placenta-derived cells three isolates, including one isolate predominately neonatal as identified by karyotyping analysis), umbilicus-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) grown in Growth Medium with penicillin/streptomycin in a gelatin-coated T75 flask.
  • Mesechymal Stem Cells were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, MD).
  • IL-8 protocol cells were thawed from liquid nitrogen and plated in gelatin-coated flasks at 5,000 cells/cm 2 , grown for 48 hours in Growth Medium and then grown for further 8 hours in 10 milliliters of serum starvation medium [DMEM-low glucose (Gibco, Carlsbad, Calif.), penicillin/streptomycin (Gibco, Carlsbad, Calif.) and 0.1% (w/v) Bovine Serum Albumin (BSA; Sigma, St. Louis, Mo.)]. After this treatment RNA was extracted and the supernatants were centrifuged at 150 ⁇ g for 5 minutes to remove cellular debris. Supernatants were then frozen at ⁇ 80° C. for ELISA analysis.
  • serum starvation medium DMEM-low glucose (Gibco, Carlsbad, Calif.), penicillin/streptomycin (Gibco, Carlsbad, Calif.) and 0.1% (w/v) Bovine Serum Albumin (BSA; Sigma, St. Louis, Mo
  • Cell culture for ELISA assay Postpartum cells derived from placenta and umbilicus, as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium in gelatin-coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen. Cells were thawed and transferred to 15-milliliter centrifuge tubes. After centrifugation at 150 ⁇ g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 milliliters culture medium and counted. Cells were grown in a 75 cm 2 flask containing 15 milliliters of Growth Medium at 375,000 cell/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation medium was collected at the end of incubation, centrifuged at 14,000 ⁇ g for 5 minutes (and stored at ⁇ 20° C.).
  • tyrpsin/EDTA Gibco, Carlsbad, Calif.
  • trypsin activity was neutralized with 8 milliliters of Growth Medium.
  • Cells were transferred to a 15 milliliters centrifuge tube and centrifuged at 150 ⁇ g for 5 minutes. Supernatant was removed and 1 milliliter Growth Medium was added to each tube to resuspend the cells. Cell number was estimated using a hemocytometer.
  • ELISA assay The amount of IL-8 secreted by the cells into serum starvation medium was analyzed using ELISA assays (R&D Systems, Minneapolis, Minn.). All assays were tested according to the instructions provided by the manufacturer.
  • RNA isolation was extracted from confluent postpartum-derived cells and fibroblasts or for IL-8 expression from cells treated as described above. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.). RNA was extracted according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, CA) and subjected to DNase treatment (2.7 U/sample) (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at ⁇ 80° C.
  • Genes identified by cDNA microarray as uniquely regulated in postpartum cells were further investigated using real-time and conventional PCR.
  • PCR was performed on cDNA samples using Assays-on-DemandTM gene expression products: oxidized LDL receptor (Hs00234028); rennin (Hs00166915); reticulon (Hs00382515); CXC ligand 3 (Hs00171061); GCP-2 (Hs00605742); IL-8 (Hs100174103); and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems, Foster City, Calif.).
  • PPDCs were fixed with cold 4% (w/v) paraformaldehyde (Sigma-Aldrich, St. Louis, Mo.) for 10 minutes at room temperature.
  • One isolate each of umbilicus- and placenta-derived cells at passage 0 (P0) (directly after isolation) and passage 11 (P11) (two isolates of placenta-derived, two isolates of umbilicus-derived cells) and fibroblasts (P11) were used.
  • Immunocytochemistry was performed using antibodies directed against the following epitopes: vimentin (1:500, Sigma, St.
  • anti-human GRO alpha—PE (1:100; Becton Dickinson, Franklin Lakes, N.J.
  • anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA)
  • anti-human oxidized LDL receptor 1 ox-LDL R1; 1:100; Santa Cruz Biotech
  • anti-human NOGA-A (1:100; Santa Cruz, Biotech).
  • fluorescence was visualized using an appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Antibody was added to aliquots as per manufactures specifications and the cells were incubated for in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess antibody. Cells requiring a secondary antibody were resuspended in 100 microliters of 3% FBS. Secondary antibody was added as per manufactures specification and the cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess secondary antibody. Washed cells were resuspended in 0.5 milliliters PBS and analyzed by flow cytometry.
  • oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROa (555042; BD Pharmingen, Bedford, Mass.), Mouse IgG1 kappa, (P-4685 and M-5284; Sigma), Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.).
  • FACScalibur Becton Dickinson San Jose, Calif.
  • results of real-time PCR for selected “signature” genes performed on cDNA from cells derived from human placentae, adult and neonatal fibroblasts and Mesenchymal Stem Cells (MSCs) indicate that both oxidized LDL receptor and rennin were expressed at higher level in the placenta-derived cells as compared to other cells.
  • the data obtained from real-time PCR were analyzed by the ⁇ CT method and expressed on a logarithmic scale. Levels of reticulon and oxidized LDL receptor expression were higher in umbilicus-derived cells as compared to other cells. No significant difference in the expression levels of CXC ligand 3 and GCP-2 were found between postpartum-derived cells and controls.
  • cytokine, IL-8 in postpartum was elevated in both Growth Medium-cultured and serum-starved postpartum-derived cells. All real-time PCR data was validated with conventional PCR and by sequencing PCR products.
  • Placenta-derived cells were also examined for the production of oxidized LDL receptor, GCP-2 and GROalpha by FACS analysis. Cells tested positive for GCP-2. Oxidized LDL receptor and GRO were not detected by this method.
  • Placenta-derived cells were also tested for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells derived from the human placenta were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Cells stained positive for both alpha-smooth muscle actin and vimentin. This pattern was preserved through passage 11. Only a few cells ( ⁇ 5%) at passage 0 stained positive for cytokeratin 18.
  • Postpartum-derived cells were evaluated in vitro for their immunological characteristics in an effort to predict the immunological response, if any, these cells would elicit upon in vivo transplantation.
  • PPDCs were assayed by flow cytometry for the presence of HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2. These proteins are expressed by antigen-presenting cells (APC) and are required for the direct stimulation of na ⁇ ve CD4 + T cells (Abbas & Lichtman, C ELLULAR AND M OLECULAR I MMUNOLOGY , 5th Ed. (2003) Saunders, Philadelphia, p. 171).
  • APC antigen-presenting cells
  • the cell lines were also analyzed by flow cytometry for the expression of HLA-G (Abbas & Lichtman, 2003, supra), CD 178 (Coumans, et al., (1999) Journal of Immunological Methods 224, 185-196), and PD-L2 (Abbas & Lichtman, 2003, supra; Brown, et. al. (2003) The Journal of Immunology 170, 1257-1266).
  • HLA-G Abbas & Lichtman, 2003, supra
  • CD 178 Cells, et al., (1999) Journal of Immunological Methods 224, 185-196
  • PD-L2 Abbas & Lichtman, 2003, supra; Brown, et. al. (2003) The Journal of Immunology 170, 1257-1266.
  • the expression of these proteins by cells residing in placental tissues is thought to mediate the immuno-privileged status of placental tissues in utero.
  • placenta- and umbilicus-derived cell lines were tested in a one-way mixed
  • Cell culture Cells were cultured to confluence in Growth Medium containing penicillin/streptomycin in T75 flasks (Corning, Corning, N.Y.) coated with 2% gelatin (Sigma, St. Louis, Mo.).
  • PBMCs Peripheral blood mononuclear cells
  • Stimulator (donor) allogeneic PBMC, autologous PBMC, and postpartum cell lines were treated with mitomycin C.
  • Autologous and mitomycin C-treated stimulator cells were added to responder (recipient) PBMCs and cultured for 4 days. After incubation, [ 3 H]thymidine was added to each sample and cultured for 18 hours. Following harvest of the cells, radiolabeled DNA was extracted, and [ 3 H]-thymidine incorporation was measured using a scintillation counter.
  • the stimulation index for the allogeneic donor was calculated as the mean proliferation of the receiver plus mitomycin C-treated allogeneic donor divided by the baseline proliferation of the receiver.
  • the stimulation index of the PPDCs was calculated as the mean proliferation of the receiver plus mitomycin C-treated postpartum cell line divided by the baseline proliferation of the receiver.
  • Mixed lymphocyte reaction umbilicus-derived cells. Six human volunteer blood donors were screened to identify a single allogeneic donor that will exhibit a robust proliferation response in a mixed lymphocyte reaction with the other five blood donors. This donor was selected as the allogeneic positive control donor. The remaining five blood donors were selected as recipients. The allogeneic positive control donor and placenta cell lines were mitomycin C-treated and cultured in a mixed lymphocyte reaction with the five individual allogeneic receivers. Reactions were performed in triplicate using two cell culture plates with three receivers per plate (Table 11-4). The average stimulation index ranged from 6.5 (plate 1) to 9 (plate 2) and the allogeneic donor positive controls ranged from 42.75 (plate 1) to 70 (plate 2) (Table 11-5).
  • Antigen presenting cell markers placenta-derived cells. Histograms of placenta-derived cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, as noted by fluorescence value consistent with the IgG control, indicating that placental cell lines lack the cell surface molecules required to directly stimulate CD4 + T cells.
  • Immunomodulating markers placenta-derived cells. Histograms of placenta-derived cells analyzed by flow cytometry show positive expression of PD-L2, as noted by the increased value of fluorescence relative to the IgG control, and negative expression of CD178 and HLA-G, as noted by fluorescence value consistent with the IgG control.
  • Antigen presenting cell markers umbilicus-derived cells. Histograms of umbilicus-derived cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, as noted by fluorescence value consistent with the IgG control, indicating that umbilical cell lines lack the cell surface molecules required to directly stimulate CD4 + T cells.
  • Immunomodulating cell markers umbilicus-derived cells. Histograms of umbilicus-derived cells analyzed by flow cytometry show positive expression of PD-L2, as noted by the increased value of fluorescence relative to the IgG control, and negative expression of CD178 and HLA-G, as noted by fluorescence value consistent with the IgG control.
  • the average stimulation index ranged from 1.3 to 3, and that of the allogeneic positive controls ranged from 46.25 to 279.
  • the average stimulation index ranged from 6.5 to 9, and that of the allogeneic positive controls ranged from 42.75 to 70.
  • Placenta- and umbilicus-derived cell lines were negative for the expression of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as measured by flow cytometry.
  • Placenta- and umbilicus-derived cell lines were negative for the expression of immuno-modulating proteins HLA-G and CD178 and positive for the expression of PD-L2, as measured by flow cytometry.
  • Allogeneic donor PBMCs contain antigen-presenting cells expressing HLA-DR, DQ, CD8, CD86, and B7-H2, thereby allowing for the stimulation of na ⁇ ve CD4 + T cells.
  • the absence of antigen-presenting cell surface molecules on placenta- and umbilicus-derived cells required for the direct stimulation of na ⁇ ve CD4 + T cells and the presence of PD-L2, an immunomodulating protein, may account for the low stimulation index exhibited by these cells in a MLR as compared to allogeneic controls.
  • HGF hepatocyte growth factor
  • MCP-1 monocyte chemotactic protein 1
  • IL-8 interleukin-8
  • keratinocyte growth factor KGF
  • basic fibroblast growth factor bFGF
  • VEGF vascular endothelial growth factor
  • TPO matrix metalloproteinase 1
  • ANG2 angiopoietin 2
  • PDGF-bb platelet derived growth factor
  • TPO thrombopoietin
  • HB-EGF stromal-derived factor 1 alpha
  • SDF-1alpha stromal-derived factor 1 alpha
  • chemokine activity such as macrophage inflammatory protein 1 alpha (MIP1a), macrophage inflammatory protein 1beta (MIP1b), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), 1309, thymus and activation-regulated chemokine (TARC), Eotaxin, macrophage-derived chemokine (MDC), IL-8).
  • MIP1a macrophage inflammatory protein 1 alpha
  • MIP1b macrophage inflammatory protein 1beta
  • MCP-1 monocyte chemoattractant-1
  • Rantes regulated on activation, normal T cell expressed and secreted
  • TARC thymus and activation-regulated chemokine
  • Eotaxin Eotaxin
  • MDC macrophage-derived chemokine
  • PPDCs from placenta and umbilicus as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium with penicillin/streptomycin on gelatin-coated T75 flasks. Cells were cryopreserved at passage 11 and stored in liquid nitrogen. After thawing of the cells, Growth Medium was added to the cells followed by transfer to a 15 milliliter centrifuge tube and centrifugation of the cells at 150 ⁇ g for 5 minutes. The supernatant was discarded. The cell pellet was resuspended in 4 milliliters Growth Medium, and cells were counted.
  • Cells were seeded at 375,000 cells/75 cm 2 flask containing 15 milliliters of Growth Medium and cultured for 24 hours. The medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin/streptomycin (Gibco)) for 8 hours. Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000 ⁇ g for 5 minutes and stored at ⁇ 20° C. To estimate the number of cells in each flask, cells were washed with PBS and detached using 2 milliliters trypsin/EDTA. Trypsin activity was inhibited by addition of 8 milliliters Growth Medium. Cells were centrifuged at 150 ⁇ g for 5 minutes. Supernatant was removed, and cells were resuspended in 1 milliliter Growth Medium. Cell number was estimated using a hemocytometer.
  • ELISA assay Cells were grown at 37° C. in 5% carbon dioxide and atmospheric oxygen. Placenta-derived cells (batch 101503) also were grown in 5% oxygen or beta-mercaptoethanol (BME). The amount of MCP-1, IL-6, VEGF, SDF-1alpha, GCP-2, IL-8, and TGF-beta 2 produced by each cell sample was measured by an ELISA assay (R&D Systems, Minneapolis, Minn.). All assays were performed according to the manufacturer's instructions.
  • the arrays are produced by spotting a 2 ⁇ 2, 3 ⁇ 3, or 4 ⁇ 4 pattern of four to 16 different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture chemiluminescent signal generated at each spot within each well of the plate. The amount of signal generated in each spot is proportional to the amount of target protein in the original standard or sample.
  • MCP-1 and IL-6 were secreted by placenta- and umbilicus-derived cells and dermal fibroblasts (Table 12-1). SDF-1 alpha was secreted by placenta-derived cells cultured in 5% O 2 and by fibroblasts.
  • GCP-2 and IL-8 were secreted by umbilicus-derived cells and by placenta-derived cells cultured in the presence of BME or 5% O 2 . GCP-2 also was secreted by human fibroblasts. TGF-beta2 was not detectable by ELISA assay.
  • TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1b, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were secreted from umbilicus-derived cells (Tables 12-2 and 12-3).
  • TIMP1, TPO, KGF, HGF, HBEGF, BDNF, MIP1a, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were secreted from placenta-derived cells (Tables 12-2 and 12-3). No Ang2, VEGF, or PDGF-bb were detected.
  • Umbilicus- and placenta-derived cells secreted a number of trophic factors. Some of these trophic factors, such as HGF, bFGF, MCP-1 and IL-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL-6, have important roles in neural regeneration.
  • placenta- and umbilicus-derived cells collectively postpartum-derived cells or PPDCs
  • mesenchymal stem cells P3; 1F2155; Cambrex, Walkersville, MD
  • osteoblasts P5; CC2538; Cambrex
  • adipose-derived cells Artecel, U.S. Pat. No. 6,555,374 B1 (P6; Donor 2)
  • neonatal human dermal fibroblasts P6; CC2509; Cambrex
  • DMEM/F12 medium Invitrogen, Carlsbad, Calif.
  • FBS fetal bovine serum
  • bFGF basic fibroblast growth factor
  • EGF epidermal growth factor
  • Peprotech penicillin/streptomycin
  • PPDCs umbilicus (022803) P11; placenta (042203) P11
  • adult human dermal fibroblasts (1F1853, P11) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached.
  • Cells were then trypsinized and seeded at similar density as in (A), but onto (1) 24 well tissue culture-treated plates (TCP, Falcon brand, VWR International), (2) TCP wells+2% (w/v) gelatin adsorbed for 1 hour at room temperature, or (3) TCP wells+20 ⁇ g/milliliter adsorbed mouse laminin (adsorbed for a minimum of 2 hours at 37° C.; Invitrogen).
  • NPE Neural Progenitor Expansion medium
  • PPDCs umbilicus (042203) P11, placenta (022803) P11), adult human dermal fibroblasts (P11; 1F1853; Cambrex) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached.
  • NPE+F+E adult rat neural progenitors isolated from hippocampus
  • Umbilicus-derived cells (P11; (042203)) were thawed and culture expanded in Growth Medium at 5,000 cells/cm 2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm 2 , onto 24 well laminin-coated plates (BD Biosciences) in the presence of NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter). In addition, some wells contained NPE+F+E+2% FBS or 10% FBS.
  • Neural progenitor co-culture protocol Adult rat hippocampal progenitors (062603) were plated as neurospheres or single cells (10,000 cells/well) onto laminin-coated 24 well dishes (BD Biosciences) in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter).
  • umbilicus-derived cells (042203) P11 and placenta-derived cells (022803) P11 were thawed and culture expanded in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter) at 5,000 cells/cm 2 for a period of 48 hours. Cells were then trypsinized and seeded at 2,500 cells/well onto existing cultures of neural progenitors. At that time, existing medium was exchanged for fresh medium. Four days later, cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nuclear protein (hNuc; Chemicon) (Table NU1-1 above) to identify PPDCs.
  • Immunocytochemistry was performed using the antibodies listed in Table NU1-1. Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature.
  • PBS phosphate-buffered saline
  • Triton Triton X-100
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Umbilicus and placenta PPDC isolates (as well as human fibroblasts and rodent neural progenitors as negative and positive control cell types, respectively) were plated on laminin (neural promoting)-coated dishes and exposed to 13 different growth conditions (and two control conditions) known to promote differentiation of neural progenitors into neurons and astrocytes.
  • laminin neural promoting
  • two control conditions two conditions were added to examine the influence of GDF5, and BMP7 on PPDC differentiation.
  • a two-step differentiation approach was taken, where the cells were first placed in neural progenitor expansion conditions for a period of 6 days, followed by full differentiation conditions for 7 days.
  • Neural progenitor and PPDC co-culture were plated onto cultures of rat neural progenitors seeded two days earlier in neural expansion conditions (NPE+F+E). While visual confirmation of plated PPDCs proved that these cells were plated as single cells, human-specific nuclear staining (hNuc) 4 days post-plating (6 days total) showed that they tended to ball up and avoid contact with the neural progenitors. Furthermore, where PPDCs attached, these cells spread out and appeared to be innervated by differentiated neurons that were of rat origin, suggesting that the PPDCs may have differentiated into muscle cells. This observation was based upon morphology under phase contrast microscopy.
  • cultures of neural progenitors and PPDCs had more cells of rat origin and larger amounts of differentiation than control cultures of neural progenitors alone, further indicating that plated PPDCs provided soluble factors and or contact-dependent mechanisms that stimulated neural progenitor survival, proliferation, and/or differentiation.
  • umbilicus and placenta-derived cells collectively postpartum-derived cells or PPDCs
  • NPE Neural Progenitor Expansion
  • NPE media was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ), herein referred to as NPE+bFGF+EGF.
  • Immunocytochemistry After a period of one month, all flasks were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against TuJ1 (BIII Tubulin; 1:500; Sigma, St. Louis, Mo.) and GFAP (glial fibrillary acidic protein; 1:2000; DakoCytomation, Carpinteria, CA).
  • TuJ1 BIII Tubulin; 1:500; Sigma, St. Louis, Mo.
  • GFAP glial fibrillary acidic protein; 1:2000; DakoCytomation, Carpinteria, CA.
  • cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens.
  • Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature.
  • NPE+bFGF+EGF media slows proliferation of PPDCs and alters their morphology.
  • PPDCs attached to the culture flasks coated with laminin. This may have been due to cell death as a function of the freeze/thaw process or because of the new growth conditions. Cells that did attach adopted morphologies different from those observed in Growth Media.
  • Clones of umbilicus-derived cells express neuronal proteins. Cultures were fixed at one month post-thawing/plating and stained for the neuronal protein TuJ1 and GFAP, an intermediate filament found in astrocytes. While all control cultures grown in Growth Medium and human fibroblasts and MSCs grown in NPE+bFGF+EGF medium were found to be TuJ1-/GFAP-, TuJ1 was detected in the umbilicus and placenta PPDCs. Expression was observed in cells with and without neuronal-like morphologies. No expression of GFAP was observed in either culture.
  • umbilicus- and placenta-derived cells collectively postpartum-derived cells or PPDCs
  • PPDCs postpartum-derived cells
  • NPE medium was washed in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement; Invitrogen), L-glutamine (4 mM; Invitrogen), and penicillin/streptomycin (Invitrogen), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) medium.
  • NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ), herein referred to as NPE+bFGF+EGF.
  • tissue minced with a scalpel Following wash, the overlying meninges were removed, and the tissue minced with a scalpel. Minced tissue was collected and trypsin/EDTA (Invitrogen) added as 75% of the total volume. DNAse (100 microliters per 8 milliliters total volume, Sigma, St. Louis, Mo.) was also added. Next, the tissue/media was sequentially passed through an 18 gauge needle, 20 gauge needle, and finally a 25 gauge needle one time each (all needles from Becton Dickinson, Franklin Lakes, N.J.). The mixture was centrifuged for 3 minutes at 250 g. Supernatant was removed, fresh NPE+bFGF+EGF was added and the pellet resuspended.
  • trypsin/EDTA Invitrogen
  • the resultant cell suspension was passed through a 40 micrometer cell strainer (Becton Dickinson), plated on laminin-coated T-75 flasks (Becton Dickinson) or low cluster 24-well plates (Becton Dickinson), and grown in NPE+bFGF+EGF media until sufficient cell numbers were obtained for the studies outlined.
  • Postpartum-derived cells (umbilicus (022803) P12, (042103) P12, (071003) P12; placenta (042203) P12) previously grown in Growth Medium were plated at 5,000 cells/transwell insert (sized for 24 well plate) and grown for a period of one week in Growth Medium in inserts to achieve confluence.
  • Neural progenitor plating Neural progenitors, grown as neurospheres or as single cells, were seeded onto laminin-coated 24 well plates at an approximate density of 2,000 cells/well in NPE+bFGF+EGF for a period of one day to promote cellular attachment. One day later, transwell inserts containing postpartum cells were added according to the following scheme:
  • Immunocytochemistry After 7 days in co-culture, all conditions were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the epitopes listed in Table 15-1. Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature.
  • PBS phosphate-buffered saline
  • Triton Triton X-100
  • fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Quantitative analysis of neural progenitor differentiation Quantification of hippocampal neural progenitor differentiation was examined. A minimum of 1000 cells were counted per condition or if less, the total number of cells observed in that condition. The percentage of cells positive for a given stain was assessed by dividing the number of positive cells by the total number of cells as determined by DAPI (nuclear) staining.
  • Mass spectrometry analysis & 2D gel electrophoresis In order to identify unique, secreted factors as a result of co-culture, conditioned media samples taken prior to culture fixation were frozen down at ⁇ 80° C. overnight. Samples were then applied to ultrafiltration spin devices (MW cutoff 30 kD). Retentate was applied to immunoaffinity chromatography (anti-Hu-albumin; IgY) (immunoaffinity did not remove albumin from the samples). Filtrate was analyzed by MALDI. The pass through was applied to Cibachron Blue affinity chromatography. Samples were analyzed by SDS-PAGE and 2D gel electrophoresis.
  • PPDC co-culture stimulates adult neural progenitor differentiation.
  • co-cultured neural progenitor cells derived from adult rat hippocampus exhibited significant differentiation along all three major lineages in the central nervous system. This effect was clearly observed after five days in co-culture, with numerous cells elaborating complex processes and losing their phase bright features characteristic of dividing progenitor cells. Conversely, neural progenitors grown alone in the absence of bFGF and EGF appeared unhealthy and survival was limited.
  • cultures were stained for markers indicative of undifferentiated stem and progenitor cells (nestin), immature and mature neurons (TuJ1), astrocytes (GFAP), and mature oligodendrocytes (MBP). Differentiation along all three lineages was confirmed while control conditions did not exhibit significant differentiation as evidenced by retention of nestin-positive staining amongst the majority of cells. While both umbilicus- and placenta-derived cells induced cell differentiation, the degree of differentiation for all three lineages was less in co-cultures with placenta-derived cells than in co-cultures with umbilicus-derived cells.
  • PPDCs postpartum-derived cells
  • Placenta- and umbilicus-derived cells were grown in Growth Medium (DMEM-low glucose (Gibco, Carlsbad Calif.), 15% (v/v) fetal bovine serum (Cat. #SH30070.03; Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma, St. Louis, Mo.), penicillin/streptomycin (Gibco)) in a gelatin-coated flasks.
  • Growth Medium DMEM-low glucose (Gibco, Carlsbad Calif.), 15% (v/v) fetal bovine serum (Cat. #SH30070.03; Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma, St. Louis, Mo.), penicillin/streptomycin (Gibco)
  • RAD16 self-assembling peptides (3D Matrix, Cambridge, Mass. under a material transfer agreement) was obtained as a sterile 1% (w/v) solution in water, which was mixed 1:1 with 1 ⁇ 10 6 cells in 10% (w/v) sucrose (Sigma, St Louis, Mo.), 10 mM HEPES in Dulbecco's modified medium (DMEM; Gibco) immediately before use.
  • the final concentration of cells in RAD16 hydrogel was 1 ⁇ 10 6 cells/100 microliters.
  • mice Mus Musculus /Fox Chase SCID/Male (Harlan Sprague Dawley, Inc., Indianapolis, Ind.), 5 weeks of age. All handling of the SCID mice took place under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligrams/kg KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligrams/kg ROMPUN (xylazine, Mobay Corp., Shawnee, Kansas) and saline.
  • KETASET ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa
  • ROMPUN xylazine, Mobay Corp., Shawnee, Kansas
  • Subcutaneous Implantation Technique Four skin incisions, each approximately 1.0 cm in length, were made on the dorsum of the mice. Two cranial sites were located transversely over the dorsal lateral thoracic region, about 5-mm caudal to the palpated inferior edge of the scapula, with one to the left and one to the right of the vertebral column. Another two were placed transversely over the gluteal muscle area at the caudal sacro-lumbar level, about 5-mm caudal to the palpated iliac crest, with one on either side of the midline. Implants were randomly placed in these sites in accordance with the experimental design.
  • the skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected for RAD16) about 1-cm caudal to the incision.
  • the appropriate test material was implanted into the subcutaneous space. The skin incision was closed with metal clips.
  • mice were individually housed in microisolator cages throughout the course of the study within a temperature range of 64° F.-79° F. and relative humidity of 30% to 70%, and maintained on an approximate 12 hour light/12 hour dark cycle. The temperature and relative humidity were maintained within the stated ranges to the greatest extent possible. Diet consisted of Irradiated Pico Mouse Chow 5058 (Purina Co.) and water fed ad libitum.
  • mice were euthanized at their designated intervals by carbon dioxide inhalation.
  • the subcutaneous implantation sites with their overlying skin were excised and frozen for histology.
  • Synthetic absorbable non-woven/foam discs (5.0 mm diameter ⁇ 1.0 mm thick) or self-assembling peptide hydrogel were seeded with either cells derived from human umbilicus or placenta and implanted subcutaneously bilaterally in the dorsal spine region of SCID mice.
  • Retinal ganglion cell (RGC) lesions have been extensively used as models for various repair strategies in the adult mammalian CNS. It has been demonstrated that retrobulbar section of adult rodent RGC axons results in abortive sprouting (Zeng et al., 1995) and progressive death of the parent cell population (Villegas-Perez et al., 1993). Numerous studies have demonstrated the stimulatory effects of various exogenous and endogenous factors on the survival of axotomized RGC's and regeneration of their axons (Yip and So, 2000; Fischer et al., 2001).
  • cell transplants can be used to promote regeneration of severed nerve axons (L1 et al., 2003; Ramon-Cueto et al., 2000).
  • cell based therapy can be utilized for the treatment of neural disorders that affect the spinal cord, peripheral nerves, pudendal nerves, optic nerves or other diseases/trauma due to injury in which nervous damage can occur.
  • Self-assembling peptides have been developed to act as a scaffold for cell-attachment to encapsulate cells in 3-D, plate cells in 2-D coatings, or as microcarriers in suspension cultures.
  • Three-dimensional cell culture has required either animal-derived materials (mouse sarcoma extract), with their inherent reproducibility and cell signaling issues, or much larger synthetic scaffolds, which fail to approximate the physical nanometer-scale and chemical attributes of native ECM.
  • RAD 16 (NH2-(RADA) 3 -COOH) and KLD (NH2-(KLDL) 3 -COOH) are synthesized in small (RAD16 is 5 nanometers) oligopeptide fragments that self-assemble into nanofibers on a scale similar to the in vivo extracellular matrix (ECM) (3D Matrix, Inc Cambridge, Mass.). The self-assembly is initiated by mono- or di-valent cations found in culture media or the physiological environment. In the protocols described in this example, RAD 16 was used as a microcarrier for the implantation of postpartum cells into the ocular defect. In this example, it is demonstrated that transplants of postpartum-derived cells PPDCs) can provide efficacy in an adult rat optic nerve axonal regeneration model.
  • ECM extracellular matrix
  • mice were injected with dexamethasone (2 milligrams/kilogram) for 10 days post transplantation.
  • dexamethasone 2 milligrams/kilogram
  • animals were maintained on oral cyclosporine A (210 milligrams/liter of drinking water; resulting blood concentration: 250-300 micrograms/liter) (Bedford Labs, Bedford, Ohio) from 2 days pre-transplantation until end of the study. Food and water were available ad libitum. Animals were sacrificed at either 30 or 60 days posttransplantation.
  • CTB Application Three days before animals were sacrificed, under anesthesia, a glass micropipette with a 30-50 millimeter tip was inserted tangentially through the sclera behind the lens, and two 4-5 microliter aliquots of a 1% retrograde tracer-cholera toxin B (CTB) aqueous solution (List Biologic, Campbell, CA) was injected into the vitreous. Animals were perfused with fixative and optic nerves were collected in the same fixative for 1 hour. The optic nerves were transferred into sucrose overnight.
  • CTB retrograde tracer-cholera toxin B
  • Rats were perfused with 4% paraformaldehyde. Rats were given 4 cubic centimeters of urethane and were then perfused with PBS (0.1 molar) then with 4% Para formaldehyde. The spinal cord was cut and the bone removed from the head to expose the colliculus. The colliculus was then removed and placed in 4% paraformaldehyde. The eye was removed by cutting around the outside of the eye and going as far back as possible. Care was given not to cut the optic nerve that lies on the underside of the eye. The eye was removed and the muscles were cut exposing the optic nerve this was then placed in 4% paraformaldehyde.
  • RAD-16 transplantation Following transplantation of RAD-16 into the cut, visible ingrowth of vascularized connective tissue was observed. However, no axonal in growth was observed between the proximal and distal stumps. The results demonstrate that application of RAD-16 alone is not sufficient for inducing axonal regeneration in this situation.
  • Transplantation of postpartum-derived cells Transplantation of postpartum-derived cells into the severed optic nerve stimulated optic nerve regrowth. Some regrowth was also observed in conditions in which fibroblast cells were implanted, although this was minimal as compared with the regrowth observed with the transplanted placenta-derived cells. Optic nerve regrowth was observed in 4/5 animals transplanted with placenta-derived cells, 3/6 animals transplanted with adult dermal fibroblasts and in 1 ⁇ 4 animals transplanted with umbilicus-derived cells. In situations where regrowth was observed, CTB labeling confirmed regeneration of retinal ganglion cell axons, which were demonstrated to penetrate through the transplant area. GFAP labeling was also performed to determine the level of glial scarring. The GFAP expression was intensified at the proximal stump with some immunostaining being observed through the reinervated graft.
  • RP retinitis pigmentosa
  • the retinal photoreceptors and adjacent retinal pigment epithelium form a functional unit.
  • the Royal College of Surgeons (RCS) rat presents with a tyrosine receptor kinase (Merkt) defect affecting outer segment phagocytosis, leading to photoreceptor cell death (1).
  • Transplantation of retinal pigment epithelial (RPE) cells into the subretinal space of RCS rats was found to limit the progress of photoreceptor loss and preserve visual function (2).
  • RPE retinal pigment epithelial
  • ERG recordings Following overnight dark adaptation, animals were prepared for ERG recording under dim red light, as previously described (3).
  • anesthesia with a mixture of 150 mg/kg i.p ketamine, and 10 mg/kg i.p. xylazine
  • the head was secured with a stereotaxic head holder and the body temperature monitored through a rectal thermometer and maintained at 38° C. using a homeothermic blanket.
  • Pupils were dilated using equal parts of topical 2.5% phenylephrine and 1% tropicamide.
  • Topical anesthesia with 0.75% bupivacaine was used to prevent any corneal reflexes and a drop of 0.9% saline was frequently applied on the cornea to prevent its dehydration and allow electrical contact with the recording electrode (gold wire loop).
  • Amplification (at 1-1000 Hz bandpass, without notch filtering), stimulus presentation, and data acquisition were provided by the UTAS-3000 system from LKC Technologies (Gaithersburg, Md.). ERGs were recorded at 60 and 90 days of age in the umbilical cell groups and at 60 days only in the placental and fibroblast groups.
  • V max from fitting the data with a Naka-Rushton curve was not used because ERG responses were often erratic at higher luminance levels in dystrophic animals and showed tendencies for depressed responses around 0.4 and 1.4 log candila/m 2 .
  • criterion amplitudes were used: 20 ⁇ V for a- and b-waves, and 10 ⁇ V for STR-like responses. The amplitude of the b-wave was measured from the a-wave negative peak up to the b-wave positive apex, and not up to the peak of oscillations, which can exceed the b-wave apex (4).
  • the double flash protocol was used to determine the isolation of rod and cone responses (5).
  • a probe flash was presented 1 sec after a conditioning flash, using a specific feature of the UTAS-3000 system (LKC Technologies) with calibrated ganzfeld; assuring complete recharge of the stimulator under the conditions used.
  • the role of the conditioning flash in the procedure was to transiently saturate rods so that they were rendered unresponsive to the probe flash.
  • Response to the probe flash was taken as reflecting cone-driven activity.
  • a rod-driven b-wave was obtained by subtracting the cone-driven response from the mixed response (obtained following presentation of a probe flash alone, i.e. not preceded by any conditioning flash).
  • Physiological retinal sensitivity testing was performed to demonstrate retinal response to dim light. Animals were anesthetized with a recovery dose of urethane at 1.25 g/kg i.p. Physiological assessment in the animals was tested post graft in animals at 90 days by recording multiunit extracellular activity in the superior colliculus to illumination of respective visual receptive fields (6). This procedure was repeated for 20 independent points (spaced 200 mm apart, with each step corresponding to approximately 10-150 displacements in the visual field), covering the visual field.
  • Visual thresholds were measured as the increase in intensity over background and maintained at 0.02 candila/m 2 (luminescence unit) [at least 2.6 logarithm units below rod saturation (7)], required for activating units in the superficial 200 ⁇ m of the superior colliculus. Response parameters were compared between transplanted and sham control eyes that received vehicle alone.
  • the retinas were then post-fixed in 1% osmium tetroxide for 1 h. After dehydration through a series of alcohols to epoxypropane, the retinas were embedded in TAAB embedding resin (TAAB Laboratories, Aldemarston, UK). Semi-thin sections were stained with 1% toluidine Blue in 1% borate buffer and the ultra thin sections were contrasted with uranyl acetate and lead citrate.
  • Umbilical cell-transplanted animals demonstrated good improvement in all outcome measures tested at 60 days (Table N5-1), a-wave (27 ⁇ 11) versus sham controls (O), mixed b-wave (117 ⁇ 67) versus sham controls (18 ⁇ 13), cone-b-wave (55 ⁇ 25) versus sham controls (28 ⁇ 11), and in rod contribution (49 ⁇ 16%) versus sham controls (6 ⁇ 7%).
  • Efficacy of transplants in preventing visual loss was monitored by assessment of electrophysiological responsiveness in two animals.
  • the threshold sensitivity response to light was used to define the area of visual field rescue in sham-injected control eyes versus eyes transplanted with umbilical cells.
  • visual thresholds never exceeded 0.5 log candila/m 2 above background.
  • the thresholds are usually in the magnitude of 4 log candila/m 2 units (8).
  • the thresholds were in the order of 2.9-4.9 log candila/m 2 units with an average threshold of 4.0 log candila/m 2 units, in some instances no recording could be attained.
  • the sham-injected rats showed some highly localized functional rescue in the temporal retina.
  • the human umbilical transplanted rats exhibited substantially greater levels of visual preservation with thresholds ranging from 0.8 to 2.1 log candila/m 2 units, with an average threshold of 1.3 log candila/m 2 units.
  • ERG measurements provide an assessment of visual function after photoreceptor loss, indicating changes in electrical activity in the retina.
  • ERG does not provide direct information as to image forming capability.
  • the measurement of collicular threshold sensitivity used in this study provides an indication of relative preservation of visual fields. The importance of this measure is based on a correlation between the amounts of functional rescue and anatomical preservation and that the data collected compares with visual field perimetry testing in humans (9).
  • the transplantation has demonstrated a retardation of the disease process in the test animals.
  • the results presented herein demonstrate clear evidence of functional efficacy of grafting human PPDCs into the subretinal space, and that rescue occurs in the general region in which the grafted cells are located.

Abstract

Cells derived from postpartum umbilicus and placenta are disclosed. Pharmaceutical compositions, devices and methods for the regeneration or repair of ocular tissue using the postpartum-derived cells are also disclosed.

Description

  • This claims benefit of U.S. Provisional Application No. 60/483,264, filed Jun. 27, 2003, the entire contents of which are incorporated by reference herein. Other related applications include the following commonly-owned, co-pending applications, the entire contents of each of which are incorporated by reference herein: U.S. Application No. [ETH-5073 NP 1], filed Jun. 25, 2004, U.S. Application No. [ETH-5073 NP2], filed Jun. 25, 2004, U.S. Application No. [ETH-5073 NP3], filed Jun. 25, 2004, U.S. Application No. [ETH-5073 NP4], filed Jun. 25, 2004, U.S. Application No. [ETH-5073 NP7], filed Jun. 25, 2004, U.S. Application No. [ETH-5073 NP8], filed Jun. 25, 2004 and U.S. Provisional Application No. 60/555,908, filed Mar. 24, 2004.
  • FIELD OF THE INVENTION
  • This invention relates to the field of cell-based or regenerative therapy for ophthalmic diseases and disorders. In particular, the invention provides pharmaceutical compositions, devices and methods for the regeneration or repair of cells and tissues of the eye, using postpartum derived cells.
  • BACKGROUND OF THE INVENTION
  • Various patents and other publications are referred to throughout the specification. Each of these publications is incorporated by reference herein, in its entirety.
  • As a complex and sensitive organ of the body, the eye can experience numerous diseases and other deleterious conditions that affect its ability to function normally. Many of these conditions are associated with damage or degeneration of specific ocular cells, and tissues made up of those cells. As one example, diseases and degenerative conditions of the optic nerve and retina are the leading causes of blindness throughout the world. Damage or degeneration of the cornea, lens and associated ocular tissues represent another significant cause of vision loss worldwide.
  • The retina contains seven layers of alternating cells and processes that convert a light signal into a neural signal. The retinal photoreceptors and adjacent retinal pigment epithelium (RPE) form a functional unit that, in many disorders, becomes unbalanced due to genetic mutations or environmental conditions (including age). This results in loss of photoreceptors through apoptosis or secondary degeneration, which leads to progressive deterioration of vision and, in some instances, to blindness (for a review, see, e.g., Lund, R. D. et al., 2001, Progress in Retinal and Eye Research 20: 415-449). Two classes of ocular disorders that fall into this pattern are age-related macular degeneration (AMD) and retinitis pigmentosa (RP).
  • AMD is the most common cause of vision loss in the United States in those 50 or older, and its prevalence increases with age. The primary disorder in AMD appears to be due to RPE dysfunction and changes in Bruch's membranes, e.g., lipid deposition, protein cross-linking and decreased permeability to nutrients (see Lund et al., 2001 supra). A variety of elements may contribute to macular degeneration, including genetic makeup, age, nutrition, smoking and exposure to sunlight.
  • RP is mainly considered an inherited disease—over 100 mutations have been associated with photoreceptor loss (see Lund et al., 2001, supra). Though the majority of mutations target photoreceptors, some affect RPE cells directly. Together, these mutations affect such processes as molecular trafficking between photoreceptors and RPE cells and phototransduction, for example.
  • Other less common, but nonetheless debilitating retinopathies can also involve progressive cellular degeneration leading to vision loss and blindness. These include, for example, diabetic retinopathy and choroidal neovascular membrane (CNVM). Diabetic retinopathy may be classified as (1) non-proliferative or background retinopathy, characterized by increased capillary permeability, edema, hemorrhage, microaneurysms, and exudates, or 2) proliferative retinopathy, characterized by neovascularization extending from the retina to the vitreous, scarring, fibrous tissue formation, and potential for retinal detachment. In CNVM, abnormal blood vessels stemming from the choroid grow up through the retinal layers. The fragile new vessels break easily, causing blood and fluid to pool within the layers of the retina.
  • Damage or progressive degeneration of the optic nerve and related nerves of the eye constitutes another leading cause of vision loss and blindness. A prime example is glaucoma, a condition of the eye that is made up of a collection of eye diseases that cause vision loss by damage to the optic nerve. Elevated intraocular pressure (IOP) due to inadequate ocular drainage is a primary cause of glaucoma, but it can also develop in the absence of elevated IOP. Glaucoma can develop as the eye ages, or it can occur as the result of an eye injury, inflammation, tumor, or in advanced cases of cataract or diabetes, or it can be caused by certain drugs, such as steroids. The primary features of the optic neuropathy in glaucoma include characteristic changes in the optic nerve head, a decrease in number of surviving retinal ganglion cells, and loss of vision. It has been proposed that a cascade of events links degeneration of the optic nerve head with the slow death of retinal ganglion cells observed in the disease, and that this cascade of events can be slowed or prevented through the use of neuroprotective agents (Osborne et al., 2003, Eur. J. Ophthalmol. 13 (Supp 3): S19-S26).
  • Cellular damage and degenerative conditions also affect other parts of the eye. For example, cataracts result from gradual opacification of the crystalline lens of the eye. It is believed that once begun, cataract development proceeds along one or more common pathways that culminate in damage to lens fibers. This condition is presently treated by surgical removal and replacement of the affected lens. Another example concerns the cornea and surrounding conjuctiva that make up the ocular surface. The limbal epithelium, located between the cornea and the bulbar conjuctiva, contains corneal epithelial stem cells. Limbal epithelial cell deficiency (LECD) is a condition that occurs, for example, in Stevens-Johnson syndrome and thermal or chemical burns. LECD often leads to an imbalance between the corneal epithelium and the conjunctival epithelium in which the cornea is covered by invading conjunctival epithelial cells, which severely compromises the corneal surface and affects visual acuity (Nakamura, T. & Kinoshita, S., 2003. Cornea 22 (Supp. 1): S75-S80).
  • The recent advent of stem cell-based therapy for tissue repair and regeneration provides promising treatments for a number of aforementioned cell-degenerative pathologies and other ocular disorders. Stem cells are capable of self-renewal and differentiation to generate a variety of mature cell lineages. Transplantation of such cells can be utilized as a clinical tool for reconstituting a target tissue, thereby restoring physiologic and anatomic functionality. The application of stem cell technology is wide-ranging, including tissue engineering, gene therapy delivery, and cell therapeutics, i.e., delivery of biotherapeutic agents to a target location via exogenously supplied living cells or cellular components that produce or contain those agents (For a review, see Tresco, P. A. et al., 2000, Advanced Drug Delivery Reviews 42: 2-37).
  • An obstacle to realization of the therapeutic potential of stem cell technology has been difficulty in obtaining sufficient numbers of stem cells. One source of stem cells is embryonic or fetal tissue. Embryonic stem and progenitor cells have been isolated from a number of mammalian species, including humans, and several such cell types have been shown capable of self-renewal and expansion, as well differentiation into a variety of cell lineages. In animal model systems, embryonic stem cells have been reported to differentiate into a RPE cell phenotype, as well as to enhance the survival of host photoreceptors following transplantation (Haruta, M. et al., 2004, Investig. Ophthalmol. Visual Sci. 45: 1020-1025; Schraermeyer, U. et al., 2001, Cell Transplantation 10: 673-680). But the derivation of stem cells from embryonic or fetal sources has raised many ethical issues that are desirable to avoid by identifying other sources of multipotent or pluripotent cells.
  • Adult tissue also can yield stem cells useful for cell-based ocular therapy. For instance, retinal and corneal stem cells themselves may be utilized for cell replacement therapy in the eye. In addition, neural stem cells from the hippocampus have been reported to integrate with the host retina, adopting certain neural and glial characteristics (see review of Lund, R. L. et al., 2003, J. Leukocyte Biol. 74: 151-160). Neural stem cells prepared from fetal rat cortex were shown to differentiate along an RPE cell pathway following transplantation into the adult rat subretinal space (Enzmann, V. et al., 2003, Investig. Ophthalmol. Visual Sci. 44: 5417-5422). Bone marrow stem cells have been reported to differentiate into retinal neural cells and photoreceptors following transplantation into host retinas (Tomita, M. et al., 2002, Stem Cells 20: 279-283; Kicic, A. et al., 2003, J. Neurosci. 23: 7742-7749). An ocular surface reconstruction in a rabbit model system, utilizing cultured mucosal epithelial stem cells, has also been reported (Nakamura, T. & Kinoshita, S., 2003, supra). While these reports show promise for the use of adult progenitor and stem cells in cell-based therapy for the eye, it must be noted that adult stem cell populations are comparatively rare and are often obtainable only by invasive procedures. Further, adult stem cells may have a more limited ability to expand in culture than do embryonic stem cells.
  • Thus, a need exists for alternative sources of adequate supplies of cells having the ability to support, augment or replace lost cellular function in the eye. A reliable, well-characterized and plentiful supply of substantially homogeneous populations of such cells would be an advantage in a variety of diagnostic and therapeutic applications in ocular repair and regeneration, including drug screening assays, ex vivo or in vitro trophic support of ocular and other useful cell types, and in vivo cell-based therapy.
  • SUMMARY OF THE INVENTION
  • This invention provides compositions and methods applicable to cell-based or regenerative therapy for ophthalmic diseases and disorders. In particular, the invention features pharmaceutical compositions, devices and methods for the regeneration or repair of ocular tissue using postpartum-derived cells.
  • One aspect of the invention features an isolated postpartum-derived cell, derived from human placental or umbilical cord tissue substantially free of blood, wherein the cell is capable of self-renewal and expansion in culture and has the potential to differentiate into a cell of a neural phenotype; wherein the cell requires L-valine for growth and is capable of growth in at least about 5% oxygen. This cell further comprises one or more of the following characteristics: (a) potential for at least about 40 doublings in culture; (b) attachment and expansion on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin, or fibronectin; (c) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; (d) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C; (e) lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; (f) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of a gene encoding: interleukin 8; reticulon 1; chemokine (C—X—C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C—X—C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C—X—C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547k1113; (g) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of a gene encoding: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis-2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); similar to neuralin 1; B cell translocation gene 1; hypothetical protein FLJ23191; and DKFZp586L151; (h) secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and (i) lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
  • In certain embodiments, the postpartum-derived cell is an umbilicus-derived cell. In other embodiments it is a placenta-derived cell. In specific embodiments, the cell has all identifying features of any one of: cell type PLA 071003 (P8) (ATCC Accession No. PTA-6074); cell type PLA 071003 (P11) (ATCC Accession No. PTA-6075); cell type PLA 071003 (P16) (ATCC Accession No. PTA-6079); cell type UMB 022803 (P7) (ATCC Accession No. PTA-6067); or cell type UMB 022803 (P17) (ATCC Accession No. PTA-6068).
  • In certain embodiments, postpartum-derived cells are isolated in the presence of one or more enzyme activities comprising metalloprotease activity, mucolytic activity and neutral protease activity. Preferably, the cells have a normal karyotype, which is maintained as the cells are passaged in culture. In preferred embodiments, the postpartum-derived cells comprise each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C and does not comprise any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
  • Another aspect of the invention features a cell population comprising the postpartum-derived cells as described above. In one embodiment, the population is a substantially homogeneous population of the postpartum-derived cells. In a specific embodiment, the population comprises a clonal cell line of the postpartum-derived cells. In another embodiment, the population is a heterogeneous population comprising the postpartum-derived cells and at least one other cell type. In certain embodiments, the other cell type is an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell or other multipotent or pluripotent stem cell. In other embodiments, the cell population is cultured in contact with one or more factors that stimulate stem cell differentiation toward a neural or epithelial lineage.
  • Also featured in accordance with the present invention is a cell lysate prepared from postpartum-derived cells. The cell lysate may be separated into a membrane enriched fraction and a soluble cell fraction. The invention also features an extracellular matrix produced by the postpartum-derived cells, as well as a conditioned medium in which the cells have been grown.
  • Another aspect of the invention features a method of treating a patient having an ocular degenerative condition, which comprises administering to the patient multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord, in an amount effective to treat the ocular degenerative condition. Preferably the cells are postpartum-derived cells, as described above. In certain embodiments, the ocular degenerative condition is an acute ocular degenerative condition such as brain trauma, optic nerve trauma or ocular lesion. In other embodiments, it is a chronic or progressive degenerative condition, such as macular degeneration, retinitis pigmentosa, diabetic retinopathy, glaucoma or limbal epithelial cell deficiency. In certain embodiments, the cells are induced in vitro to differentiate into a neural or epithelial lineage cells prior to administration.
  • In certain embodiments, the cells are administered with at least one other cell type, such as an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell. In these embodiments, the other cell type can be administered simultaneously with, or before, or after, the postpartum-derived cells. Likewise, in these and other embodiments, the cells are administered with at least one other agent, such as a drug for ocular therapy, or another beneficial adjunctive agent such as an anti-inflammatory agent, anti-apoptotic agents, antioxidants or growth factors. In these embodiments, the other agent can be administered simultaneously with, or before, or after, the postpartum cells.
  • In various embodiments, the cells are administered to the surface of an eye, or they are administered to the interior of an eye or to a location in proximity to the eye, e.g., behind the eye. The cells can be administered through a cannula or from a device implanted in the patient's body within or in proximity to the eye, or they may be administered by implantation of a matrix or scaffold containing the cells.
  • Another aspect of the invention features a pharmaceutical composition for treating a patient having an ocular degenerative condition, comprising a pharmaceutically acceptable carrier and multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord in an amount effective to treat the ocular degenerative condition. Preferably the postpartum cells are postpartum-derived cells as described above. The ocular degenerative condition may be an acute, chronic or progressive condition. In certain embodiments, the composition comprises cells that have been induced in vitro to differentiate into a neural or epithelial lineage cells prior to formulation of the composition.
  • In certain embodiments, the pharmaceutical composition comprises at least one other cell type, such as an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell. In these or other embodiments, the pharmaceutical composition comprises at least one other agent, such as a drug for treating the ocular degenerative disorder or other beneficial adjunctive agents, e.g., anti-inflammatory agents, anti-apoptotic agents, antioxidants or growth factors.
  • In certain embodiments, the pharmaceutical compositions are formulated for administration to the surface of an eye. Alternatively, they can be formulated for administration to the interior of an eye or in proximity to the eye (e.g., behind the eye). The compositions also can be formulated as a matrix or scaffold containing the cells.
  • According to yet another aspect of the invention, a kit is provided for treating a patient having an ocular degenerative condition. The kit comprises a pharmaceutically acceptable carrier, a population of multipotent or pluripotent cells isolated from postpartum placenta or umbilicus, preferably the postpartum-derived cells described above, and instructions for using the kit in a method of treating the patient. The kit may also contain one or more additional components, such as reagents and instructions for culturing the cells, or a population of at least one other cell type, or one or more agents useful in the treatment of an ocular degenerative condition.
  • According to another aspect of the invention, a method is provided for treating a patient having an ocular degenerative condition, which comprises administering to the patient a preparation made from multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord, in an amount effective to treat the ocular degenerative condition, wherein the preparation comprises a cell lysate of the cells (or fraction thereof), or a conditioned medium in which the cells were grown, or an extracellular matrix of the cells. Preferably, the cells are the postpartum-derived cells described above. In another aspect, the invention features a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a preparation made from the postpartum cells, which may be a cell lysate (or fraction thereof) of the postpartum cells, an extracellular matrix of the postpartum cells or a conditioned medium in which the postpartum cells were grown. Kits for practicing this aspect of the invention are also provided. These may include the one or more of a pharmaceutically acceptable carrier or other agent or reagent, one or more of a cell lysate or fraction thereof, an extracellular matrix or a conditioned medium from the postpartum cells, and instructions for use of the kit components.
  • Another aspect of the invention features a method for increasing the survival, growth or activity of cells for transplantation to treat an ocular degenerative disorder. The method comprises co-culturing the cells for transplantation with cultured cells derived from postpartum placental or umbilical tissue, under conditions effective to increase the survival, growth or activity of the cells for transplantation. Preferably, the postpartum cells are the postpartum-derived cells described above. A kit for practicing the method is also provided. The kit comprises the cultured postpartum cells and instructions for co-culturing the cells for transplantation with the postpartum cells under conditions effective to increase the survival, growth or activity of the cells for transplantation.
  • Other features and advantages of the invention will be apparent from the detailed description and examples that follow.
  • DETAILED DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
  • Definitions
  • Various terms used throughout the specification and claims are defined as set forth below.
  • Stem cells are undifferentiated cells defined by the ability of a single cell both to self-renew, and to differentiate to produce progeny cells, including self-renewing progenitors, non-renewing progenitors, and terminally differentiated cells. Stem cells are also characterized by their ability to differentiate in vitro into functional cells of various cell lineages from multiple germ layers (endoderm, mesoderm and ectoderm), as well as to give rise to tissues of multiple germ layers following transplantation, and to contribute substantially to most, if not all, tissues following injection into blastocysts.
  • Stem cells are classified according to their developmental potential as: (1) totipotent; (2) pluripotent; (3) multipotent; (4) oligopotent; and (5) unipotent. Totipotent cells are able to give rise to all embryonic and extraembryonic cell types. Pluripotent cells are able to give rise to all embryonic cell types. Multipotent cells include those able to give rise to a subset of cell lineages, but all within a particular tissue, organ, or physiological system (for example, hematopoietic stem cells (HSC) can produce progeny that include HSC (self-renewal), blood cell-restricted oligopotent progenitors, and all cell types and elements (e.g., platelets) that are normal components of the blood). Cells that are oligopotent can give rise to a more restricted subset of cell lineages than multipotent stem cells; and cells that are unipotent are able to give rise to a single cell lineage (e.g., spermatogenic stem cells).
  • Stem cells are also categorized on the basis of the source from which they may be obtained. An adult stem cell is generally a multipotent undifferentiated cell found in tissue comprising multiple differentiated cell types. The adult stem cell can renew itself. Under normal circumstances, it can also differentiate to yield the specialized cell types of the tissue from which it originated, and possibly other tissue types. An embryonic stem cell is a pluripotent cell from the inner cell mass of a blastocyst-stage embryo. A fetal stem cell is one that originates from fetal tissues or membranes. A postpartum stem cell is a multipotent or pluripotent cell that originates substantially from extraembryonic tissue available after birth, namely, the placenta and the umbilical cord. These cells have been found to possess features characteristic of pluripotent stem cells, including rapid proliferation and the potential for differentiation into many cell lineages. Postpartum stem cells may be blood-derived (e.g., as are those obtained from umbilical cord blood) or non-blood-derived (e.g., as obtained from the non-blood tissues of the umbilical cord and placenta).
  • Embryonic tissue is typically defined as tissue originating from the embryo (which in humans refers to the period from fertilization to about six weeks of development. Fetal tissue refers to tissue originating from the fetus, which in humans refers to the period from about six weeks of development to parturition. Extraembryonic tissue is tissue associated with, but not originating from, the embryo or fetus. Extraembryonic tissues include extraembryonic membranes (chorion, amnion, yolk sac and allantois), umbilical cord and placenta (which itself forms from the chorion and the maternal decidua basalis).
  • Differentiation is the process by which an unspecialized (“uncommitted”) or less specialized cell acquires the features of a specialized cell, such as a nerve cell or a muscle cell, for example. A differentiated cell is one that has taken on a more specialized (“committed”) position within the lineage of a cell. The term committed, when applied to the process of differentiation, refers to a cell that has proceeded in the differentiation pathway to a point where, under normal circumstances, it will continue to differentiate into a specific cell type or subset of cell types, and cannot, under normal circumstances, differentiate into a different cell type or revert to a less differentiated cell type. De-differentiation refers to the process by which a cell reverts to a less specialized (or committed) position within the lineage of a cell. As used herein, the lineage of a cell defines the heredity of the cell, i.e. which cells it came from and what cells it can give rise to. The lineage of a cell places the cell within a hereditary scheme of development and differentiation.
  • In a broad sense, a progenitor cell is a cell that has the capacity to create progeny that are more differentiated than itself, and yet retains the capacity to replenish the pool of progenitors. By that definition, stem cells themselves are also progenitor cells, as are the more immediate precursors to terminally differentiated cells. When referring to the cells of the present invention, as described in greater detail below, this broad definition of progenitor cell may be used. In a narrower sense, a progenitor cell is often defined as a cell that is intermediate in the differentiation pathway, i.e., it arises from a stem cell and is intermediate in the production of a mature cell type or subset of cell types. This type of progenitor cell is generally not able to self-renew. Accordingly, if this type of cell is referred to herein, it will be referred to as a non-renewing progenitor cell or as an intermediate progenitor or precursor cell.
  • As used herein, the phrase differentiates into an ocular lineage or phenotype refers to a cell that becomes partially or fully committed to a specific ocular phenotype, including without limitation, retinal and corneal stem cells, pigment epithelial cells of the retina and iris, photoreceptors, retinal ganglia and other optic neural lineages (e.g., retinal glia, microglia, astrocytes, Mueller cells), cells forming the crystalline lens, and epithelial cells of the sclera, cornea, limbus and conjunctiva. The phrase differentiates into a neural lineage or phenotype refers to a cell that becomes partially or fully committed to a specific neural phenotype of the CNS or PNS, i.e., a neuron or a glial cell, the latter category including without limitation astrocytes, oligodendrocytes, Schwann cells and microglia.
  • The cells exemplified herein and preferred for use in present invention are generally referred to as postpartum-derived cells (or PPDCs). They also may sometimes be referred to more specifically as umbilicus-derived cells or placenta-derived cells (UDCs or PDCs). In addition, the cells may be described as being stem or progenitor cells, the latter term being used in the broad sense. The term derived is used to indicate that the cells have been obtained from their biological source and grown or otherwise manipulated in vitro (e.g., cultured in a Growth Medium to expand the population and/or to produce a cell line). The in vitro manipulations of umbilical stem cells and the unique features of the umbilicus-derived cells of the present invention are described in detail below. Cells isolated from postpartum placenta and umbilicus by other means are also considered suitable for use in the present invention. These other cells are referred to herein as postpartum cells (rather than postpartum-derived cells).
  • Various terms are used to describe cells in culture. Cell culture refers generally to cells taken from a living organism and grown under controlled condition (“in culture” or “cultured”). A primary cell culture is a culture of cells, tissues, or organs taken directly from an organism(s) before the first subculture. Cells are expanded in culture when they are placed in a Growth Medium under conditions that facilitate cell growth and/or division, resulting in a larger population of the cells. When cells are expanded in culture, the rate of cell proliferation is sometimes measured by the amount of time needed for the cells to double in number. This is referred to as doubling time.
  • A cell line is a population of cells formed by one or more subcultivations of a primary cell culture. Each round of subculturing is referred to as a passage. When cells are subcultured, they are referred to as having been passaged. A specific population of cells, or a cell line, is sometimes referred to or characterized by the number of times it has been passaged. For example, a cultured cell population that has been passaged ten times may be referred to as a P10 culture. The primary culture, i.e., the first culture following the isolation of cells from tissue, is designated P0. Following the first subculture, the cells are described as a secondary culture (P1 or passage 1). After the second subculture, the cells become a tertiary culture (P2 or passage 2), and so on. It will be understood by those of skill in the art that there may be many population doublings during the period of passaging; therefore the number of population doublings of a culture is greater than the passage number. The expansion of cells (i.e., the number of population doublings) during the period between passaging depends on many factors, including but not limited to the seeding density, substrate, medium, growth conditions, and time between passaging.
  • A conditioned medium is a medium in which a specific cell or population of cells has been cultured, and then removed. When cells are cultured in a medium, they may secrete cellular factors that can provide trophic support to other cells. Such trophic factors include, but are not limited to hormones, cytokines, extracellular matrix (ECM), proteins, vesicles, antibodies, and granules. The medium containing the cellular factors is the conditioned medium.
  • Generally, a trophic factor is defined as a substance that promotes survival, growth, differentiation, proliferation and/or maturation of a cell, or stimulates increased activity of a cell. The interaction between cells via trophic factors may occur between cells of different types. Cell interaction by way of trophic factors is found in essentially all cell types, and is a particularly significant means of communication among neural cell types. Trophic factors also can function in an autocrine fashion, i.e., a cell may produce trophic factors that affect its own survival, growth, differentiation, proliferation and/or maturation.
  • When referring to cultured vertebrate cells, the term senescence (also replicative senescence or cellular senescence) refers to a property attributable to finite cell cultures; namely, their inability to grow beyond a finite number of population doublings (sometimes referred to as Hayflick's limit). Although cellular senescence was first described using fibroblast-like cells, most normal human cell types that can be grown successfully in culture undergo cellular senescence. The in vitro lifespan of different cell types varies, but the maximum lifespan is typically fewer than 100 population doublings (this is the number of doublings for all the cells in the culture to become senescent and thus render the culture unable to divide). Senescence does not depend on chronological time, but rather is measured by the number of cell divisions, or population doublings, the culture has undergone. Thus, cells made quiescent by removing essential growth factors are able to resume growth and division when the growth factors are re-introduced, and thereafter carry out the same number of doublings as equivalent cells grown continuously. Similarly, when cells are frozen in liquid nitrogen after various numbers of population doublings and then thawed and cultured, they undergo substantially the same number of doublings as cells maintained unfrozen in culture. Senescent cells are not dead or dying cells; they are actually resistant to programmed cell death (apoptosis), and have been maintained in their nondividing state for as long as three years. These cells are very much alive and metabolically active, but they do not divide. The nondividing state of senescent cells has not yet been found to be reversible by any biological, chemical, or viral agent.
  • The terms ocular, ophthalmic and optic are used interchangeably herein to define “of, or about, or related to the eye.”
  • The term ocular degenerative condition (or disorder) is an inclusive term encompassing acute and chronic conditions, disorders or diseases of the eye, inclusive of the neural connection between the eye and the brain, involving cell damage, degeneration or loss. An ocular degenerative condition may be age-related, or it may result from injury or trauma, or it may be related to a specific disease or disorder. Acute ocular degenerative conditions include, but are not limited to, conditions associated with cell death or compromise affecting the eye including conditions arising from cerebrovascular insufficiency, focal or diffuse brain trauma, diffuse brain damage, infection or inflammatory conditions of the eye, retinal tearing or detachment, intra-ocular lesions (contusion penetration, compression, laceration) or other physical injury (e.g., physical or chemical burns). Chronic ocular degenerative conditions (including progressive conditions) include, but are not limited to, retinopathies and other retinal/macular disorders such as retinitis pigmentosa (RP), age-related macular degeneration (AMD), choroidal neovascular membrane (CNVM); retinopathies such as diabetic retinopathy, occlusive retinopathy, sickle cell retinopathy and hypertensive retinopathy, central retinal vein occlusion, stenosis of the carotid artery, optic neuropathies such as glaucoma and related syndromes; disorders of the lens and outer eye, e.g., limbal stem cell deficiency (LSCD), also referred to as limbal epithelial cell deficiency (LECD), such as occurs in chemical or thermal injury, Steven-Johnson syndrome, contact lens-induced keratopathy, ocular cicatricial pemphigoid, congenital diseases of aniridia or ectodermal dysplasia, and multiple endocrine deficiency-associated keratitis.
  • The term treating (or treatment of) an ocular degenerative condition refers to ameliorating the effects of, or delaying, halting or reversing the progress of, or delaying or preventing the onset of, an ocular degenerative condition as defined herein.
  • The term effective amount refers to a concentration or amount of a reagent or pharmaceutical composition, such as a growth factor, differentiation agent, trophic factor, cell population or other agent, that is effective for producing an intended result, including cell growth and/or differentiation in vitro or in vivo, or treatment of ocular degenerative conditions, as described herein. With respect to growth factors, an effective amount may range from about 1 nanogram/milliliter to about 1 microgram/milliliter. With respect to PPDCs as administered to a patient in vivo, an effective amount may range from as few as several hundred or fewer to as many as several million or more. In specific embodiments, an effective amount may range from 103-1011, more specifically at least about 104 cells. It will be appreciated that the number of cells to be administered will vary depending on the specifics of the disorder to be treated, including but not limited to size or total volume/surface area to be treated, as well as proximity of the site of administration to the location of the region to be treated, among other factors familiar to the medicinal biologist.
  • The terms effective period (or time) and effective conditions refer to a period of time or other controllable conditions (e.g., temperature, humidity for in vitro methods), necessary or preferred for an agent or pharmaceutical composition to achieve its intended result.
  • The term patient or subject refers to animals, including mammals, preferably humans, who are treated with the pharmaceutical compositions or in accordance with the methods described herein.
  • The term pharmaceutically acceptable carrier (or medium), which may be used interchangeably with the term biologically compatible carrier or medium, refers to reagents, cells, compounds, materials, compositions, and/or dosage forms that are not only compatible with the cells and other agents to be administered therapeutically, but also are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other complication commensurate with a reasonable benefit/risk ratio.
  • Several terms are used herein with respect to cell replacement therapy. The terms autologous transfer, autologous transplantation, autograft and the like refer to treatments wherein the cell donor is also the recipient of the cell replacement therapy. The terms allogeneic transfer, allogeneic transplantation, allograft and the like refer to treatments wherein the cell donor is of the same species as the recipient of the cell replacement therapy, but is not the same individual. A cell transfer in which the donor's cells and have been histocompatibly matched with a recipient is sometimes referred to as a syngeneic transfer. The terms xenogeneic transfer, xenogeneic transplantation, xenograft and the like refer to treatments wherein the cell donor is of a different species than the recipient of the cell replacement therapy.
  • DESCRIPTION
  • Ocular degenerative conditions, which encompass acute, chronic and progressive disorders and diseases having divergent causes, have as a common feature the dysfunction or loss of a specific or vulnerable group of ocular cells. This commonality enables development of similar therapeutic approaches for the repair or regeneration of vulnerable, damaged or lost ocular tissue, one of which is cell-based therapy. Development of cell therapy for ocular degenerative conditions heretofore has been limited to a comparatively few types of stem or progenitor cells, including ocular-derived stem cells themselves (e.g., retinal and corneal stem cells), embryonic stem cells and a few types of adult stem or progenitor cells (e.g., neural, mucosal epithelial and bone marrow stem cells). The present inventors have identified a significant new source of stem cells for this purpose, namely, cells isolated from the postpartum placenta and umbilical cord. Accordingly, in its various embodiments described herein, the present invention features methods and pharmaceutical compositions for repair and regeneration of ocular tissues, which utilize progenitor cells and cell populations isolated from postpartum tissues. The invention is applicable to any ocular degenerative condition, but is expected to be particularly suitable for a number of ocular disorders for which treatment or cure heretofore has been difficult or unavailable. These include, without limitation, age-related macular degeneration, retinitis pigmentosa, diabetic and other retinopathies, glaucoma and other optic neuropathies, and disorders associated with limbal stem cell deficiency.
  • Stem or progenitor cells isolated from postpartum placenta or umbilical cord in accordance with any method known in the art are expected to be suitable for use in the present invention. In a preferred embodiment, however, the invention utilizes postpartum-derived cells (PPDCs) as defined above, which are derived from placental or umbilical cord tissue that has been rendered substantially free of blood, preferably in accordance with the method set forth below. The PPDCs are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of other phenotypes. Certain embodiments features populations comprising such cells, pharmaceutical compositions comprising the cells or components or products thereof, and methods of using the pharmaceutical compositions for treatment of patients with acute or chronic ocular degenerative conditions. The postpartum-derived cells have been characterized by their growth properties in culture, by their cell surface markers, by their gene expression, by their ability to produce certain biochemical trophic factors, and by their immunological properties.
  • Preparation of PPDCs
  • According to the methods described herein, a mammalian placenta and umbilical cord are recovered upon or shortly after termination of either a full-term or pre-term pregnancy, for example, after expulsion after birth. The postpartum tissue may be transported from the birth site to a laboratory in a sterile container such as a flask, beaker, culture dish, or bag. The container may have a solution or medium, including but not limited to a salt solution, such as, for example, Dulbecco's Modified Eagle's Medium (DMEM) or phosphate buffered saline (PBS), or any solution used for transportation of organs used for transplantation, such as University of Wisconsin solution or perfluorochemical solution. One or more antibiotic and/or antimycotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin, may be added to the medium or buffer. The postpartum tissue may be rinsed with an anticoagulant solution such as heparin-containing solution. It is preferable to keep the tissue at about 4-10° C. prior to extraction of PPDCs. It is even more preferable that the tissue not be frozen prior to extraction of PPDCs.
  • Isolation of PPDCs preferably occurs in an aseptic environment. The umbilical cord may be separated from the placenta by means known in the art. Alternatively, the umbilical cord and placenta are used without separation. Blood and debris are preferably removed from the postpartum tissue prior to isolation of PPDCs. For example, the postpartum tissue may be washed with buffer solution, such as but not limited to phosphate buffered saline. The wash buffer also may comprise one or more antimycotic and/or antibiotic agents, such as but not limited to penicillin, streptomycin, amphotericin B, gentamicin, and nystatin.
  • Postpartum tissue comprising a whole placenta or a fragment or section thereof is disaggregated by mechanical force (mincing or shear forces). In a presently preferred embodiment, the isolation procedure also utilizes an enzymatic digestion process. Many enzymes are known in the art to be useful for the isolation of individual cells from complex tissue matrices to facilitate growth in culture. Ranging from weakly digestive (e.g. deoxyribonucleases and the neutral protease, dispase) to strongly digestive (e.g. papain and trypsin), such enzymes are available commercially. A nonexhaustive list of enzymes compatible herewith includes mucolytic enzyme activities, metalloproteases, neutral proteases, serine proteases (such as trypsin, chymotrypsin, or elastase), and deoxyribonucleases. Presently preferred are enzyme activities selected from metalloproteases, neutral proteases and mucolytic activities. For example, collagenases are known to be useful for isolating various cells from tissues. Deoxyribonucleases can digest single-stranded DNA and can minimize cell-clumping during isolation. Preferred methods involve enzymatic treatment with for example collagenase and dispase, or collagenase, dispase, and hyaluronidase, and such methods are provided wherein in certain preferred embodiments, a mixture of collagenase and the neutral protease dispase are used in the dissociating step. More preferred are those methods which employ digestion in the presence of at least one collagenase from Clostridium histolyticum, and either of the protease activities, dispase and thermolysin. Still more preferred are methods employing digestion with both collagenase and dispase enzyme activities. Also preferred are methods which include digestion with a hyaluronidase activity in addition to collagenase and dispase activities. The skilled artisan will appreciate that many such enzyme treatments are known in the art for isolating cells from various tissue sources. For example, the LIBERASE Blendzyme (Roche) series of enzyme combinations are suitable for use in the instant methods. Other sources of enzymes are known, and the skilled artisan may also obtain such enzymes directly from their natural sources. The skilled artisan is also well-equipped to assess new, or additional enzymes or enzyme combinations for their utility in isolating the cells of the invention. Preferred enzyme treatments are 0.5, 1, 1.5, or 2 hours long or longer. In other preferred embodiments, the tissue is incubated at 37° C. during the enzyme treatment of the dissociation step.
  • In some embodiments of the invention, postpartum tissue is separated into sections comprising various aspects of the tissue, such as neonatal, neonatal/maternal, and maternal aspects of the placenta, for instance. The separated sections then are dissociated by mechanical and/or enzymatic dissociation according to the methods described herein. Cells of neonatal or maternal lineage may be identified by any means known in the art, for example, by karyotype analysis or in situ hybridization for a Y chromosome.
  • Isolated cells or postpartum tissue from which PPDCs grow out may be used to initiate, or seed, cell cultures. Isolated cells are transferred to sterile tissue culture vessels either uncoated or coated with extracellular matrix or ligands such as laminin, collagen (native, denatured or crosslinked), gelatin, fibronectin, and other extracellular matrix proteins. PPDCs are cultured in any culture medium capable of sustaining growth of the cells such as, but not limited to, DMEM (high or low glucose), advanced DMEM, DMEM/MCDB 201, Eagle's basal medium, Ham's F10 medium (F10), Ham's F-12 medium (F12), Iscove's modified Dulbecco's medium, Mesenchymal Stem Cell Growth Medium (MSCGM), DMEM/F12, RPMI 1640, and CELL-GRO-FREE. The culture medium may be supplemented with one or more components including, for example, fetal bovine serum (FBS), preferably about 2-15% (v/v); equine serum (ES); human serum (HS); beta-mercaptoethanol (BME or 2-ME), preferably about 0.001% (v/v); one or more growth factors, for example, platelet-derived growth factor (PDGF), epidermal growth factor (EGF), fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), insulin-like growth factor-1 (IGF-1), leukocyte inhibitory factor (LIF) and erythropoietin; amino acids, including L-valine; and one or more antibiotic and/or antimycotic agents to control microbial contamination, such as, for example, penicillin G, streptomycin sulfate, amphotericin B, gentamicin, and nystatin, either alone or in combination. The culture medium preferably comprises Growth Medium (DMEM-low glucose, serum, BME, and an antibiotic agent).
  • The cells are seeded in culture vessels at a density to allow cell growth. In a preferred embodiment, the cells are cultured at about 0 to about 5 percent by volume CO2 in air. In some preferred embodiments, the cells are cultured at about 2 to about 25 percent O2 in air, preferably about 5 to about 20 percent O2 in air. The cells preferably are cultured at about 25 to about 40° C. and more preferably are cultured at 37° C. The cells are preferably cultured in an incubator. The medium in the culture vessel can be static or agitated, for example, using a bioreactor. PPDCs preferably are grown under low oxidative stress (e.g., with addition of glutathione, Vitamin C, Catalase, Vitamin E, N-Acetylcysteine). “Low oxidative stress”, as used herein, refers to conditions of no or minimal free radical damage to the cultured cells.
  • Methods for the selection of the most appropriate culture medium, medium preparation, and cell culture techniques are well known in the art and are described in a variety of sources, including Doyle et al., (eds.), 1995, CELL & TISSUE CULTURE: LABORATORY PROCEDURES, John Wiley & Sons, Chichester; and Ho and Wang (eds.), 1991, ANIMAL CELL BIOREACTORS, Butterworth-Heinemann, Boston, which are incorporated herein by reference.
  • After culturing the isolated cells or tissue fragments for a sufficient period of time, PPDCs will have grown out, either as a result of migration from the postpartum tissue or cell division, or both. In some embodiments of the invention, PPDCs are passaged, or removed to a separate culture vessel containing fresh medium of the same or a different type as that used initially, where the population of cells can be mitotically expanded. The cells of the invention may be used at any point between passage 0 and senescence. The cells preferably are passaged between about 3 and about 25 times, more preferably are passaged about 4 to about 12 times, and preferably are passaged 10 or 11 times. Cloning and/or subcloning may be performed to confirm that a clonal population of cells has been isolated.
  • In some aspects of the invention, the different cell types present in postpartum tissue are fractionated into subpopulations from which the PPDCs can be isolated. This may be accomplished using standard techniques for cell separation including, but not limited to, enzymatic treatment to dissociate postpartum tissue into its component cells, followed by cloning and selection of specific cell types, for example but not limited to selection based on morphological and/or biochemical markers; selective growth of desired cells (positive selection), selective destruction of unwanted cells (negative selection); separation based upon differential cell agglutinability in the mixed population as, for example, with soybean agglutinin; freeze-thaw procedures; differential adherence properties of the cells in the mixed population; filtration; conventional and zonal centrifugation; centrifugal elutriation (counter-streaming centrifugation); unit gravity separation; countercurrent distribution; electrophoresis; and fluorescence activated cell sorting (FACS). For a review of clonal selection and cell separation techniques, see Freshney, 1994, CULTURE OF ANIMAL CELLS: A MANUAL OF BASIC TECHNIQUES, 3rd Ed., Wiley-Liss, Inc., New York, which is incorporated herein by reference.
  • The culture medium is changed as necessary, for example, by carefully aspirating the medium from the dish, for example, with a pipette, and replenishing with fresh medium. Incubation is continued until a sufficient number or density of cells accumulate in the dish. The original explanted tissue sections may be removed and the remaining cells trypsinized using standard techniques or using a cell scraper. After trypsinization, the cells are collected, removed to fresh medium and incubated as above. In some embodiments, the medium is changed at least once at approximately 24 hours post-trypsinization to remove any floating cells. The cells remaining in culture are considered to be PPDCs.
  • PPDCs may be cryopreserved. Accordingly, in a preferred embodiment described in greater detail below, PPDCs for autologous transfer (for either the mother or child) may be derived from appropriate postpartum tissues following the birth of a child, then cryopreserved so as to be available in the event they are later needed for transplantation.
  • Characteristics of PPDCs
  • PPDCs may be characterized, for example, by growth characteristics (e.g., population doubling capability, doubling time, passages to senescence), karyotype analysis (e.g., normal karyotype; maternal or neonatal lineage), flow cytometry (e.g., FACS analysis), immunohistochemistry and/or immunocytochemistry (e.g., for detection of epitopes), gene expression profiling (e.g., gene chip arrays; polymerase chain reaction (for example, reverse transcriptase PCR, real time PCR, and conventional PCR)), protein arrays, protein secretion (e.g., by plasma clotting assay or analysis of PDC-conditioned medium, for example, by Enzyme Linked ImmunoSorbent Assay (ELISA)), mixed lymphocyte reaction (e.g., as measure of stimulation of PBMCs), and/or other methods known in the art.
  • Examples of PPDCs derived from placental tissue were deposited with the American Type Culture Collection (ATCC, Manassas, Va.) and assigned ATCC Accession Numbers as follows: (1) strain designation PLA 071003 (P8) was deposited Jun. 15, 2004 and assigned Accession No. PTA-6074; (2) strain designation PLA 071003 (P111) was deposited Jun. 15, 2004 and assigned Accession No. PTA-6075; and (3) strain designation PLA 071003 (P16) was deposited Jun. 16, 2004 and assigned Accession No. PTA-6079. Examples of PPDCs derived from umbilicus tissue were deposited with the American Type Culture Collection on Jun. 10, 2004, and assigned ATCC Accession Numbers as follows: (1) strain designation UMB 022803 (P7) was assigned Accession No. PTA-6067; and (2) strain designation UMB 022803 (P17) was assigned Accession No. PTA-6068.
  • In various embodiments, the PPDCs possess one or more of the following growth features (1) they require L-valine for growth in culture; (2) they are capable of growth in atmospheres containing oxygen from about 5% to at least about 20% (3) they have the potential for at least about 40 doublings in culture before reaching senescence; and (4) they attach and expand on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin or fibronectin.
  • In certain embodiments the PPDCs possess a normal karyotype, which is maintained as the cells are passaged. Karyotyping is particularly useful for identifying and distinguishing neonatal from maternal cells derived from placenta. Methods for karyotyping are available and known to those of skill in the art.
  • In other embodiments, the PPDCs may be characterized by production of certain proteins, including (1) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; and (2) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C cell surface markers, as detected by flow cytometry. In other embodiments, the PPDCs may be characterized by lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ cell surface markers, as detected by flow cytometry. Particularly preferred are cells that produce at least two of tissue factor, vimentin, and alpha-smooth muscle actin. More preferred are those cells producing all three of the proteins tissue factor, vimentin, and alpha-smooth muscle actin.
  • In other embodiments, the PPDCs may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for a gene encoding at least one of interleukin 8; reticulon 1; chemokine (C—X—C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C—X—C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C—X—C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547k1113.
  • In yet other embodiments, the PPDCs may be characterized by gene expression, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for a gene encoding at least one of: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; and cytochrome c oxidase subunit VIIa polypeptide 1 (muscle).
  • In other embodiments, the PPDCs may be characterized by secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1. In alternative embodiments, the PPDCs may be characterized by lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
  • In preferred embodiments, the cell comprises two or more of the above-listed growth, protein/surface marker production, gene expression or substance-secretion characteristics. More preferred are those cells comprising, three, four, or five or more of the characteristics. Still more preferred are PPDCs comprising six, seven, or eight or more of the characteristics. Still more preferred presently are those cells comprising all of above characteristics.
  • Among cells that are presently preferred for use with the invention in several of its aspects are postpartum cells having the characteristics described above and more particularly those wherein the cells have normal karyotypes and maintain normal karyotypes with passaging, and further wherein the cells express each of the markers CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C, wherein the cells produce the immunologically-detectable proteins which correspond to the listed markers. Still more preferred are those cells which in addition to the foregoing do not produce proteins corresponding to any of the markers CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
  • Certain cells having the potential to differentiate along lines leading to various phenotypes are unstable and thus can spontaneously differentiate. Presently preferred for use with the invention are cells that do not spontaneously differentiate, for example along neural lines. Preferred cells, when grown in Growth Medium, are substantially stable with respect to the cell markers produced on their surface, and with respect to the expression pattern of various genes, for example as determined using an Affymetrix GENECHIP. The cells remain substantially constant, for example in their surface marker characterisitics over passaging, through multiple population doublings.
  • However, one feature of PPDCs is that they may be deliberately induced to differentiate into various lineage phenotypes by subjecting them to differentiation-inducing cell culture conditions. Of use in treatment of certain ocular degenerative conditions, the PPDCs may be induced to differentiate into neural phenotypes using one or more methods known in the art. For instance, as exemplified herein, PPDCs may be plated on flasks coated with laminin in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine and Penicillin/Streptomycin, the combination of which is referred to herein as Neural Progenitor Expansion (NPE) medium. NPE media may be further supplemented with bFGF and/or EGF. Alternatively, PPDCs may be induced to differentiate in vitro by (1) co-culturing the PPDCs with neural progenitor cells, or (2) growing the PPDCs in neural progenitor cell-conditioned medium.
  • Differentiation of the PPDCs into neural phenotypes may be demonstrated by a bipolar cell morphology with extended processes. The induced cell populations may stain positive for the presence of nestin. Differentiated PPDCs may be assessed by detection of nestin, TuJ1 (BIII tubulin), GFAP, tyrosine hydroxylase, GABA, 04 and/or MBP. In some embodiments, PPDCs have exhibited the ability to form three-dimensional bodies characteristic of neuronal stem cell formation of neurospheres.
  • Cell Populations, Modifications, Components and Products
  • Another aspect of the invention features populations of cells isolated from placental or umbilical tissue. In a preferred embodiment, the populations comprise the PPDCs described above, and these cell populations are described in the section below. In some embodiments, the cell population is heterogeneous. A heterogeneous cell population of the invention may comprise at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, or 95% PPDCs. The heterogeneous cell populations of the invention may further comprise stem cells or other progenitor cells, such as epithelial or neural progenitor cells, or it may further comprise fully differentiated cells. In some embodiments, the population is substantially homogeneous, i.e., comprises substantially only PPDCs (preferably at least about 96%, 97%, 98%, 99% or more PPDCs). The homogeneous cell population of the invention may comprise umbilicus- or placenta-derived cells. Homogeneous populations of umbilicus-derived cells are preferably free of cells of maternal lineage. Homogeneous populations of placenta-derived cells may be of neonatal or maternal lineage. Homogeneity of a cell population may be achieved by any method known in the art, for example, by cell sorting (e.g., flow cytometry) or by clonal expansion in accordance with known methods. Thus, preferred homogeneous PPDC populations may comprise a clonal cell line of postpartum-derived cells. Such populations are particularly useful when a cell clone with highly desirable functionality has been isolated.
  • Also provided herein are populations of cells incubated in the presence of one or more factors, or under conditions, that stimulate stem cell differentiation along a desired pathway (e.g., neural, epithelial). Such factors are known in the art and the skilled artisan will appreciate that determination of suitable conditions for differentiation can be accomplished with routine experimentation. Optimization of such conditions can be accomplished by statistical experimental design and analysis, for example response surface methodology allows simultaneous optimization of multiple variables, for example in a biological culture. Presently preferred factors include, but are not limited to factors, such as growth or trophic factors, demethylating agents, co-culture with neural or epithelial lineage cells or culture in neural or epithelial lineage cell-conditioned medium, as well other conditions known in the art to stimulate stem cell differentiation along these pathways (for factors useful in neural differentiation, see, e.g., Lang, K. J. D. et al., 2004, J. Neurosci. Res. 76: 184-192; Johe, K. K. et al., 1996, Genes Devel. 10: 3129-3140; Gottleib, D., 2002, Ann. Rev. Neurosci. 25: 381-407).
  • Postpartum cells, preferably PPDCs, may also be genetically modified to produce therapeutically useful gene products, or to produce antineoplastic agents for treatment of tumors, for example. Genetic modification may be accomplished using any of a variety of vectors including, but not limited to, integrating viral vectors, e.g., retrovirus vector or adeno-associated viral vectors; non-integrating replicating vectors, e.g., papilloma virus vectors, SV40 vectors, adenoviral vectors; or replication-defective viral vectors. Other methods of introducing DNA into cells include the use of liposomes, electroporation, a particle gun, or by direct DNA injection.
  • Hosts cells are preferably transformed or transfected with DNA controlled by or in operative association with, one or more appropriate expression control elements such as promoter or enhancer sequences, transcription terminators, polyadenylation sites, among others, and a selectable marker. Any promoter may be used to drive the expression of the inserted gene. For example, viral promoters include, but are not limited to, the CMV promoter/enhancer, SV 40, papillomavirus, Epstein-Barr virus or elastin gene promoter. In some embodiments, the control elements used to control expression of the gene of interest can allow for the regulated expression of the gene so that the product is synthesized only when needed in vivo. If transient expression is desired, constitutive promoters are preferably used in a non-integrating and/or replication-defective vector. Alternatively, inducible promoters could be used to drive the expression of the inserted gene when necessary. Inducible promoters include, but are not limited to, those associated with metallothionein and heat shock proteins.
  • Following the introduction of the foreign DNA, engineered cells may be allowed to grow in enriched media and then switched to selective media. The selectable marker in the foreign DNA confers resistance to the selection and allows cells to stably integrate the foreign DNA as, for example, on a plasmid, into their chromosomes and grow to form foci which, in turn, can be cloned and expanded into cell lines. This method can be advantageously used to engineer cell lines that express the gene product.
  • Cells may be genetically engineered to “knock out” or “knock down” expression of factors that promote inflammation or rejection at the implant site. Negative modulatory techniques for the reduction of target gene expression levels or target gene product activity levels are discussed below. “Negative modulation,” as used herein, refers to a reduction in the level and/or activity of target gene product relative to the level and/or activity of the target gene product in the absence of the modulatory treatment. The expression of a gene native to a neuron or glial cell can be reduced or knocked out using a number of techniques including, for example, inhibition of expression by inactivating the gene using the homologous recombination technique. Typically, an exon encoding an important region of the protein (or an exon 5′ to that region) is interrupted by a positive selectable marker, e.g., neo, preventing the production of normal mRNA from the target gene and resulting in inactivation of the gene. A gene may also be inactivated by creating a deletion in part of a gene, or by deleting the entire gene. By using a construct with two regions of homology to the target gene that are far apart in the genome, the sequences intervening the two regions can be deleted (Mombaerts et al., 1991, Proc. Nat. Acad. Sci. U.S.A. 88:3084). Antisense, DNAzymes, ribozymes, small interfering RNA (siRNA) and other such molecules that inhibit expression of the target gene can also be used to reduce the level of target gene activity. For example, antisense RNA molecules that inhibit the expression of major histocompatibility gene complexes (HLA) have been shown to be most versatile with respect to immune responses. Still further, triple helix molecules can be utilized in reducing the level of target gene activity. These techniques are described in detail by L. G. Davis et al. (eds), 1994, BASIC METHODS IN MOLECULAR BIOLOGY, 2nd ed., Appleton & Lange, Norwalk, CN.
  • In other aspects, the invention provides cell lysates and cell soluble fractions prepared from postpartum stem cells, preferably PPDCs, or heterogeneous or homogeneous cell populations comprising PPDCs, as well as PPDCs or populations thereof that have been genetically modified or that have been stimulated to differentiate along a neurogenic pathway. Such lysates and fractions thereof have many utilities. Use of the cell lysate soluble fraction (i.e., substantially free of membranes) in vivo, for example, allows the beneficial intracellular milieu to be used allogeneically in a patient without introducing an appreciable amount of the cell surface proteins most likely to trigger rejection, or other adverse immunological responses. Methods of lysing cells are well-known in the art and include various means of mechanical disruption, enzymatic disruption, or chemical disruption, or combinations thereof. Such cell lysates may be prepared from cells directly in their Growth Medium and thus containing secreted growth factors and the like, or may be prepared from cells washed free of medium in, for example, PBS or other solution. Washed cells may be resuspended at concentrations greater than the original population density if preferred.
  • In one embodiment, whole cell lysates are prepared, e.g., by disrupting cells without subsequent separation of cell fractions. In another embodiment, a cell membrane fraction is separated from a soluble fraction of the cells by routine methods known in the art, e.g., centrifugation, filtration, or similar methods.
  • Cell lysates or cell soluble fractions prepared from populations of postpartum-derived cells may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, partially purified, combined with pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions. Cell lysates or fractions thereof may be used in vitro or in vivo, alone or for example, with autologous or syngeneic live cells. The lysates, if introduced in vivo, may be introduced locally at a site of treatment, or remotely to provide, for example needed cellular growth factors to a patient.
  • In a further embodiment, postpartum cells, preferably PPDCs, can be cultured in vitro to produce biological products in high yield. For example, such cells, which either naturally produce a particular biological product of interest (e.g., a trophic factor), or have been genetically engineered to produce a biological product, can be clonally expanded using the culture techniques described herein. Alternatively, cells may be expanded in a medium that induces differentiation to a desired lineage. In either case, biological products produced by the cell and secreted into the medium can be readily isolated from the conditioned medium using standard separation techniques, e.g., such as differential protein precipitation, ion-exchange chromatography, gel filtration chromatography, electrophoresis, and HPLC, to name a few. A “bioreactor” may be used to take advantage of the flow method for feeding, for example, a three-dimensional culture in vitro. Essentially, as fresh media is passed through the three-dimensional culture, the biological product is washed out of the culture and may then be isolated from the outflow, as above.
  • Alternatively, a biological product of interest may remain within the cell and, thus, its collection may require that the cells be lysed, as described above. The biological product may then be purified using any one or more of the above-listed techniques.
  • In other embodiments, the invention provides conditioned medium from cultured postpartum cells, preferably PPDCs, for use in vitro and in vivo as described below. Use of such conditioned medium allows the beneficial trophic factors secreted by the postpartum cells to be used allogeneically in a patient without introducing intact cells that could trigger rejection, or other adverse immunological responses. Conditioned medium is prepared by culturing cells in a culture medium, then removing the cells from the medium.
  • Conditioned medium prepared from populations of postpartum-derived cells may be used as is, further concentrated, by for example, ultrafiltration or lyophilization, or even dried, partially purified, combined with pharmaceutically-acceptable carriers or diluents as are known in the art, or combined with other compounds such as biologicals, for example pharmaceutically useful protein compositions. Conditioned medium may be used in vitro or in vivo, alone or for example, with autologous or syngeneic live cells. The conditioned medium, if introduced in vivo, may be introduced locally at a site of treatment, or remotely to provide, for example needed cellular growth or trophic factors to a patient.
  • In another embodiment, an extracellular matrix (ECM) produced by culturing postpartum cells (preferably PPDCs) on liquid, solid or semi-solid substrates is prepared, collected and utilized as an alternative to implanting live cells into a subject in need of tissue repair or replacement. The cells are cultured in vitro, on a three dimensional framework as described elsewhere herein, under conditions such that a desired amount of ECM is secreted onto the framework. The comprising the new tissue are removed, and the ECM processed for further use, for example, as an injectable preparation. To accomplish this, cells on the framework are killed and any cellular debris removed from the framework. This process may be carried out in a number of different ways. For example, the living tissue can be flash-frozen in liquid nitrogen without a cryopreservative, or the tissue can be immersed in sterile distilled water so that the cells burst in response to osmotic pressure.
  • Once the cells have been killed, the cellular membranes may be disrupted and cellular debris removed by treatment with a mild detergent rinse, such as EDTA, CHAPS or a zwitterionic detergent. Alternatively, the tissue can be enzymatically digested and/or extracted with reagents that break down cellular membranes and allow removal of cell contents. Example of such enzymes include, but are not limited to, hyaluronidase, dispase, proteases, and nucleases. Examples of detergents include non-ionic detergents such as, for example, alkylaryl polyether alcohol (TRITON X-100), octylphenoxy polyethoxy-ethanol (Rohm and Haas Philadelphia, Pa.), BRIJ-35, a polyethoxyethanol lauryl ether (Atlas Chemical Co., San Diego, Calif.), polysorbate 20 (TWEEN 20), a polyethoxyethanol sorbitan monolaureate (Rohm and Haas), polyethylene lauryl ether (Rohm and Haas); and ionic detergents such as, for example, sodium dodecyl sulphate, sulfated higher aliphatic alcohols, sulfonated alkanes and sulfonated alkylarenes containing 7 to 22 carbon atoms in a branched or unbranched chain.
  • The collection of the ECM can be accomplished in a variety of ways, depending, for example, on whether the new tissue has been formed on a three-dimensional framework that is biodegradable or non-biodegradable. For example, if the framework is non-biodegradable, the ECM can be removed by subjecting the framework to sonication, high pressure water jets, mechanical scraping, or mild treatment with detergents or enzymes, or any combination of the above.
  • If the framework is biodegradable, the ECM can be collected, for example, by allowing the framework to degrade or dissolve in solution. Alternatively, if the biodegradable framework is composed of a material that can itself be injected along with the ECM, the framework and the ECM can be processed in toto for subsequent injection. Alternatively, the ECM can be removed from the biodegradable framework by any of the methods described above for collection of ECM from a non-biodegradable framework. All collection processes are preferably designed so as not to denature the ECM.
  • After it has been collected, the ECM may be processed further. For example, the ECM can be homogenized to fine particles using techniques well known in the art such as by sonication, so that it can pass through a surgical needle. The components of the ECM can be crosslinked, if desired, by gamma irradiation. Preferably, the ECM can be irradiated between 0.25 to 2 mega rads to sterilize and crosslink the ECM. Chemical crosslinking using agents that are toxic, such as glutaraldehyde, is possible but not generally preferred.
  • The amounts and/or ratios of proteins, such as the various types of collagen present in the ECM, may be adjusted by mixing the ECM produced by the cells of the invention with ECM of one or more other cell types. In addition, biologically active substances such as proteins, growth factors and/or drugs, can be incorporated into the ECM. Exemplary biologically active substances include tissue growth factors, such as TGF-beta, and the like, which promote healing and tissue repair at the site of the injection. Such additional agents may be utilized in any of the embodiments described herein above, e.g., with whole cell lysates, soluble cell fractions, or further purified components and products produced by the cells.
  • Pharmaceutical Compositions
  • In another aspect, the invention provides pharmaceutical compositions that utilize postpartum cells, preferably PPDCs, cell populations and cell components and products in various methods for treatment of ocular degenerative conditions. Certain embodiments encompass pharmaceutical compositions comprising live cells (e.g., PPDCs alone or admixed with other cell types). Other embodiments encompass pharmaceutical compositions comprising PPDC cellular components (e.g., cell lysates, soluble cell fractions, conditioned medium, ECM, or components of any of the foregoing) or products (e.g., trophic and other biological factors produced naturally by PPDCs or through genetic modification, conditioned medium from PPDC culture). In either case, the pharmaceutical composition may further comprise other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative, neuroprotective or ophthalmic drugs as known in the art.
  • Examples of other components that may be added to PPDC pharmaceutical compositions include, but are not limited to: (1) other neuroprotective or neurobeneficial drugs; (2) selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs (alternatively, PPDCs may be genetically engineered to express and produce growth factors); (3) anti-apoptotic agents (e.g., erythropoietin (EPO), EPO mimetibody, thrombopoietin, insulin-like growth factor (IGF)-I, IGF-II, hepatocyte growth factor, caspase inhibitors); (4) anti-inflammatory compounds (e.g., p38 MAP kinase inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and non-steroidal anti-inflammatory drugs (NSAIDS) (such as TEPOXALIN, TOLMETIN, and SUPROFEN); (5) immunosuppressive or immunomodulatory agents, such as calcineurin inhibitors, mTOR inhibitors, antiproliferatives, corticosteroids and various antibodies; (6) antioxidants such as probucol, vitamins C and E, conenzyme Q-10, glutathione, L-cysteine and N-acetylcysteine; and (6) local anesthetics, to name a few.
  • Pharmaceutical compositions of the invention comprise postpartum cells (preferably PPDCs), or components or products thereof, formulated with a pharmaceutically acceptable carrier or medium. Suitable pharmaceutically acceptable carriers include water, salt solution (such as Ringer's solution), alcohols, oils, gelatins, and carbohydrates, such as lactose, amylose, or starch, fatty acid esters, hydroxymethylcellulose, and polyvinyl pyrolidine. Such preparations can be sterilized, and if desired, mixed with auxiliary agents such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, and coloring. Typically, but not exclusively, pharmaceutical compositions comprising cellular components or products, but not live cells, are formulated as liquids. Pharmaceutical compositions comprising PPDC live cells are typically formulated as liquids, semisolids (e.g., gels) or solids (e.g., matrices, scaffolds and the like, as appropriate for ophthalmic tissue engineering).
  • Pharmaceutical compositions may comprise auxiliary components as would be familiar to medicinal chemists or biologists. For example, they may contain antioxidants in ranges that vary depending on the kind of antioxidant used. Reasonable ranges for commonly used antioxidants are about 0.01% to about 0.15% weight by volume of EDTA, about 0.01% to about 2.0% weight volume of sodium sulfite, and about 0.01% to about 2.0% weight by volume of sodium metabisulfite. One skilled in the art may use a concentration of about 0.1% weight by volume for each of the above. Other representative compounds include mercaptopropionyl glycine, N-acetyl cysteine, beta-mercaptoethylamine, glutathione and similar species, although other anti-oxidant agents suitable for ocular administration, e.g. ascorbic acid and its salts or sulfite or sodium metabisulfite may also be employed.
  • A buffering agent may be used to maintain the pH of eye drop formulations in the range of about 4.0 to about 8.0; so as to minimize irritation of the eye. For direct intravitreal or intraocular injection, formulations should be at pH 7.2 to 7.5, preferably at pH 7.3-7.4. The ophthalmologic compositions may also include tonicity agents suitable for administration to the eye. Among those suitable is sodium chloride to make formulations approximately isotonic with 0.9% saline solution.
  • In certain embodiments, pharmaceutical compositions are formulated with viscosity enhancing agents. Exemplary agents are hydroxyethylcellulose, hydroxypropylcellulose, methylcellulose, and polyvinylpyrrolidone. The pharmaceutical compositions may have cosolvents added if needed. Suitable cosolvents may include glycerin, polyethylene glycol (PEG), polysorbate, propylene glycol, and polyvinyl alcohol. Preservatives may also be included, e.g., benzalkonium chloride, benzethonium chloride, chlorobutanol, phenylmercuric acetate or nitrate, thimerosal, or methyl or propylparabens.
  • Formulations for injection are preferably designed for single-use administration and do not contain preservatives. Injectable solutions should have isotonicity equivalent to 0.9% sodium chloride solution (osmolality of 290-300 milliosmoles). This may be attained by addition of sodium chloride or other co-solvents as listed above, or excipients such as buffering agents and antioxidants, as listed above.
  • The tissues of the anterior chamber of the eye are bathed by the aqueous humor, while the retina is under continuous exposure to the vitreous. These fluids/gels exist in a highly reducing redox state because they contains antioxidant compounds and enzymes. Therefore, it may be advantageous to include a reducing agent in the ophthalmologic compositions. Suitable reducing agents include N-acetylcysteine, ascorbic acid or a salt form, and sodium sulfite or metabisulfite, with ascorbic acid and/or N-acetylcysteine or glutathione being particularly suitable for injectable solutions.
  • Pharmaceutical compositions comprising cells, cell components or cell products may be delivered to the eye of a patient in one or more of several delivery modes known in the art. In one embodiment that may be suitable for use in some instances, the compositions are topically delivered to the eye in eye drops or washes. In another embodiment, the compositions may be delivered to various locations within the eye via periodic intraocular injection or by infusion in an irrigating solution such as BSS or BSS PLUS (Alcon USA, Fort Worth, Tex.). Alternatively, the compositions may be applied in other ophthalmologic dosage forms known to those skilled in the art, such as pre-formed or in situ-formed gels or liposomes, for example as disclosed in U.S. Pat. No. 5,718,922 to Herrero-Vanrell. In another embodiment, the composition may be delivered to or through the lens of an eye in need of treatment via a contact lens (e.g. Lidofilcon B, Bausch & Lomb CW79 or DELTACON (Deltafilcon A) or other object temporarily resident upon the surface of the eye. In other embodiments, supports such as a collagen corneal shield (e.g. BIO-COR dissolvable corneal shields, Summit Technology, Watertown, Mass.) can be employed. The compositions can also be administered by infusion into the eyeball, either through a cannula from an osmotic pump (ALZET, Alza Corp., Palo Alto, Calif.) or by implantation of timed-release capsules (OCCUSENT) or biodegradable disks (OCULEX, OCUSERT). These routes of administration have the advantage of providing a continuous supply of the pharmaceutical composition to the eye. This may be an advantage for local delivery to the cornea, for example.
  • Pharmaceutical compositions comprising live cells in a semi-solid or solid carrier are typically formulated for surgical implantation at the site of ocular damage or distress. It will be appreciated that liquid compositions also may be administered by surgical procedures. In particular embodiments, semi-solid or solid pharmaceutical compositions may comprise semi-permeable gels, lattices, cellular scaffolds and the like, which may be non-biodegradable or biodegradable. For example, in certain embodiments, it may be desirable or appropriate to sequester the exogenous cells from their surroundings, yet enable the cells to secrete and deliver biological molecules to surrounding cells. In these embodiments, cells may be formulated as autonomous implants comprising living PPDCs or cell population comprising PPDCs surrounded by a non-degradable, selectively permeable barrier that physically separates the transplanted cells from host tissue. Such implants are sometimes referred to as “immunoprotective,” as they have the capacity to prevent immune cells and macromolecules from killing the transplanted cells in the absence of pharmacologically induced immunosuppression (for a review of such devices and methods, see, e.g., P. A. Tresco et al., 2000, Adv. Drug Delivery Rev. 42: 3-27).
  • In other embodiments, different varieties of degradable gels and networks are utilized for the pharmaceutical compositions of the invention. For example, degradable materials particularly suitable for sustained release formulations include biocompatible polymers, such as poly(lactic acid), poly (lactic-co-glycolic acid), methylcellulose, hyaluronic acid, collagen, and the like. The structure, selection and use of degradable polymers in drug delivery vehicles have been reviewed in several publications, including, A. Domb et al., 1992, Polymers for Advanced Technologies 3:279. U.S. Pat. No. 5,869,079 to Wong et al. discloses combinations of hydrophilic and hydrophobic entities in a biodegradable sustained release ocular implant. In addition, U.S. Pat. No. 6,375,972 to Guo et al., U.S. Pat. No. 5,902,598 to Chen et al., U.S. Pat. No. 6,331,313 to Wong et al., U.S. Pat. No. 5,707,643 to Ogura et al., U.S. Pat. No. 5,466,233 to Weiner et al. and U.S. Pat. No. 6,251,090 to Avery et al. each describes intraocular implant devices and systems that may be used to deliver pharmaceutical compositions.
  • In other embodiments, e.g., for repair of neural lesions, such as a damaged or severed optic nerve, it may be desirable or appropriate to deliver the cells on or in a biodegradable, preferably bioresorbable or bioabsorbable, scaffold or matrix. These typically three-dimensional biomaterials contain the living cells attached to the scaffold, dispersed within the scaffold, or incorporated in an extracellular matrix entrapped in the scaffold. Once implanted into the target region of the body, these implants become integrated with the host tissue, wherein the transplanted cells gradually become established (see, e.g., P. A. Tresco et al., 2000, supra; see also D. W. Hutmacher, 2001, J. Biomater. Sci. Polymer Edn. 12: 107-174).
  • Examples of scaffold or matrix (sometimes referred to collectively as “framework”) material that may be used in the present invention include nonwoven mats, porous foams, or self assembling peptides. Nonwoven mats may, for example, be formed using fibers comprised of a synthetic absorbable copolymer of glycolic and lactic acids (PGA/PLA), sold under the tradename VICRYL (Ethicon, Inc., Somerville, NJ), Foams, composed of, for example, poly(epsilon-caprolactone)/poly(glycolic acid) (PCL/PGA) copolymer, formed by processes such as freeze-drying, or lyophilized, as discussed in U.S. Pat. No. 6,355,699, also may be utilized. Hydrogels such as self-assembling peptides (e.g., RAD16) may also be used. In situ-forming degradable networks are also suitable for use in the invention (see, e.g., Anseth, K. S. et al., 2002, J. Controlled Release 78: 199-209; Wang, D. et al., 2003, Biomaterials 24: 3969-3980; U.S. Patent Publication 2002/0022676 to He et al.). These materials are formulated as fluids suitable for injection, then may be induced by a variety of means (e.g., change in temperature, pH, exposure to light) to form degradable hydrogel networks in situ or in vivo.
  • In another embodiment, the framework is a felt, which can be composed of a multifilament yarn made from a bioabsorbable material, e.g., PGA, PLA, PCL copolymers or blends, or hyaluronic acid. The yarn is made into a felt using standard textile processing techniques consisting of crimping, cutting, carding and needling. In another embodiment, cells are seeded onto foam scaffolds that may be composite structures.
  • In many of the abovementioned embodiments, the framework may be molded into a useful shape. Furthermore, it will be appreciated that PPDCs may be cultured on pre-formed, non-degradable surgical or implantable devices, e.g., in a manner corresponding to that used for preparing fibroblast-containing GDC endovascular coils, for instance (Marx, W. F. et al., 2001, Am. J. Neuroradiol. 22: 323-333).
  • The matrix, scaffold or device may be treated prior to inoculation of cells in order to enhance cell attachment. For example, prior to inoculation, nylon matrices can be treated with 0.1 molar acetic acid and incubated in polylysine, PBS, and/or collagen to coat the nylon. Polystyrene can be similarly treated using sulfuric acid. The external surfaces of a framework may also be modified to improve the attachment or growth of cells and differentiation of tissue, such as by plasma coating the framework or addition of one or more proteins (e.g., collagens, elastic fibers, reticular fibers), glycoproteins, glycosaminoglycans (e.g., heparin sulfate, chondroitin-4-sulfate, chondroitin-6-sulfate, dermatan sulfate, keratin sulfate), a cellular matrix, and/or other materials such as, but not limited to, gelatin, alginates, agar, agarose, and plant gums, among others.
  • Frameworks containing living cells are prepared according to methods known in the art. For example, cells can be grown freely in a culture vessel to sub-confluency or confluency, lifted from the culture and inoculated onto the framework. Growth factors may be added to the culture medium prior to, during, or subsequent to inoculation of the cells to trigger differentiation and tissue formation, if desired. Alternatively, the frameworks themselves may be modified so that the growth of cells thereon is enhanced, or so that the risk of rejection of the implant is reduced. Thus, one or more biologically active compounds, including, but not limited to, anti-inflammatories, immunosuppressants or growth factors, may be added to the framework for local release.
  • Methods of Use
  • Postpartum cells, preferably PPDCs, or cell populations, components of or products produced by such cells, may be used in a variety of ways to support and facilitate repair and regeneration of ocular cells and tissues. Such utilities encompass in vitro, ex vivo and in vivo methods. The methods set forth below are directed to PPDCs, but other postpartum cells may also be suitable for use in those methods.
  • In Vitro and Ex Vivo Methods:
  • In one embodiment, PPDCs may be used in vitro to screen a wide variety of compounds for effectiveness and cytotoxicity of pharmaceutical agents, growth factors, regulatory factors, and the like. For example, such screening may be performed on substantially homogeneous populations of PPDCs to assess the efficacy or toxicity of candidate compounds to be formulated with, or co-administered with, the PPDCs, for treatment of a an ocular condition. Alternatively, such screening may be performed on PPDCs that have been stimulated to differentiate into a cell type found in the eye, or progenitor thereof, for the purpose of evaluating the efficacy of new pharmaceutical drug candidates. In this embodiment, the PPDCs are maintained in vitro and exposed to the compound to be tested. The activity of a potentially cytotoxic compound can be measured by its ability to damage or kill cells in culture. This may readily be assessed by vital staining techniques. The effect of growth or regulatory factors may be assessed by analyzing the number or robustness of the cultured cells, as compared with cells not exposed to the factors. This may be accomplished using standard cytological and/or histological techniques, including the use of immunocytochemical techniques employing antibodies that define type-specific cellular antigens.
  • In a further embodiment, as discussed above, PPDCs can be cultured in vitro to produce biological products that are either naturally produced by the cells, or produced by the cells when induced to differentiate into other lineages, or produced by the cells via genetic modification. For instance, TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1b, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were found to be secreted from umbilicus-derived cells grown in Growth Medium. TIMP1, TPO, KGF, HGF, HBEGF, BDNF, MIP1a, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were found to be secreted from placenta-derived PPDCs cultured in Growth Medium (see Examples). Some of these trophic factors, such as BDNF and IL-6, have important roles in neural regeneration. Other trophic factors, as yet undetected or unexamined, of use in repair and regeneration of ocular tissues, are likely to be produced by PPDCs and possibly secreted into the medium.
  • In this regard, another embodiment of the invention features use of PPDCs for production of conditioned medium, either from undifferentiated PPDCs or from PPDCs incubated under conditions that stimulate differentiation. Such conditioned media are contemplated for use in in vitro or ex vivo culture of epithelial or neural precursor cells, for example, or in vivo to support transplanted cells comprising homogeneous populations of PPDCs or heterogeneous populations comprising PPDCs and other progenitors, for example.
  • Yet another embodiment comprises the use of PPCD cell lysates, soluble cell fractions or components thereof, or ECM or components thereof, for a variety of purposes. As mentioned above, some of these components may be used in pharmaceutical compositions. In other embodiments, a cell lysate or ECM is used to coat or otherwise treat substances or devices to be used surgically, or for implantation, or for ex vivo purposes, to promote healing or survival of cells or tissues contacted in the course of such treatments.
  • As described in Examples 13 and 15, PPDCs have demonstrated the ability to support survival, growth and differentiation of adult neural progenitor cells when grown in co-culture with those cells. Likewise, the experimental results set forth in Example 18 indicates that PPDCs may function to support cells of the retina via trophic mechanisms. Accordingly, in another embodiment, PPDCs are used advantageously in co-cultures in vitro to provide trophic support to other cells, in particular neural cells and neural and ocular progenitors (e.g., neural stem cells and retinal or corneal epithelial stem cells). For co-culture, it may be desirable for the PPDCs and the desired other cells to be co-cultured under conditions in which the two cell types are in contact. This can be achieved, for example, by seeding the cells as a heterogeneous population of cells in culture medium or onto a suitable culture substrate. Alternatively, the PPDCs can first be grown to confluence, and then will serve as a substrate for the second desired cell type in culture. In this latter embodiment, the cells may further be physically separated, e.g., by a membrane or similar device, such that the other cell type may be removed and used separately, following the co-culture period. Use of PPDCs in co-culture to promote expansion and differentiation of neural or ocular cell types may find applicability in research and in clinical/therapeutic areas. For instance, PPDC co-culture may be utilized to facilitate growth and differentiation of such cells in culture, for basic research purposes or for use in drug screening assays, for example. PPDC co-culture may also be utilized for ex vivo expansion of neural or ocular progenitors for later administration for therapeutic purposes. For example, neural or ocular progenitor cells may be harvested from an individual, expanded ex vivo in co-culture with PPDCs, then returned to that individual (autologous transfer) or another individual (syngeneic or allogeneic transfer). In these embodiments, it will be appreciated that, following ex vivo expansion, the mixed population of cells comprising the PPDCs and progenitors could be administered to a patient in need of treatment. Alternatively, in situations where autologous transfer is appropriate or desirable, the co-cultured cell populations may be physically separated in culture, enabling removal of the autologous progenitors for administration to the patient.
  • In Vivo Methods:
  • As set forth in Examples 16, 17, and 18, PPDCs have been shown to be effectively transplanted into the body, and to supply lost neural or retinal function in animal models accepted for their predictability of efficacy in humans. These results support a preferred embodiment of the invention, wherein PPDCs are used in cell therapy for treating an ocular degenerative condition. Once transplanted into a target location in the eye, PPDCs may themselves differentiate into one or more phenotypes, or they may provide trophic support for ocular cells in situ, or they may exert a beneficial effect in both of those fashions, among others.
  • PPDCs may be administered alone (e.g., as substantially homogeneous populations) or as admixtures with other cells. As described above, PPDCs may be administered as formulated in a pharmaceutical preparation with a matrix or scaffold, or with conventional pharmaceutically acceptable carriers. Where PPDCs are administered with other cells, they may be administered simultaneously or sequentially with the other cells (either before or after the other cells). Cells that may be administered in conjunction with PPDCs include, but are not limited to, neurons, astrocytes, oligodendrocytes, neural progenitor cells, neural stem cells, ocular progenitor cells, retinal or corneal epithelial stem cells and/or other multipotent or pluripotent stem cells. The cells of different types may be admixed with the PPDCs immediately or shortly prior to administration, or they may be co-cultured together for a period of time prior to administration.
  • The PPDCs may be administered with other beneficial drugs, biological molecules, such as growth factors, trophic factors, conditioned medium (from postpartum cells or from progenitor or differentiated cell cultures), or other active agents, such as anti-inflammatory agents, anti-apoptotic agents, antioxidants, growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art. When PPDCs are administered with other agents, they may be administered together in a single pharmaceutical composition, or in separate pharmaceutical compositions, simultaneously or sequentially with the other agents (either before or after administration of the other agents).
  • Examples of other components that may be administered with postpartum cells include, but are not limited to: (1) other neuroprotective or neurobeneficial drugs; (2) selected extracellular matrix components, such as one or more types of collagen known in the art, and/or growth factors, platelet-rich plasma, and drugs (alternatively, the cells may be genetically engineered to express and produce growth factors); (3) anti-apoptotic agents (e.g., erythropoietin (EPO), EPO mimetibody, thrombopoietin, insulin-like growth factor (IGF)-I, IGF-II, hepatocyte growth factor, caspase inhibitors); (4) anti-inflammatory compounds (e.g., p38 MAP kinase inhibitors, TGF-beta inhibitors, statins, IL-6 and IL-1 inhibitors, PEMIROLAST, TRANILAST, REMICADE, SIROLIMUS, and non-steroidal anti-inflammatory drugs (NSAIDS) (such as TEPOXALIN, TOLMETIN, and SUPROFEN); (5) immunosuppressive or immunomodulatory agents, such as calcineurin inhibitors, mTOR inhibitors, antiproliferatives, corticosteroids and various antibodies; (6) antioxidants such as probucol, vitamins C and E, conenzyme Q-10, glutathione, L-cysteine and N-acetylcysteine; and (6) local anesthetics, to name a few.
  • In one embodiment, PPDCs are administered as undifferentiated cells, i.e., as cultured in Growth Medium. Alternatively, PPDCs may be administered following exposure in culture to conditions that stimulate differentiation toward a desired phenotype.
  • The cells may be surgically implanted, injected or otherwise administered directly or indirectly to the site of ocular damage or distress. When cells are administered in semi-solid or solid devices, surgical implantation into a precise location in the body is typically a suitable means of administration. Liquid or fluid pharmaceutical compositions, however, may be administered to a more general location in the eye (e.g., topically or intra-ocularly).
  • Other embodiments encompass methods of treating ocular degenerative conditions by administering pharmaceutical compositions comprising PPDC cellular components (e.g., cell lysates or components thereof) or products (e.g., trophic and other biological factors produced naturally by PPDCs or through genetic modification, conditioned medium from PPDC culture). Again, these methods may further comprise administering other active agents, such as growth factors, neurotrophic factors or neuroregenerative or neuroprotective drugs as known in the art.
  • Dosage forms and regimes for administering PPDCs or any of the other pharmaceutical compositions described herein are developed in accordance with good medical practice, taking into account the condition of the individual patient, e.g., nature and extent of the ocular degenerative condition, age, sex, body weight and general medical condition, and other factors known to medical practitioners. Thus, the effective amount of a pharmaceutical composition to be administered to a patient is determined by these considerations as known in the art.
  • It may be desirable or appropriate to pharmacologically immunosuppress a patient prior to initiating cell therapy. This may be accomplished through the use of systemic or local immunosuppressive agents, or it may be accomplished by delivering the cells in an encapsulated device, as described above. These and other means for reducing or eliminating an immune response to the transplanted cells are known in the art. As an alternative, PPDCs may be genetically modified to reduce their immunogenicity, as mentioned above.
  • Survival of transplanted cells in a living patient can be determined through the use of a variety of scanning techniques, e.g., computerized axial tomography (CAT or CT) scan, magnetic resonance imaging (MRI) or positron emission tomography (PET) scans. Determination of transplant survival can also be done post mortem by removing the tissue and examining it visually or through a microscope. Alternatively, cells can be treated with stains that are specific for neural or ocular cells or products thereof, e.g., neurotransmitters. Transplanted cells can also be identified by prior incorporation of tracer dyes such as rhodamine- or fluorescein-labeled microspheres, fast blue, ferric microparticles, bisbenzamide or genetically introduced reporter gene products, such as beta-galactosidase or beta-glucuronidase.
  • Functional integration of transplanted cells into ocular tissue of a subject can be assessed by examining restoration of the ocular function that was damaged or diseased. For example, effectiveness in the treatment of macular degeneration or other retinopathies may be determined by improvement of visual acuity and evaluation for abnormalities and grading of stereoscopic color fundus photographs. (Age-Related Eye Disease Study Research Group, NEI, NIH, AREDS Report No. 8, 2001, Arch. Ophthalmol. 119: 1417-1436).
  • Kits and Banks
  • In another aspect, the invention provides kits that utilize postpartum cells, preferably PPDCs, cell populations, components and products thereof in various methods for ocular regeneration and repair as described above. Where used for treatment of ocular degenerative conditions, or other scheduled treatment, the kits may include one or more cell populations, including at least postpartum cells and a pharmaceutically acceptable carrier (liquid, semi-solid or solid). The kits also optionally may include a means of administering the cells, for example by injection. The kits further may include instructions for use of the cells. Kits prepared for field hospital use, such as for military use may include full-procedure supplies including tissue scaffolds, surgical sutures, and the like, where the cells are to be used in conjunction with repair of acute injuries. Kits for assays and in vitro methods as described herein may contain, for example, one or more of (1) PPDCs or components or products of PPDCs, (2) reagents for practicing the in vitro method, (3) other cells or cell populations, as appropriate, and (4) instructions for conducting the in vitro method.
  • In yet another aspect, the invention also provides for banking of tissues, cells, cellular components and cell populations of the invention. As discussed above, the cells are readily cryopreserved. The invention therefore provides methods of cryopreserving the cells in a bank, wherein the cells are stored frozen and associated with a complete characterization of the cells based on immunological, biochemical and genetic properties of the cells. The frozen cells can be thawed and expanded or used directly for autologous, syngeneic, or allogeneic therapy, depending on the requirements of the procedure and the needs of the patient. Preferably, the information on each cryopreserved sample is stored in a computer, which is searchable based on the requirements of the surgeon, procedure and patient with suitable matches being made based on the characterization of the cells or populations. Preferably, the cells of the invention are grown and expanded to the desired quantity of cells and therapeutic cell compositions are prepared either separately or as co-cultures, in the presence or absence of a matrix or support. While for some applications it may be preferable to use cells freshly prepared, the remainder can be cryopreserved and banked by freezing the cells and entering the information in the computer to associate the computer entry with the samples. Even where it is not necessary to match a source or donor with a recipient of such cells, for immunological purposes, the bank system makes it easy to match, for example, desirable biochemical or genetic properties of the banked cells to the therapeutic needs. Upon matching of the desired properties with a banked sample, the sample is retrieved and prepared for therapeutic use. Cell lysates, ECM or cellular components prepared as described herein may also be cryopreserved or otherwise preserved (e.g., by lyophilization) and banked in accordance with the present invention.
  • The following examples are provided to describe the invention in greater detail. They are intended to illustrate, not to limit, the invention.
  • As used in the following examples and elsewhere in the specification, the term Growth Medium generally refers to a medium sufficient for the culturing of PPDCs. In particular, one presently preferred medium for the culturing of the cells of the invention in comprises Dulbecco's Modified Essential Media (also abbreviated DMEM herein). Particularly preferred is DMEM-low glucose (also DMEM-LG herein) (Invitrogen, Carlsbad, Calif.). The DMEM-low glucose is preferably supplemented with 15% (v/v) fetal bovine serum (e.g. defined fetal bovine serum, Hyclone, Logan Utah), antibiotics/antimycotics ((preferably 50-100 Units/milliliter penicillin, 50-100 microgram/milliliter streptomycin, and 0-0.25 microgram/milliliter amphotericin B; Invitrogen, Carlsbad, Calif.)), and 0.001% (v/v) 2-mercaptoethanol (Sigma, St. Louis Mo.). As used in the Examples below, Growth Medium refers to DMEM-low glucose with 15% fetal bovine serum and antibiotics/antimycotics (when penicillin/streptomycin are included, it is preferably at 50 U/ml and 50 microgram/ml respectively; when penicillin/streptomycin/amphotericin B are use, it is preferably at 100 U/ml, 100 microgram/ml and 0.25 microgram/ml, respectively). In some cases different growth media are used, or different supplementations are provided, and these are normally indicated in the text as supplementations to Growth Medium.
  • The following abbreviations may appear in the examples and elsewhere in the specification and claims: ANG2 (or Ang2) for angiopoietin 2; APC for antigen-presenting cells; BDNF for brain-derived neurotrophic factor; bFGF for basic fibroblast growth factor; bid (BID) for “bis in die” (twice per day); CK18 for cytokeratin 18; CNS for central nervous system; CXC ligand 3 for chemokine receptor ligand 3; DMEM for Dulbecco's Minimal Essential Medium; DMEM:1 g (or DMEM:Lg, DMEM:LG) for DMEM with low glucose; EDTA for ethylene diamine tetraacetic acid; EGF (or E) for epidermal growth factor; FACS for fluorescent activated cell sorting; FBS for fetal bovine serum; FGF (or F) for fibroblast growth factor; GCP-2 for granulocyte chemotactic protein-2; GFAP for glial fibrillary acidic protein; HB-EGF for heparin-binding epidermal growth factor; HCAEC for Human coronary artery endothelial cells; HGF for hepatocyte growth factor; hMSC for Human mesenchymal stem cells; BNF-1 alpha for hepatocyte-specific transcription factor; HUVEC for Human umbilical vein endothelial cells; 1309 for a chemokine and the ligand for the CCR8 receptor; IGF-1 for insulin-like growth factor 1; IL-6 for interleukin-6; IL-8 for interleukin 8; K19 for keratin 19; K8 for keratin 8; KGF for keratinocyte growth factor; LIF for leukemia inhibitory factor; MBP for myelin basic protein; MCP-1 for monocyte chemotactic protein 1; MDC for macrophage-derived chemokine; MIP1alpha for macrophage inflammatory protein 1 alpha; MIP1beta for macrophage inflammatory protein 1beta; MMP for matrix metalloprotease (MMP); MSC for mesenchymal stem cells; NHDF for Normal Human Dermal Fibroblasts; NPE for Neural Progenitor Expansion media; O4 for oligodendrocyte or glial differentiation marker O4; PBMC for Peripheral blood mononuclear cell; PBS for phosphate buffered saline; PDGFbb for platelet derived growth factor; PO for “per os” (by mouth); PNS for peripheral nervous system; Rantes (or RANTES) for regulated on activation, normal T cell expressed and secreted; rhGDF-5 for recombinant human growth and differentiation factor 5; SC for subcutaneously; SDF-1alpha for stromal-derived factor 1 alpha; SHH for sonic hedgehog; SOP for standard operating procedure; TARC for thymus and activation-regulated chemokine; TCP for Tissue culture plastic; TCPS for tissue culture polystyrene; TGFbeta2 for transforming growth factor beta2; TGF beta-3 for transforming growth factor beta-3; TIMP1 for tissue inhibitor of matrix metalloproteinase 1; TPO for thrombopoietin; TuJ1 for BIII Tubulin; VEGF for vascular endothelial growth factor; vWF for von Willebrand factor; and alphaFP for alpha-fetoprotein.
  • EXAMPLE 1 Derivation of Cells from Postpartum Tissue
  • This example describes the preparation of postpartum-derived cells from placental and umbilical cord tissues. Postpartum umbilical cords and placentae were obtained upon birth of either a full term or pre-term pregnancy. Cells were harvested from 5 separate donors of umbilicus and placental tissue. Different methods of cell isolation were tested for their ability to yield cells with: 1) the potential to differentiate into cells with different phenotypes, a characteristic common to stem cells, or 2) the potential to provide trophic factors useful for other cells and tissues.
  • Methods & Materials
  • Umbilical cell isolation. Umbilical cords were obtained from National Disease Research Interchange (NDR1, Philadelphia, Pa.). The tissues were obtained following normal deliveries. The cell isolation protocol was performed aseptically in a laminar flow hood. To remove blood and debris, the cord was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of antimycotic and antibiotic (100 units/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B). The tissues were then mechanically dissociated in 150 cm2 tissue culture plates in the presence of 50 milliliters of medium (DMEM-Low glucose or DMEM-High glucose; Invitrogen), until the tissue was minced into a fine pulp. The chopped tissues were transferred to 50 milliliter conical tubes (approximately 5 grams of tissue per tube). The tissue was then digested in either DMEM-Low glucose medium or DMEM-High glucose medium, each containing antimycotic and antibiotic as described above. In some experiments, an enzyme mixture of collagenase and dispase was used (“C:D;” collagenase (Sigma, St Louis, Mo.), 500 Units/milliliter; and dispase (Invitrogen), 50 Units/milliliter in DMEM:-Low glucose medium). In other experiments a mixture of collagenase, dispase and hyaluronidase (“C:D:H”) was used (collagenase, 500 Units/milliliter; dispase, 50 Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter, in DMEM:-Low glucose). The conical tubes containing the tissue, medium and digestion enzymes were incubated at 37° C. in an orbital shaker (Environ, Brooklyn, NY) at 225 rpm for 2 hrs.
  • After digestion, the tissues were centrifuged at 150×g for 5 minutes, the supernatant was aspirated. The pellet was resuspended in 20 milliliters of Growth Medium (DMEM:Low glucose (Invitrogen), 15 percent (v/v) fetal bovine serum (FBS; defined bovine serum; Lot#AND18475; Hyclone, Logan, Utah), 0.001% (v/v) 2-mercaptoethanol (Sigma), 1 milliliter per 100 milliliters of antibiotic/antimycotic as described above. The cell suspension was filtered through a 70-micrometer nylon cell strainer (BD Biosciences). An additional 5 milliliters rinse comprising Growth Medium was passed through the strainer. The cell suspension was then passed through a 40-micrometer nylon cell strainer (BD Biosciences) and chased with a rinse of an additional 5 milliliters of Growth Medium.
  • The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150×g for 5 minutes. The supernatant was aspirated and the cells were resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more.
  • Upon the final centrifugation supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. The number of viable cells was determined using Trypan Blue staining. Cells were then cultured under standard conditions.
  • The cells isolated from umbilical cords were seeded at 5,000 cells/cm2 onto gelatin-coated T-75 cm2 flasks (Corning Inc., Corning, N.Y.) in Growth Medium with antibiotics/antimycotics as described above. After 2 days (in various experiments, cells were incubated from 2-4 days), spent medium was aspirated from the flasks. Cells were washed with PBS three times to remove debris and blood-derived cells. Cells were then replenished with Growth Medium and allowed to grow to confluence (about 10 days from passage 0) to passage 1. On subsequent passages (from passage 1 to 2 and so on), cells reached sub-confluence (75-85 percent confluence) in 4-5 days. For these subsequent passages, cells were seeded at 5000 cells/cm2. Cells were grown in a humidified incubator with 5 percent carbon dioxide and atmospheric oxygen, at 37° C.
  • Placental Cell Isolation. Placental tissue was obtained from NDR1 (Philadelphia, Pa.). The tissues were from a pregnancy and were obtained at the time of a normal surgical delivery. Placental cells were isolated as described for umbilical cell isolation.
  • The following example applies to the isolation of separate populations of maternal-derived and neonatal-derived cells from placental tissue.
  • The cell isolation protocol was performed aseptically in a laminar flow hood. The placental tissue was washed in phosphate buffered saline (PBS; Invitrogen, Carlsbad, Calif.) in the presence of antimycotic and antibiotic (as described above) to remove blood and debris. The placental tissue was then dissected into three sections: top-line (neonatal side or aspect), mid-line (mixed cell isolation neonatal and maternal) and bottom line (maternal side or aspect).
  • The separated sections were individually washed several times in PBS with antibiotic/antimycotic to further remove blood and debris. Each section was then mechanically dissociated in 150 cm2 tissue culture plates in the presence of 50 milliliters of DMEM/Low glucose, to a fine pulp. The pulp was transferred to 50 milliliter conical tubes. Each tube contained approximately 5 grams of tissue. The tissue was digested in either DMEM-Low glucose or DMEM-High glucose medium containing antimycotic and antibiotic (100 U/milliliter penicillin, 100 micrograms/milliliter streptomycin, 0.25 micrograms/milliliter amphotericin B) and digestion enzymes. In some experiments an enzyme mixture of collagenase and dispase (“C:D”) was used containing collagenase (Sigma, St Louis, Mo.) at 500 Units/milliliter and dispase (Invitrogen) at 50 Units/milliliter in DMEM-Low glucose medium. In other experiments a mixture of collagenase, dispase and hyaluronidase (C:D:H) was used (collagenase, 500 Units/milliliter; dispase, 50 Units/milliliter; and hyaluronidase (Sigma), 5 Units/milliliter in DMEM-Low glucose). The conical tubes containing the tissue, medium, and digestion enzymes were incubated for 2 h at 37° C. in an orbital shaker (Environ, Brooklyn, N.Y.) at 225 rpm.
  • After digestion, the tissues were centrifuged at 150×g for 5 minutes, the resultant supernatant was aspirated off. The pellet was resuspended in 20 milliliters of Growth Medium with penicillin/streptomycin/amphotericin B. The cell suspension was filtered through a 70 micometer nylon cell strainer (BD Biosciences), chased by a rinse with an additional 5 milliliters of Growth Medium. The total cell suspension was passed through a 40 micometer nylon cell strainer (BD Biosciences) followed with an additional 5 milliliters of Growth Medium as a rinse.
  • The filtrate was resuspended in Growth Medium (total volume 50 milliliters) and centrifuged at 150×g for 5 minutes. The supernatant was aspirated and the cell pellet was resuspended in 50 milliliters of fresh Growth Medium. This process was repeated twice more. After the final centrifugation, supernatant was aspirated and the cell pellet was resuspended in 5 milliliters of fresh Growth Medium. A cell count was determined using the Trypan Blue Exclusion test. Cells were then cultured at standard conditions.
  • LIBERASE Cell Isolation. Cells were isolated from umbilicus tissues in DMEM-Low glucose medium with LIBERASE (Boehringer Mannheim Corp., Indianapolis, Ind.) (2.5 milligrams per milliliter, Blendzyme 3; Roche Applied Sciences, Indianapolis, Ind.) and hyaluronidase (5 Units/milliliter, Sigma). Digestion of the tissue and isolation of the cells was as described for other protease digestions above, using the LIBERASE/hyaluronidase mixture in place of the C:D or C:D:H enzyme mixture. Tissue digestion with LIBERASE resulted in the isolation of cell populations from postpartum tissues that expanded readily.
  • Cell isolation using other enzyme combinations. Procedures were compared for isolating cells from the umbilical cord using differing enzyme combinations. Enzymes compared for digestion included: i) collagenase; ii) dispase; iii) hyaluronidase; iv) collagenase:dispase mixture (C;D); v) collagenase:hyaluronidase mixture (C:H); vi) dispase:hyaluronidase mixture (D:H); and vii) collagenase:dispase:hyaluronidase mixture (C:D:H). Differences in cell isolation utilizing these different enzyme digestion conditions were observed (Table 1-1).
    TABLE 1-1
    Isolation of cells from umbilical cord tissue using varying enzyme
    combinations
    Enzyme Digest Cells Isolated Cell Expansion
    Collagenase X X
    Dispase  + (>10 h) +
    Hyaluronidase X X
    Collagenase:Dispase ++ (<3 h) ++
    Collagenase:Hyaluronidase ++ (<3 h) +
    Dispase:Hyaluronidase  + (>10 h) +
    Collagenase:Dispase:Hyaluronidase +++ (<3 h)  +++

    Key: + = good,

    ++ = very good,

    +++ = excellent,

    X = no success under conditions tested
  • Isolation of cells from residual blood in the cords. Other attempts were made to isolate pools of cells from umbilical cord by different approaches. In one instance umbilical cord was sliced and washed with Growth Medium to dislodge the blood clots and gelatinous material. The mixture of blood, gelatinous material and Growth Medium was collected and centrifuged at 150×g. The pellet was resuspended and seeded onto gelatin-coated flasks in Growth Medium. From these experiments a cell population was isolated that readily expanded.
  • Isolation of cells from cord blood. Cells have also been isolated from cord blood samples attained from NDR1. The isolation protocol used here was that of International Patent Application US0229971 by Ho et al (Ho, T. W. et al., WO2003025149 A2). Samples (50 milliliter and 10.5 milliliters, respectively) of umbilical cord blood (NDR1, Philadelphia Pa.) were mixed with lysis buffer (filter-sterilized 155 mM ammonium chloride, 10 millimolar potassium bicarbonate, 0.1 millimolar EDTA buffered to pH 7.2 (all components from Sigma, St. Louis, Mo.)). Cells were lysed at a ratio of 1:20 cord blood to lysis buffer. The resulting cell suspension was vortexed for 5 seconds, and incubated for 2 minutes at ambient temperature. The lysate was centrifuged (10 minutes at 200×g). The cell pellet was resuspended in complete minimal essential medium (Gibco, Carlsbad Calif.) containing 10 percent fetal bovine serum (Hyclone, Logan Utah), 4 millimolar glutamine (Mediatech Hemdon, VA), 100 Units penicillin per 100 milliliters and 100 micrograms streptomycin per 100 milliliters (Gibco, Carlsbad, Calif.). The resuspended cells were centrifuged (10 minutes at 200×g), the supernatant was aspirated, and the cell pellet was washed in complete medium. Cells were seeded directly into either T75 flasks (Corning, N.Y.), T75 laminin-coated flasks, or T175 fibronectin-coated flasks (both Becton Dickinson, Bedford, Mass.).
  • Isolation of cells using different enzyme combinations and growth conditions. To determine whether cell populations could be isolated under different conditions and expanded under a variety of conditions immediately after isolation, cells were digested in Growth Medium with or without 0.001 percent (v/v) 2-mercaptoethanol (Sigma, St. Louis, Mo.), using the enzyme combination of C:D:H, according to the procedures provided above. Placental-derived cells so isolated were seeded under a variety of conditions. All cells were grown in the presence of penicillin/streptomycin. (Table 1-2).
    TABLE 1-2
    Isolation and culture expansion of postpartum cells under varying conditions:
    Condition Medium 15% FBS BME Gelatin 20% O2 Growth Factors
    1 DMEM-Lg Y Y Y Y N
    2 DMEM-Lg Y Y Y N (5%) N
    3 DMEM-Lg Y Y N Y N
    4 DMEM-Lg Y Y N N (5%) N
    5 DMEM-Lg N (2%) Y N (Laminin) Y EGF/FGF (20 ng/mL)
    6 DMEM-Lg N (2%) Y N (Laminin) N (5%) EGF/FGF (20 ng/mL)
    7 DMEM-Lg N (2%) Y N (Fibrone) Y PDGF/VEGF
    8 DMEM-Lg N (2%) Y N (Fibrone) N (5%) PDGF/VEGF
    9 DMEM-Lg Y N Y Y N
    10 DMEM-Lg Y N Y N (5%) N
    11 DMEM-Lg Y N N Y N
    12 DMEM-Lg Y N N N (5%) N
    13 DMEM-Lg N (2%) N N (Laminin) Y EGF/FGF (20 ng/mL)
    14 DMEM-Lg N (2%) N N (Laminin) N (5%) EGF/FGF (20 ng/mL)
    15 DMEM-Lg N (2%) N N (Fibrone) Y PDGF/VEGF
    16 DMEM-Lg N (2%) N N (Fibrone) N (5%) PDGF/VEGF
  • Isolation of cells using different enzyme combinations and growth conditions. In all conditions cells attached and expanded well between passage 0 and 1 (Table 1-2). Cells in conditions 5-8 and 13-16 were demonstrated to proliferate well up to 4 passages after seeding at which point they were cryopreserved and banked.
  • Cell isolation using different enzyme combinations. The combination of C:D:H, provided the best cell yield following isolation, and generated cells which expanded for many more generations in culture than the other conditions (Table 1). An expandable cell population was not attained using collagenase or hyaluronidase alone. No attempt was made to determine if this result is specific to the collagen that was tested.
  • Isolation of cells using different enzyme combinations and growth conditions. Cells attached and expanded well between passage 0 and 1 under all conditions tested for enzyme digestion and growth (Table 2). Cells in experimental conditions 5-8 and 13-16 proliferated well up to 4 passages after seeding, at which point they were cryopreserved. All cells were banked for further investigation.
  • Isolation of cells from residual blood in the cords. Nucleated cells attached and grew rapidly. These cells were analyzed by flow cytometry and were similar to cells obtained by enzyme digestion.
  • Isolation of cells from cord blood. The preparations contained red blood cells and platelets. No nucleated cells attached and divided during the first 3 weeks. The medium was changed 3 weeks after seeding and no cells were observed to attach and grow.
  • Summary. Populations of cells can be derived from umbilical cord and placental tissue efficiently using the enzyme combination collagenase (a matrix metalloprotease), dispase (a neutral protease) and hyaluronidase (a mucolytic enzyme that breaks down hyaluronic acid). LIBERASE, which is a Blendzyme, may also be used. Specifically, Blendzyme 3, which is collagenase (4 Wunsch units/g) and thermolysin (1714 casein Units/g) was also used together with hyaluronidase to isolate cells. These cells expanded readily over many passages when cultured in Growth Medium on gelatin coated plastic.
  • Cells were also isolated from residual blood in the cords, but not cord blood. The presence of cells in blood clots washed from the tissue, that adhere and grow under the conditions used, may be due to cells being released during the dissection process.
  • EXAMPLE 2 Growth Characteristics of Postpartum-Derived Cells
  • The cell expansion potential of postpartum-derived cells (PPDCs) was compared to other populations of isolated stem cells. The process of cell expansion to senescence is referred to as Hayflick's limit (Hayflick L. 1974a, 1974b). Postpartum-derived cells are highly suited for therapeutic use because they can be readily expanded to sufficient cell numbers.
  • Materials and Methods
  • Gelatin-coating flasks. Tissue culture plastic flasks were coated by adding 20 milliliters 2% (w/v) porcine gelatin (Type B: 225 Bloom; Sigma, St Louis, Mo.) to a T75 flask (Corning, Corning, N.Y.) for 20 minutes at room temperature. After removing the gelatin solution, 10 milliliters phosphate-buffered saline (PBS) (Invitrogen, Carlsbad, Calif.) was added and then aspirated.
  • Comparison of expansion potential of PPDCs with other cell populations. For comparison of growth expansion potential the following cell populations were utilized; i) Mesenchymal stem cells (MSC; Cambrex, Walkersville, MD); ii) Adipose-derived cells (U.S. U.S. Pat. No. 6,555,374 B1; U.S. Patent Application US20040058412); iii) Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, MD); iv) Umbilicus-derived cells; and v) Placenta-derived cells. Cells were initially seeded at 5,000 cells/cm2 on gelatin-coated T75 flasks in Growth Medium with penicillin/streptomycin/amphotericin B. For subsequent passages, cell cultures were treated as follows. After trypsinization, viable cells were counted after Trypan Blue staining. Cell suspension (50 microliters) was combined with Trypan Blue (50 milliliters, Sigma, St. Louis Mo.). Viable cell numbers were estimated using a hemocytometer.
  • Following counting, cells were seeded at 5,000 cells/cm2 onto gelatin-coated T 75 flasks in 25 milliliters of fresh Growth Medium. Cells were grown under standard conditions at 37° C. The Growth Medium was changed twice per week. When cells reached about 85 percent confluence they were passaged; this process was repeated until the cells reached senescence.
  • At each passage, cells were trypsinized and counted. The viable cell yield, population doubling [ln (cell final/cell initial)/ln 2] and doubling time (time in culture (h)/population doubling) were calculated. For the purposes of determining optimal cell expansion, the total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor=cell final/cell initial).
  • Expansion potential of cell banks at low density. The expansion potential of cells banked at passage 10 was also tested, using a different set of conditions. Normal dermal skin fibroblasts (cc-2509 lot # 9F0844; Cambrex, Walkersville, MD), umbilicus-derived cells, and placenta-derived cells were tested. These cell populations had been banked at passage 10 previously, having been cultured at 5,000 cells/cm2 and grown to confluence at each passage to that point. The effect of cell density on the cell populations following cell thaw at passage 10 was determined. Cells were thawed under standard conditions and counted using Trypan Blue staining. Thawed cells were then seeded at 1000 cells/cm2 in DMEM:Low glucose Growth Medium with antibiotic/antimycotic as described above. Cells were grown under standard atmospheric conditions at 37° C. Growth Medium was changed twice a week and cells were passaged as they reached about 85% confluence. Cells were subsequently passaged until senescence, i.e., until they could not be expanded any further. Cells were trypsinized and counted at each passage. The cell yield, population doubling (ln (cell final/cell initial)/ln 2) and doubling time (time in culture (h)/population doubling). The total cell yield per passage was determined by multiplying total yield for the previous passage by the expansion factor for each passage (i.e., expansion factor=cell final/cell initial).
  • Expansion of PPDCs at low density from initial cell seeding. The expansion potential of freshly isolated PPDCs under low cell seeding conditions was tested. PPDDs were prepared as described herein. Cells were seeded at 1000 cells/cm2 and passaged as described above until senescence. Cells were grown under standard atmospheric conditions at 37° C. Growth Medium was changed twice per week. Cells were passaged as they reached about 85% confluence. At each passage, cells were trypsinized and counted by Trypan Blue staining. The cell yield, population doubling (In (cell final/cell initial)/ln 2) and doubling time (time in culture (h)/population doubling) were calculated for each passage. The total cell yield per passage was determined by multiplying the total yield for the previous passage by the expansion factor for each passage (i.e. expansion factor=cell final/cell initial). Cells were grown on gelatin and non-gelatin coated flasks.
  • Expansion of clonal neonatal placenta-derived cells. Cloning was used in order to expand a population of neonatal cells from placental tissue. Following isolation of three differential cell populations from the placenta (as described herein), these cell populations were expanded under standard growth conditions and then karyotyped to reveal the identity of the isolated cell populations. Because the cells were isolated from a mother who delivered a boy, it was straightforward to distinguish between the male and female chromosomes by performing metaphase spreads. These experiments demonstrated that fetal-aspect cells were karyotype positive for neonatal phenotpye, mid-layer cells were karyotype positive for both neonatal and maternal phenotypes and maternal-aspect cells were karyotype positive for maternal cells.
  • Expansion of cells in low oxygen culture conditions. It has been demonstrated that low oxygen cell culture conditions can improve cell expansion in certain circumstances (US20040005704). To determine if cell expansion of PPDCs could be improved by altering cell culture conditions, cultures of umbilical-derived cells were grown in low oxygen conditions. Cells were seeded at 5000 cells/cm2 in Growth Medium on gelatin coated flasks. Cells were initially cultured under standard atmospheric conditions through passage 5, at which point they were transferred to low oxygen (5% O2) culture conditions.
  • Other growth conditions. In other protocols, cells were expanded on non-coated, collagen-coated, fibronectin-coated, laminin-coated and extracellular matrix protein-coated plates. Cultures have been demonstrated to expand well on these different matrices.
  • Results
  • Comparison of expansion potential of PPDCs with other stem cell and non-stem cell populations. Both umbilical-derived and placenta-derived cells expanded for greater than 40 passages generating cell yields of >1E17 cells in 60 days. In contrast, MSCs and fibroblasts senesced after <25 days and <60 days, respectively. Although adipose-derived cells expanded for almost 60 days they generated total cell yields of 4.5E12. Thus, when seeded at 5000 cells/cm2 under the experimental conditions utilized, postpartum-derived cells expanded much better than the other cell types grown under the same conditions (Table 2-1).
    TABLE 2-1
    Growth characteristics for different cell populations grown to senescence
    Total Population Total Cell
    Cell Type Senescence Doublings Yield
    MSC 24 d 8 4.72E7 
    Adipose 57 d 24  4.5E12
    Fibroblasts 53 d 26 2.82E13
    Umbilicus 65 d 42 6.15E17
    Placenta 80 d 46 2.49E19
  • Expansion potential of cell banks at low density. Umbilicus-derived, placenta-derived and fibroblast cells expanded for greater than 10 passages generating cell yields of >1E11 cells in 60 days (Table 2-2). After 60 days under these conditions the fibroblasts became senescent whereas the umbilicus-derived and placenta-derived cell populations senesced after 80 days, completing >50 and >40 population doublings respectively.
    TABLE 2-2
    Growth characteristics for different cell populations using low density
    growth expansion from passage 10 till senescence
    Total Population Total Cell
    Cell Type Senescence Doublings Yield
    Fibroblast (P10) 80 d 43.68 2.59E11
    Umbilicus (P10) 80 d 53.6 1.25E14
    Placenta (P10) 60 d 32.96 6.09E12
  • Expansion of PPDCs at low density from initial cell seeding. PPDCs were expanded at low density (1,000 cells/cm2) on gelatin-coated and uncoated plates or flasks. Growth potential of these cells under these conditions was good. The cells expanded readily in a log phase growth. The rate of cell expansion was similar to that observed when placenta-derived cells were seeded at 5000 cells/cm2 on gelatin-coated flasks in Growth Medium. No differences were observed in cell expansion potential between culturing on either uncoated flasks or gelatin-coated flasks. However, cells appeared phenotypically much smaller on gelatin-coated flasks and more larger cell phenotypes were observed on uncoated flasks.
  • Expansion of clonal neonatal or maternal placenta-derived cells. A clonal neonatal or maternal cell population can be expanded from placenta-derived cells isolated from the neonatal aspect or the maternal aspect, respectively, of the placenta. Cells are serially diluted and then seeded onto gelatin-coated plates in Growth medium for expansion at 1 cell/well in 96-well gelatin coated plates. From this initial cloning, expansive clones are identified, trypsinized, and reseeded in 12-well gelatin-coated plates in Growth medium and then subsequently passaged into T25 gelatin-coated flasks at 5,000 cells/cm2 in Growth medium. Subcloning is performed to ensure that a clonal population of cells has been identified. For subcloning experiments, cells are trypsinized and reseeded at 0.5 cells/well. The subclones that grow well are expanded in gelatin-coated T25 flasks at 5,000 cells cm2/flask. Cells are passaged at 5,000 cells cm2/T75 flask. The growth characteristics of a clone may be plotted to demonstrate cell expansion. Karyotyping analysis can confirm that the clone is either neonatal or maternal.
  • Expansion of cells in low oxygen culture conditions. Cells expanded well under the reduced oxygen conditions, however, culturing under low oxygen conditions did not appear to have a significant effect on cell expansion of PPDCs under the conditions used.
  • Summary. Cell expansion conditions comprising growing isolated postpartum-derived cells at densities of about 5000 cells/cm2, in Growth Medium on gelatin-coated or uncoated flasks, under standard atmospheric oxygen, are sufficient to generate large numbers of cells at passage 11. Furthermore, the data suggests that the cells can be readily expanded using lower density culture conditions (e.g. 1000 cells/cm2). Postpartum-derived cell expansion in low oxygen conditions also facilitates cell expansion, although no incremental improvement in cell expansion potential has yet been observed when utilizing these conditions for growth. Presently, culturing postpartum-derived cells under standard atmospheric conditions is preferred for generating large pools of cells. However, when the culture conditions are altered, postpartum-derived cell expansion can likewise be altered. This strategy may be used to enhance the proliferative and differentiative capacity of these cell populations.
  • Under the conditions utilized, while the expansion potential of MSC and adipose-derived cells is limited, postpartum-derived cells expand readily to large numbers.
  • References for Example 2
    • 1) Hayflick L. 1974a. J Am Geriatr Soc. 22:1-12.
    • 2) Hayflick L. 1974b. Gerontologist. 14:37-45.
    • 3) U.S. Patent publication US20040058412
    • 4) U.S. Patent publication US20040048372
    • 5) U.S. Patent publication US20040005704.
    EXAMPLE 3 Evaluation of Growth Media for Placenta-Derived Cells
  • Several cell culture media were evaluated for their ability to support the growth of placenta-derived cells. The growth of placenta-derived cells in normal (20%) and low (5%) oxygen was assessed after 3 days using the MTS calorimetric assay.
  • Methods & Materials
  • Placenta-derived cells at passage 8 (P8) were seeded at 1×103 cells/well in 96 well plates in Growth Medium with penicillin/streptomycin. After 8 hours the medium was changed as described below and cells were incubated in normal (atmospheric) or low (5%, v/v) oxygen at 37° C., 5% CO2 for 48 hours. MTS was added to the culture medium (CELLTITER 96 AQueous One Solution Cell Proliferation Assay, Promega, Madison, Wis.) for 3 hours and the absorbance measured at 490 nanometers (Molecular Devices, Sunnyvale Calif.).
    TABLE 3-1
    Culture Media
    Added
    fetal bovine
    Culture Medium Supplier serum % (v/v)
    DMEM low glucose Gibco Carlsbad CA 0, 2 10
    DMEM high glucose Gibco 0, 2 10
    RPMI 1640 Mediatech, Inc. 0, 2 10
    Herndon, VA
    Cell gro-free (Serum-free, Mediatech, Inc.
    Protein-free
    Ham's F10 Mediatech, Inc. 0, 2 10
    MSCGM (complete with Cambrex, Walkersville, 0, 2 10
    serum) MD
    Complete-serum free Mediatech, Inc.
    w/albumin
    Growth Medium NA
    Ham's F12 Mediatech, Inc. 0, 2 10
    Iscove's Mediatech, Inc. 0, 2 10
    Basal Medium Eagle's Mediatech, Inc.
    DMEM/F12 (1:1) Mediatech, Inc. 0, 2 10

    Results
  • Standard curves for the MTS assay established a linear correlation between an increase in absorbance and an increase in cell number. The absorbance values obtained were converted into estimated cell numbers and the change (%) relative to the initial seeding was calculated.
  • The Effect of Serum. The addition of serum to media at normal oxygen conditions resulted in a reproducible dose-dependent increase in absorbance and thus the viable cell number. The addition of serum to complete MSCGM resulted in a dose-dependent decrease in absorbance. In the media without added serum, cells only grew appreciably in CELLGRO-FREE, Ham's FIO and DMEM.
  • The Effect of Oxygen. Reduced oxygen appeared to increase the growth rate of cells in Growth Medium, Ham's F10, and MSCGM. In decreasing order of growth, the media resulting in the best growth of the cells were Growth Medium>MSCGM>Iscove's+10% FBS=DMEM-H+10% FBS=Ham's F12+10% FBS=RPMI 1640+10% FBS.
  • Summary. Placenta-derived cells may be grown in a variety of culture media in normal or low oxygen. Short term growth of placenta-derived cells was determined in twelve basal media with 0, 2 and 10% (v/v) serum in 5% or atmospheric oxygen. In general, placenta-derived cells did not grow as well in serum-free conditions with the exception of Ham's F10 and CELLGRO-Free, which are also protein-free. Growth in these serum-free media was about 25-33% of the maximal growth observed with media containing 15% serum.
  • EXAMPLE 4 Growth of Postpartum-Derived Cells in Medium Containing D-Valine
  • It has been reported that medium containing D-valine instead of the normal L-valine isoform can be used to selectively inhibit the growth of fibroblast-like cells in culture (Hongpaisan, 2000; Sordillo et al., 1988). It was not previously known whether postpartum-derived cells could grow in medium containing D-valine.
  • Methods & Materials
  • Placenta-derived cells (P3), fibroblasts (P9) and umbilical-derived cells (P5) were seeded at 5×103 cells/cm2 in gelatin-coated T75 flasks (Corning, Corning, N.Y.). After 24 hours the medium was removed and the cells were washed with phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) to remove residual medium. The medium was replaced with a Modified Growth Medium (DMEM with D-valine (special order Gibco), 15% (v/v) dialyzed fetal bovine serum (Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma), penicillin/streptomycin (Gibco)).
  • Results
  • Placenta-derived, umbilical-derived, and fibroblast cells seeded in the D-valine-containing medium did not proliferate, unlike cells seeded in Growth Medium containing dialyzed serum. Fibroblasts cells changed morphologically, increasing in size and changing shape. All of the cells died and eventually detached from the flask surface after 4 weeks. These results indicate that medium containing D-valine is not suitable for selectively growing postpartum-derived cells.
  • References for Example 4
    • 1) Hongpaisan J. 2000. Cell Biol Int. 24:1-7.
    • 2) Sordillo L M, Oliver S P, Akers R M. 1988). Cell Biol Int Rep. 12:355-64.
    EXAMPLE 5 Cryopreservation Media for Placenta-Derived Cells
  • Cryopreservation media for the cryopreservation of placenta-derived cells were evaluated.
  • Methods & Materials
  • Placenta-derived cells grown in Growth Medium in a gelatin-coated T75 flask were washed with PBS and trypsinized using 1 milliliter Trypsin/EDTA (Gibco). The trypsinization was stopped by adding 10 milliliters Growth Medium. The cells were centrifuged at 150×g, supernatant removed, and the cell pellet was resuspended in 1 milliliter Growth Medium. An aliquot of cell suspension, 60 microliters, was removed and added to 60 microliters trypan blue (Sigma). The viable cell number was estimated using a hemocytometer. The cell suspension was divided into four equal aliquots each containing 88×104 cells each. The cell suspension was centrifuged and resuspended in 1 milliliter of each media below and transferred into Cryovials (Nalgene).
  • 1.) Growth Medium+10% (v/v) DMSO (Hybrimax, Sigma, St. Louis, Mo.)
  • 2.) Cell Freezing medium w/DMSO, w/methyl cellulose, serum-free (C6295, Sigma, St. Louis, Mo.)
  • 3.) Cell Freezing medium serum-free (C2639, Sigma, St. Louis, Mo.)
  • 4.) Cell Freezing Medium w/glycerol (C6039, Sigma, St. Louis, Mo.)
  • The cells were cooled at approximately −1° C./min overnight in a −80° C. freezer using a “Mr Frosty” freezing container according to the manufacturer's instructions (Nalgene, Rochester, N.Y.). Vials of cells were transferred into liquid nitrogen for 2 days before thawing rapidly in a 37° C. water bath. The cells were added to 10 milliliters Growth Medium and centrifuged before the cell number and viability was estimated. Cells were seeded onto gelatin-coated flasks at 5,000 cells/cm2 to determine whether the cells would attach and proliferate.
  • Results
  • The initial viability of the cells to be cryopreserved was assessed by trypan blue staining to be 100%. The initial viability of the cells to be cryopreserved was assessed by trypan blue staining to be 100%.
  • There was a commensurate reduction in cell number with viability for C6295 due to cells lysis. The viable cells cryopreserved in all four solutions attached, divided, and produced a confluent monolayer within 3 days. There was no discernable difference in estimated growth rate.
  • Summary. The cryopreservation of cells is one procedure available for preparation of a cell bank or a cell product. Four cryopreservation mixtures were compared for their ability to protect human placenta-derived cells from freezing damage. Dulbecco's modified Eagle's medium (DMEM) and 10% (v/v) dimethylsulfoxide (DMSO) is the preferred medium of those compared for cryopreservation of placenta-derived cells.
  • EXAMPLE 6 Karyotype Analysis of Postpartum-Derived Cells
  • Cell lines used in cell therapy are preferably homogeneous and free from any contaminating cell type. Cells used in cell therapy should have a normal chromosome number (46) and structure. To identify placenta- and umbilicus-derived cell lines that are homogeneous and free from cells of non-postpartum tissue origin, karyotypes of cell samples were analyzed.
  • Materials and Methods
  • PPDCs from postpartum tissue of a male neonate were cultured in Growth Medium containing penicillin/streptomycin. Postpartum tissue from a male neonate (X,Y) was selected to allow distinction between neonatal-derived cells and maternal derived cells (X,X). Cells were seeded at 5,000 cells per square centimeter in Growth Medium in a T25 flask (Corning, Corning, N.Y.) and expanded to 80% confluence. A T25 flask containing cells was filled to the neck with Growth Medium. Samples were delivered to a clinical cytogenetics laboratory by courier (estimated lab to lab transport time is one hour). Cells were analyzed during metaphase when the chromosomes are best visualized. Of twenty cells in metaphase counted, five were analyzed for normal homogeneous karyotype number (two). A cell sample was characterized as homogeneous if two karyotypes were observed. A cell sample was characterized as heterogeneous if more than two karyotypes were observed. Additional metaphase cells were counted and analyzed when a heterogeneous karyotype number (four) was identified.
  • Results
  • All cell samples sent for chromosome analysis were interpreted as exhibiting a normal appearance. Three of the sixteen cell lines analyzed exhibited a heterogeneous phenotype (XX and XY) indicating the presence of cells derived from both neonatal and maternal origins (Table 6-1). Cells derived from tissue Placenta-N were isolated from the neonatal aspect of placenta. At passage zero, this cell line appeared homogeneous XY. However, at passage nine, the cell line was heterogeneous (XX/XY), indicating a previously undetected presence of cells of maternal origin.
    TABLE 6-1
    Results of PPDC karyotype analysis
    Tissue passage Metaphase cells counted Metaphase cells analyzed
    Figure US20060234376A1-20061019-P00899
    mber of karyotype
    ISCN Karyotype
    Placenta 22 20 5 2 46, XX
    Umbilical 23 20 5 2 46, XX
    Umbilical 6 20 5 2 46, XY
    Placenta 2 20 5 2 46, XX
    Umbilical 3 20 5 2 46, XX
    Placenta-N 0 20 5 2 46, XY
    Placenta-V 0 20 5 2 46, XY
    Placenta-M 0 21 5 4 46, XY[18]/46, XX[3]
    Placenta-M 4 20 5 2 46, XX
    Placenta-N 9 25 5 4 46, XY[5]/46, XX[20]
    Placenta-N C1 1 20 5 2 46, XY
    Placenta-N C3 1 20 6 4 46, XY[2]/46, XX[18]
    Placenta-N C4 1 20 5 2 46, XY
    Placenta-N C15 1 20 5 2 46, XY
    Placenta-N C20 1 20 5 2 46, XY

    Key:

    N—Neonatal aspect;

    V—villous region;

    M—maternal aspect;

    C—clone
  • Summary. Chromosome analysis identified placenta- and umbilicus-derived cells whose karyotypes appeared normal as interpreted by a clinical cytogenetic laboratory. Karyotype analysis also identified cell lines free from maternal cells, as determined by homogeneous karyotype.
  • EXAMPLE 7 Evaluation of Human Postpartum-Derived Cell Surface Markers by Flow Cytometry
  • Characterization of cell surface proteins or “markers” by flow cytometry can be used to determine a cell line's identity. The consistency of expression can be determined from multiple donors, and in cells exposed to different processing and culturing conditions. Postpartum-derived cell (PPDC) lines isolated from the placenta and umbilicus were characterized (by flow cytometry), providing a profile for the identification of these cell lines.
  • Materials and Methods
  • Media and culture vessels. Cells were cultured in Growth Medium (Gibco Carlsbad, Calif.) with penicillin/streptomycin. Cells were cultured in plasma-treated T75, T150, and T225 tissue culture flasks (Corning, Corning, N.Y.) until confluent. The growth surfaces of the flasks were coated with gelatin by incubating 2% (w/v) gelatin (Sigma, St. Louis, Mo.) for 20 minutes at room temperature.
  • Antibody Staining and flow cytometry analysis. Adherent cells in flasks were washed in PBS and detached with Trypsin/EDTA. Cells were harvested, centrifuged, and resuspended in 3% (v/v) FBS in PBS at a cell concentration of 1×107 per milliliter. In accordance to the manufacture's specifications, antibody to the cell surface marker of interest (see below) was added to one hundred microliters of cell suspension and the mixture was incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were resuspended in 500 microliter PBS and analyzed by flow cytometry. Flow cytometry analysis was performed with a FACScalibur instrument (Becton Dickinson, San Jose, Calif.).
  • The following antibodies to cell surface markers were used.
    Antibody Manufacture Catalog Number
    CD10 BD Pharmingen (San Diego, CA) 555375
    CD13 BD Pharmingen 555394
    CD31 BD Pharmingen 555446
    CD34 BD Pharmingen 555821
    CD44 BD Pharmingen 555478
    CD45RA BD Pharmingen 555489
    CD73 BD Pharmingen 550257
    CD90 BD Pharmingen 555596
    CD117 BD Pharmingen 340529
    CD141 BD Pharmingen 559781
    PDGFr-alpha BD Pharmingen 556002
    HLA-A, B, C BD Pharmingen 555553
    HLA-DR, DP, BD Pharmingen 555558
    DQ
    IgG-FITC Sigma (St. Louis, MO) F-6522
    IgG-PE Sigma P-4685
  • Placenta and umbilicus comparison. Placenta-derived cells were compared to umbilicus-derive cells at passage 8.
  • Passage to passage comparison. Placenta- and umbilicus-derived cells were analyzed at passages 8, 15, and 20.
  • Donor to donor comparison. To compare differences among donors, placenta-derived cells from different donors were compared to each other, and umbilicus-derived cells from different donors were compared to each other.
  • Surface coating comparison. Placenta-derived cells cultured on gelatin-coated flasks was compared to placenta-derived cells cultured on uncoated flasks. Umbilicus-derived cells cultured on gelatin-coated flasks was compared to umbilicus-derived cells cultured on uncoated flasks.
  • Digestion enzyme comparison. Four treatments used for isolation and preparation of cells were compared. Cells isolated from placenta by treatment with 1) collagenase; 2) collagenase/dispase; 3) collagenase/hyaluronidase; and 4) collagenase/hyaluronidase/dispase were compared.
  • Placental layer comparison. Cells derived from the maternal aspect of placental tissue were compared to cells derived from the villous region of placental tissue and cells derived from the neonatal fetal aspect of placenta.
  • Results
  • Placenta vs. umbilicus comparison. Placenta- and umbilicus-derived cells analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for detectable expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values comparable to the IgG control. Variations in fluorescence values of positive curves were accounted for. The mean (i.e. CD 13) and range (i.e. CD90) of the positive curves showed some variation, but the curves appeared normal, confirming a homogenous population. Both curves individually exhibited values greater than the IgG control.
  • Passage to passage comparison—placenta-derived cells. Placenta-derived cells at passages 8, 15, and 20 analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, as reflected in the increased value of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ having fluorescence values consistent with the IgG control.
  • Passage to passage comparison—umbilicus-derived cells. Umbilicus-derived cells at passage 8, 15, and 20 analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, indicated by increased fluorescence relative to the IgG control. These cells were negative for CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, indicated by fluorescence values consistent with the IgG control.
  • Donor to donor comparison—placenta-derived cells. Placenta-derived cells isolated from separate donors analyzed by flow cytometry each expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. The cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence value consistent with the IgG control.
  • Donor to donor comparison—umbilicus derived cells. Umbilicus-derived cells isolated from separate donors analyzed by flow cytometry each showed positive expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ with fluorescence values consistent with the IgG control.
  • The effect of surface coating with gelatin on placenta-derived cells. Placenta-derived cells expanded on either gelatin-coated or uncoated flasks analyzed by flow cytometry all expressed of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, reflected in the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ indicated by fluorescence values consistent with the IgG control.
  • The effect of surface coating with gelatin on umbilicus-derived cells. Umbilicus-derived cells expanded on gelatin and uncoated flasks analyzed by flow cytometry all were positive for expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, with increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ, with fluorescence values consistent with the IgG control.
  • Effect of enzyme digestion procedure used for preparation of the cells on the cell surface marker profile. Placenta-derived cells isolated using various digestion enzymes analyzed by flow cytometry all expressed CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased values of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control
  • Placental layer comparison. Cells isolated from the maternal, villous, and neonatal layers of the placenta, respectively, analyzed by flow cytometry showed positive expression of CD10, CD13, CD44, CD73, CD 90, PDGFr-alpha and HLA-A, B, C, as indicated by the increased value of fluorescence relative to the IgG control. These cells were negative for expression of CD31, CD34, CD45, CD117, CD141, and HLA-DR, DP, DQ as indicated by fluorescence values consistent with the IgG control.
  • Summary. Analysis of placenta- and umbilicus-derived cells by flow cytometry has established of an identity of these cell lines. Placenta- and umbilicus-derived cells are positive for CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, HLA-A,B,C and negative for CD31, CD34, CD45, CD117, CD141 and HLA-DR, DP, DQ. This identity was consistent between variations in variables including the donor, passage, culture vessel surface coating, digestion enzymes, and placental layer. Some variation in individual fluorescence value histogram curve means and ranges was observed, but all positive curves under all conditions tested were normal and expressed fluorescence values greater than the IgG control, thus confirming that the cells comprise a homogenous population that has positive expression of the markers.
  • EXAMPLE 8 Immunohistochemical Characterization of Postpartum Tissue Phenotypes
  • The phenotypes of cells found within human postpartum tissues, namely umbilical cord and placenta, was analyzed by immunohistochemistry.
  • Materials & Methods
  • Tissue Preparation. Human umbilical cord and placenta tissue was harvested and immersion fixed in 4% (w/v) paraformaldehyde overnight at 4° C. Immunohistochemistry was performed using antibodies directed against the following epitopes: vimentin (1:500; Sigma, St. Louis, Mo.), desmin (1:150, raised against rabbit; Sigma; or 1:300, raised against mouse; Chemicon, Temecula, CA), alpha-smooth muscle actin (SMA; 1:400; Sigma), cytokeratin 18 (CK18; 1:400; Sigma), von Willebrand Factor (vWF; 1:200; Sigma), and CD34 (human CD34 Class III; 1:100; DAKOCytomation, Carpinteria, CA). In addition, the following markers were tested: anti-human GROalpha—PE (1:100; Becton Dickinson, Franklin Lakes, N.J.), anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA), anti-human oxidized LDL receptor 1 (ox-LDL R1; 1:100; Santa Cruz Biotech), and anti-human NOGO-A (1:100; Santa Cruz Biotech). Fixed specimens were trimmed with a scalpel and placed within OCT embedding compound (Tissue-Tek OCT; Sakura, Torrance, Calif.) on a dry ice bath containing ethanol. Frozen blocks were then sectioned (10 μm thick) using a standard cryostat (Leica Microsystems) and mounted onto glass slides for staining.
  • Immunohistochemistry. Immunohistochemistry was performed similar to previous studies (e.g., Messina, et al., 2003, Exper. Neurol. 184: 816-829). Tissue sections were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 1 hour to access intracellular antigens. In instances where the epitope of interest would be located on the cell surface (CD34, ox-LDL R1), Triton was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL R1, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout the procedure. Primary antibodies, diluted in blocking solution, were then applied to the sections for a period of 4 hours at room temperature. Primary antibody solutions were removed, and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG—Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and/or goat anti-rabbit IgG—Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG—FITC (1:150; Santa Cruz Biotech). Cultures were washed, and 10 micromolar DAPI (Molecular Probes) was applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). Positive staining was represented by fluorescence signal above control staining. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Results
  • Umbilical cord characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 markers were expressed in a subset of the cells found within umbilical cord. In particular, vWF and CD34 expression were restricted to blood vessels contained within the cord. CD34+cells were on the innermost layer (lumen side). Vimentin expression was found throughout the matrix and blood vessels of the cord. SMA was limited to the matrix and outer walls of the artery & vein, but not contained with the vessels themselves. CK18 and desmin were observed within the vessels only, desmin being restricted to the middle and outer layers.
  • Placenta characterization. Vimentin, desmin, SMA, CK18, vWF, and CD34 were all observed within the placenta and regionally specific.
  • GROalpha, GCP-2, ox-LDL R1, and NOGO-A Tissue Expression. None of these markers were observed within umbilical cord or placental tissue.
  • Summary. Vimentin, desmin, alpha-smooth muscle actin, cytokeratin 18, von Willebrand Factor, and CD 34 are expressed in cells within human umbilical cord and placenta.
  • EXAMPLE 9 Analysis of Postpartum Tissue-Derived Cells using Oligonucleotide Arrays
  • Affymetrix GENECHIP arrays were used to compare gene expression profiles of umbilicus- and placenta-derived cells with fibroblasts, human mesenchymal stem cells, and another cell line derived from human bone marrow. This analysis provided a characterization of the postpartum-derived cells and identified unique molecular markers for these cells.
  • Materials and Methods
  • Isolation and culture of cells. Human umbilical cords and placenta were obtained from National Disease Research Interchange (NDR1, Philadelphia, Pa.) from normal full term deliveries with patient consent. The tissues were received and cells were isolated as described in Example 1. Cells were cultured in Growth Medium (using DMEM-LG) on gelatin-coated tissue culture plastic flasks. The cultures were incubated at 37° C. with 5% CO2.
  • Human dermal fibroblasts were purchased from Cambrex Incorporated (Walkersville, MD; Lot number 9F0844) and ATCC CRL-1501 (CCD39SK). Both lines were cultured in DMEM/F 12 medium (Invitrogen, Carlsbad, Calif.) with 10% (v/v) fetal bovine serum (Hyclone) and penicillin/streptomycin (Invitrogen). The cells were grown on standard tissue-treated plastic.
  • Human mesenchymal stem cells (hMSC) were purchased from Cambrex Incorporated (Walkersville, MD; Lot numbers 2F1655, 2F1656 and 2F1657) and cultured according to the manufacturer's specifications in MSCGM Media (Cambrex). The cells were grown on standard tissue cultured plastic at 37-C with 5% CO2.
  • Human ileac crest bone marrow was received from NDR1 with patient consent. The marrow was processed according to the method outlined by Ho, et al. (WO03/025149). The marrow was mixed with lysis buffer (155 mM NH4Cl, 10 mM KHCO3, and 0.1 mM EDTA, pH 7.2) at a ratio of 1 part bone marrow to 20 parts lysis buffer. The cell suspension was vortexed, incubated for 2 minutes at ambient temperature, and centrifuged for 10 minutes at 500×g. The supernatant was discarded and the cell pellet was resuspended in Minimal Essential Medium-alpha (Invitrogen) supplemented with 10% (v/v) fetal bovine serum and 4 mM glutamine. The cells were centrifuged again and the cell pellet was resuspended in fresh medium. The viable mononuclear cells were counted using trypan-blue exclusion (Sigma, St. Louis, Mo.). The mononuclear cells were seeded in tissue-cultured plastic flasks at 5×104 cells/cm2. The cells were incubated at 37° C. with 5% CO2 at either standard atmospheric O2 or at 5% O2. Cells were cultured for 5 days without a media change. Media and non-adherent cells were removed after 5 days of culture. The adherent cells were maintained in culture.
  • Isolation of mRNA and GENECHIP Analysis. Actively growing cultures of cells were removed from the flasks with a cell scraper in cold PBS. The cells were centrifuged for 5 minutes at 300×g. The supernatant was removed and the cells were resuspended in fresh PBS and centrifuged again. The supernatant was removed and the cell pellet was immediately frozen and stored at −80° C. Cellular mRNA was extracted and transcribed into cDNA, which was then transcribed into cRNA and biotin-labeled. The biotin-labeled cRNA was hybridized with HG-U133A GENECHIP oligonucleotide array (Affymetrix, Santa Clara Calif.). The hybridization and data collection was performed according to the manufacturer's specifications. Analyses were performed using “Significance Analysis of Microarrays” (SAM) version 1.21 computer software (Stanford University, www-stat.stanford.edu/-tibs/SAM; Tusher, V. G. et al., 2001, Proc. Natl. Acad. Sci. USA 98: 5116-5121).
  • Results
  • Fourteen different populations of cells were analyzed. The cells along with passage information, culture substrate, and culture media are listed in Table 9-1.
    TABLE 9-1
    Cells analyzed by the microarray study. Cell lines are listed by
    identification code along with passage at time of analysis, cell
    growth substrate and growth medium.
    Cell Population Passage Substrate Medium
    Umbilicus (022803) 2 Gelatin DMEM, 15% FBS, 2-ME
    Umbilicus (042103) 3 Gelatin DMEM, 15% FBS, 2-ME
    Umbilicus (071003) 4 Gelatin DMEM, 15% FBS, 2-ME
    Placenta (042203) 12 Gelatin DMEM, 15% FBS, 2-ME
    Placenta (042903) 4 Gelatin DMEM, 15% FBS, 2-ME
    Placenta (071003) 3 Gelatin DMEM, 15% FBS, 2-ME
    ICBM (070203) 3 Plastic MEM, 10% FBS
    (5% 02)
    ICBM (062703) 5 Plastic MEM, 10% FBS
    (std. O2)
    ICBM (062703) 5 Plastic MEM, 10% FBS
    (5% 02)
    hMSC (Lot 2F1655) 3 Plastic MSCGM
    hMSC (Lot 2F1656) 3 Plastic MSCGM
    hMSC (Lot 2F 1657) 3 Plastic MSCGM
    hFibroblast (9F0844) 9 Plastic DMEM-F12, 10% FBS
    hFibroblast 4 Plastic DMEM-F12, 10% FBS
    (CCD39SK)
  • The data were evaluated by a Principle Component Analysis, analyzing the 290 genes that were differentially expressed in the cells. This analysis allows for a relative comparison for the similarities between the populations. Table 9-2 shows the Euclidean distances that were calculated for the comparison of the cell pairs. The Euclidean distances were based on the comparison of the cells based on the 290 genes that were differentially expressed among the cell types. The Euclidean distance is inversely proportional to similarity between the expression of the 290 genes (i.e., the greater the distance, the less similarity exists).
    TABLE 9-2
    The Euclidean Distances for the Cell Pairs.
    Cell Pair Euclidean Distance
    ICBM-hMSC 24.71
    Placenta-umbilical 25.52
    ICBM-Fibroblast 36.44
    ICBM-placenta 37.09
    Fibroblast-MSC 39.63
    ICBM-Umbilical 40.15
    Fibroblast-Umbilical 41.59
    MSC-Placenta 42.84
    MSC-Umbilical 46.86
    ICBM-placenta 48.41
  • Tables 9-3, 9-4, and 9-5 show the expression of genes increased in placenta-derived cells (Table 9-3), increased in umbilicus-derived cells (Table 9-4), and reduced in umbilicus- and placenta-derived cells (Table 9-5). The column entitled “Probe Set ID” refers to the manufacturer's identification code for the sets of several oligonucleotide probes located on a particular site on the chip, which hybridize to the named gene (column “Gene Name”), comprising a sequence that can be found within the NCBI (GenBank) database at the specified accession number (column “NCBI Accession Number”).
    TABLE 9-3
    Genes shown to have specifically increased expression in the placenta-derived cells
    as compared to other cell lines assayed
    Genes Increased in Placenta-Derived Cells
    NCBI Accession
    Probe Set ID Gene Name Number
    209732_at C-type (calcium dependent, carbohydrate-recognition domain) AF070642
    lectin, superfamily member 2 (activation-induced)
    206067_s_at Wilms tumor 1 NM_024426
    207016_s_at aldehyde dehydrogenase 1 family, member A2 AB015228
    206367_at renin NM_000537
    210004_at oxidized low density lipoprotein (lectin-like) receptor 1 AF035776
    214993_at Homo sapiens, clone IMAGE: 4179671, mRNA, partial cds AF070642
    202178_at protein kinase C, zeta NM_002744
    209780_at hypothetical protein DKFZp564F013 AL136883
    204135_at downregulated in ovarian cancer 1 NM_014890
    213542_at Homo sapiens mRNA; cDNA DKFZp547K1113 (from clone Al246730
    DKFZp547K1113)
  • TABLE 9-4
    Genes shown to have specifically increased expression in the
    umbilicus-derived cells as compared to other cell lines assayed
    Genes Increased in Umbilicus-Derived Cells
    NCBI Accession
    Probe Set ID Gene Name Number
    202859_x_at interleukin 8 NM_000584
    211506_s_at interleukin 8 AF043337
    210222_s_at reticulon 1 BC000314
    204470_at chemokine (C—X—C motif) ligand 1 NM_001511
    (melanoma growth stimulating
    activity
    206336_at chemokine (C—X—C motif) ligand 6 NM_002993
    (granulocyte chemotactic protein 2)
    207850_at chemokine (C—X—C motif) ligand 3 NM_002090
    203485_at reticulon 1 NM_021136
    202644_s_at tumor necrosis factor, alpha-induced NM_006290
    protein 3
  • TABLE 9-5
    Genes shown to have decreased expression in umbilicus- and placenta-derived cells
    as compared to other cell lines assayed
    Genes Decreased in Umbilicus- and Placenta-Derived Cells
    NCBI Accession
    Probe Set ID Gene name Number
    210135_s_at short stature homeobox 2 AF022654.1
    205824_at heat shock 27 kDa protein 2 NM_001541.1
    209687_at chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1) U19495.1
    203666_at chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1) NM_000609.1
    212670_at elastin (supravalvular aortic stenosis, Williams-Beuren AA479278
    syndrome)
    213381_at Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone N91149
    DKFZp586M2022)
    206201_s_at mesenchyme homeo box 2 (growth arrest-specific homeo box) NM_005924.1
    205817_at sine oculis homeobox homolog 1 (Drosophila) NM_005982.1
    209283_at crystallin, alpha B AF007162.1
    212793_at dishevelled associated activator of morphogenesis 2 BF513244
    213488_at DKFZP586B2420 protein AL050143.1
    209763_at similar to neuralin 1 AL049176
    205200_at tetranectin (plasminogen binding protein) NM_003278.1
    205743_at src homology three (SH3) and cysteine rich domain NM_003149.1
    200921_s_at B-cell translocation gene 1, anti-proliferative NM_001731.1
    206932_at cholesterol 25-hydroxylase NM_003956.1
    204198_s_at runt-related transcription factor 3 AA541630
    219747_at hypothetical protein FLJ23191 NM_024574.1
    204773_at interleukin 11 receptor, alpha NM_004512.1
    202465_at procollagen C-endopeptidase enhancer NM_002593.2
    203706_s_at frizzled homolog 7 (Drosophila) NM_003507.1
    212736_at hypothetical gene BC008967 BE299456
    214587_at collagen, type VIII, alpha 1 BE877796
    201645_at tenascin C (hexabrachion) NM_002160.1
    210239_at iroquois homeobox protein 5 U90304.1
    203903_s_at Hephaestin NM_014799.1
    205816_at integrin, beta 8 NM_002214.1
    203069_at synaptic vesicle glycoprotein 2 NM_014849.1
    213909_at Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744 AU147799
    206315_at cytokine receptor-like factor 1 NM_004750.1
    204401_at potassium intermediate/small conductance calcium-activated NM_002250.1
    channel, subfamily N, member 4
    216331_at integrin, alpha 7 AK022548.1
    209663_s_at integrin, alpha 7 AF072132.1
    213125_at DKFZP586L151 protein AW007573
    202133_at transcriptional co-activator with PDZ-binding motif (TAZ) AA081084
    206511_s_at sine oculis homeobox homolog 2 (Drosophila) NM_016932.1
    213435_at KIAA1034 protein AB028957.1
    206115_at early growth response 3 NM_004430.1
    213707_s_at distal-less homeo box 5 NM_005221.3
    218181_s_at hypothetical protein FLJ20373 NM_017792.1
    209160_at aldo-keto reductase family 1, member C3 (3-alpha AB018580.1
    hydroxysteroid dehydrogenase, type II)
    213905_x_at Biglycan AA845258
    201261_x_at Biglycan BC002416.1
    202132_at transcriptional co-activator with PDZ-binding motif (TAZ) AA081084
    214701_s_at fibronectin 1 AJ276395.1
    213791_at Proenkephalin NM_006211.1
    205422_s_at integrin, beta-like 1 (with EGF-like repeat domains) NM_004791.1
    214927_at Homo sapiens mRNA full length insert cDNA clone AL359052.1
    EUROIMAGE 1968422
    206070_s_at EphA3 AF213459.1
    212805_at KIAA0367 protein AB002365.1
    219789_at natriuretic peptide receptor C/guanylate cyclase C AI628360
    (atrionatriuretic peptide receptor C)
    219054_at hypothetical protein FLJ14054 NM_024563.1
    213429_at Homo sapiens mRNA; cDNA DKFZp564B222 (from clone AW025579
    DKFZp564B222)
    204929_s_at vesicle-associated membrane protein 5 (myobrevin) NM_006634.1
    201843_s_at EGF-containing fibulin-like extracellular matrix protein 1 NM_004105.2
    221478_at BCL2/adenovirus E1B 19 kDa interacting protein 3-like AL132665.1
    201792_at AE binding protein 1 NM_001129.2
    204570_at cytochrome c oxidase subunit VIIa polypeptide 1 (muscle) NM_001864.1
    201621_at neuroblastoma, suppression of tumorigenicity 1 NM_005380.1
    202718_at insulin-like growth factor binding protein 2, 36 kDa NM_000597.1
  • Tables 9-6, 9-7, and 9-8 show the expression of genes increased in human fibroblasts (Table 9-6), ICBM cells (Table 9-7), and MSCs (Table 9-8).
    TABLE 9-6
    Genes that were shown to have increased expression in
    fibroblasts as compared to the other cell lines assayed.
    Genes increased in fibroblasts
    dual specificity phosphatase 2
    KIAA0527 protein
    Homo sapiens cDNA: FLJ23224 fis, clone ADSU02206
    dynein, cytoplasmic, intermediate polypeptide 1
    ankyrin 3, node of Ranvier (ankyrin G)
    inhibin, beta A (activin A, activin AB alpha polypeptide)
    ectonucleotide pyrophosphatase/phosphodiesterase 4 (putative function)
    KIAA1053 protein
    microtubule-associated protein 1A
    zinc finger protein 41
    HSPC019 protein
    Homo sapiens cDNA: FLJ23564 fis, clone LNG10773
    Homo sapiens mRNA; cDNA DKFZp564A072 (from clone
    DKFZp564A072)
    LIM protein (similar to rat protein kinase C-binding enigma)
    inhibitor of kappa light polypeptide gene enhancer in B-cells, kinase
    complex-associated protein
    hypothetical protein FLJ22004
    Human (clone CTG-A4) mRNA sequence
    ESTs, Moderately similar to cytokine receptor-like factor 2; cytokine
    receptor CRL2 precursor [Homo sapiens]
    transforming growth factor, beta 2
    hypothetical protein MGC29643
    antigen identified by monoclonal antibody MRC OX-2
    putative X-linked retinopathy protein
  • TABLE 9-7
    Genes that were shown to have increased expression in
    the ICBM-derived cells as compared to the other cell lines assayed.
    Genes Increased In ICBM Cells
    cardiac ankyrin repeat protein
    MHC class I region ORF
    integrin, alpha 10
    hypothetical protein FLJ22362
    UDP-N-acetyl-alpha-D-galactosamine:polypeptide
    N-acetylgalactosaminyltransferase 3 (GalNAc-T3)
    interferon-induced protein 44
    SRY (sex determining region Y)-box 9
    (campomelic dysplasia, autosomal sex-reversal)
    keratin associated protein 1-1
    hippocalcin-like 1
    jagged 1 (Alagille syndrome)
    proteoglycan 1, secretory granule
  • TABLE 9-8
    Genes that were shown to have increased expression in
    the MSC cells as compared to the other cell lines assayed.
    Genes Increased In MSC Cells
    interleukin 26
    maltase-glucoamylase (alpha-glucosidase)
    nuclear receptor subfamily 4, group A, member 2
    v-fos FBJ murine osteosarcoma viral oncogene homolog
    hypothetical protein DC42
    nuclear receptor subfamily 4, group A, member 2
    FBJ murine osteosarcoma viral oncogene homolog B
    WNT1 inducible signaling pathway protein 1
    MCF.2 cell line derived transforming sequence
    potassium channel, subfamily K, member 15
    cartilage paired-class homeoprotein 1
    Homo sapiens cDNA FLJ12232 fis, clone MAMMA1001206
    Homo sapiens cDNA FLJ34668 fis, clone LIVER2000775
    jun B proto-oncogene
    B-cell CLL/lymphoma 6 (zinc finger protein 51)
    zinc finger protein 36, C3H type, homolog (mouse)
  • Summary. The present examination was performed to provide a molecular characterization of the postpartum cells derived from umbilical cord and placenta. This analysis included cells derived from three different umbilical cords and three different placentas. The examination also included two different lines of dermal fibroblasts, three lines of mesenchymal stem cells, and three lines of ileac crest bone marrow cells. The mRNA that was expressed by these cells was analyzed using an oligonucleotide array that contained probes for 22,000 genes. Results showed that 290 genes are differentially expressed in these five different cell types. These genes include ten genes that are specifically increased in the placenta-derived cells and seven genes specifically increased in the umbilical cord-derived cells. Fifty-four genes were found to have specifically lower expression levels in placenta and umbilical cord, as compared with the other cell types. The expression of selected genes has been confirmed by PCR (see the example that follows). These results demonstrate that the postpartum-derived cells have a distinct gene expression profile, for example, as compared to bone marrow-derived cells and fibroblasts.
  • EXAMPLE 10 Cell Markers in Postpartum-Derived Cells
  • In the preceding example, similarities and differences in cells derived from the human placenta and the human umbilical cord were assessed by comparing their gene expression profiles with those of cells derived from other sources (using an oligonucleotide array). Six “signature” genes were identified: oxidized LDL receptor 1, interleukin-8, rennin, reticulon, chemokine receptor ligand 3 (CXC ligand 3), and granulocyte chemotactic protein 2 (GCP-2). These “signature” genes were expressed at relatively high levels in postpartum-derived cells.
  • The procedures described in this example were conducted to verify the microarray data and find concordance/discordance between gene and protein expression, as well as to establish a series of reliable assay for detection of unique identifiers for placenta- and umbilicus-derived cells.
  • Methods & Materials
  • Cells. Placenta-derived cells (three isolates, including one isolate predominately neonatal as identified by karyotyping analysis), umbilicus-derived cells (four isolates), and Normal Human Dermal Fibroblasts (NHDF; neonatal and adult) grown in Growth Medium with penicillin/streptomycin in a gelatin-coated T75 flask. Mesechymal Stem Cells (MSCs) were grown in Mesenchymal Stem Cell Growth Medium Bullet kit (MSCGM; Cambrex, Walkerville, MD).
  • For the IL-8 protocol, cells were thawed from liquid nitrogen and plated in gelatin-coated flasks at 5,000 cells/cm2, grown for 48 hours in Growth Medium and then grown for further 8 hours in 10 milliliters of serum starvation medium [DMEM-low glucose (Gibco, Carlsbad, Calif.), penicillin/streptomycin (Gibco, Carlsbad, Calif.) and 0.1% (w/v) Bovine Serum Albumin (BSA; Sigma, St. Louis, Mo.)]. After this treatment RNA was extracted and the supernatants were centrifuged at 150×g for 5 minutes to remove cellular debris. Supernatants were then frozen at −80° C. for ELISA analysis.
  • Cell culture for ELISA assay. Postpartum cells derived from placenta and umbilicus, as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium in gelatin-coated T75 flasks. Cells were frozen at passage 11 in liquid nitrogen. Cells were thawed and transferred to 15-milliliter centrifuge tubes. After centrifugation at 150×g for 5 minutes, the supernatant was discarded. Cells were resuspended in 4 milliliters culture medium and counted. Cells were grown in a 75 cm2 flask containing 15 milliliters of Growth Medium at 375,000 cell/flask for 24 hours. The medium was changed to a serum starvation medium for 8 hours. Serum starvation medium was collected at the end of incubation, centrifuged at 14,000×g for 5 minutes (and stored at −20° C.).
  • To estimate the number of cells in each flask, 2 milliliters of tyrpsin/EDTA (Gibco, Carlsbad, Calif.) was added each flask. After cells detached from the flask, trypsin activity was neutralized with 8 milliliters of Growth Medium. Cells were transferred to a 15 milliliters centrifuge tube and centrifuged at 150×g for 5 minutes. Supernatant was removed and 1 milliliter Growth Medium was added to each tube to resuspend the cells. Cell number was estimated using a hemocytometer.
  • ELISA assay. The amount of IL-8 secreted by the cells into serum starvation medium was analyzed using ELISA assays (R&D Systems, Minneapolis, Minn.). All assays were tested according to the instructions provided by the manufacturer.
  • Total RNA isolation. RNA was extracted from confluent postpartum-derived cells and fibroblasts or for IL-8 expression from cells treated as described above. Cells were lysed with 350 microliters buffer RLT containing beta-mercaptoethanol (Sigma, St. Louis, Mo.) according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, Calif.). RNA was extracted according to the manufacturer's instructions (RNeasy Mini Kit; Qiagen, Valencia, CA) and subjected to DNase treatment (2.7 U/sample) (Sigma St. Louis, Mo.). RNA was eluted with 50 microliters DEPC-treated water and stored at −80° C.
  • Reverse transcription. RNA was also extracted from human placenta and umbilicus. Tissue (30 milligram) was suspended in 700 microliters of buffer RLT containing 2-mercaptoethanol. Samples were mechanically homogenized and the RNA extraction proceeded according to manufacturer's specification. RNA was extracted with 50 microliters of DEPC-treated water and stored at −80° C. RNA was reversed transcribed using random hexamers with the TaqMan reverse transcription reagents (Applied Biosystems, Foster City, Calif.) at 25° C. for 10 minutes, 37° C. for 60 minutes, and 95° C. for 10 minutes. Samples were stored at −20° C.
  • Genes identified by cDNA microarray as uniquely regulated in postpartum cells (signature genes—including oxidized LDL receptor, interleukin-8, rennin and reticulon), were further investigated using real-time and conventional PCR.
  • Real-time PCR. PCR was performed on cDNA samples using Assays-on-Demand™ gene expression products: oxidized LDL receptor (Hs00234028); rennin (Hs00166915); reticulon (Hs00382515); CXC ligand 3 (Hs00171061); GCP-2 (Hs00605742); IL-8 (Hs100174103); and GAPDH (Applied Biosystems, Foster City, Calif.) were mixed with cDNA and TaqMan Universal PCR master mix according to the manufacturer's instructions (Applied Biosystems, Foster City, Calif.) using a 7000 sequence detection system with ABI Prism 7000 SDS software (Applied Biosystems, Foster City, Calif.). Thermal cycle conditions were initially 50° C. for 2 min and 95° C. for 10 min, followed by 40 cycles of 95° C. for 15 sec and 60° C. for 1 min. PCR data was analyzed according to manufacturer's specifications (User Bulletin #2 from Applied Biosystems for ABI Prism 7700 Sequence Detection System).
  • Conventional PCR. Conventional PCR was performed using an ABI PRISM 7700 (Perkin Elmer Applied Biosystems, Boston, Mass., USA) to confirm the results from real-time PCR. PCR was performed using 2 microliters of cDNA solution, 1× AmpliTaq Gold universal mix PCR reaction buffer (Applied Biosystems, Foster City, Calif.) and initial denaturation at 94° C. for 5 minutes. Amplification was optimized for each primer set. For IL-8, CXC ligand 3, and reticulon (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 30 cycles); for rennin (94° C. for 15 seconds, 53° C. for 15 seconds and 72° C. for 30 seconds for 38 cycles); for oxidized LDL receptor and GAPDH (94° C. for 15 seconds, 55° C. for 15 seconds and 72° C. for 30 seconds for 33 cycles). Primers used for amplification are listed in Table 1. Primer concentration in the final PCR reaction was 1 micromolar except for GAPDH, which was 0.5 micromolar. GAPDH primers were the same as real-time PCR, except that the manufacturer's TaqMan probe was not added to the final PCR reaction. Samples were run on 2% (w/v) agarose gel and stained with ethidium bromide (Sigma, St. Louis, Mo.). Images were captured using a 667 Universal Twinpack film (VWR International, South Plainfield, NJ) using a focal-length Polaroid camera (VWR International, South Plainfield, NJ).
    TABLE 10-1
    Primers used
    Primer name Primers
    Oxidized LDL S: 5′-GAGAAATCCAAAGAGCAAATGG-3′ (SEQ ID NO:1)
    receptor A: 5′-AGAATGGAAAACTGGAATAGG-3′ (SEQ ID NO:2)
    Renin S: 5′-TCTTCGATGCTTCGGATTCC-3′ (SEQ ID NO:3)
    A: 5′-GAATTCTCGGAATCTCTGTTG-3′ (SEQ ID NO:4)
    Reticulon S: 5′-TTACAAGCAGTGCAGAAAACC-3′ (SEQ ID NO:5)
    A: 5′-AGTAAACATTGAAACCACAGCC-3′ (SEQ ID NO:6)
    Interleukin-8 S: 5′-TCTGCAGCTCTGTGTGAAGG-3′ (SEQ ID NO:7)
    A: 5′-CTTCAAAAACTTCTCCACAACC-3′ (SEQ ID NO:8)
    Chemokine S: 5′-CCCACGCCACGCTCTCC-3′ (SEQ ID NO:9)
    (CXC) ligand 3 A: 5′-TCCTGTCAGTTGGTGCTCC-3′ (SEQ ID NO:10)
  • Immunofluorescence. PPDCs were fixed with cold 4% (w/v) paraformaldehyde (Sigma-Aldrich, St. Louis, Mo.) for 10 minutes at room temperature. One isolate each of umbilicus- and placenta-derived cells at passage 0 (P0) (directly after isolation) and passage 11 (P11) (two isolates of placenta-derived, two isolates of umbilicus-derived cells) and fibroblasts (P11) were used. Immunocytochemistry was performed using antibodies directed against the following epitopes: vimentin (1:500, Sigma, St. Louis, Mo.), desmin (1:150; Sigma—raised against rabbit; or 1:300; Chemicon, Temecula, Calif.—raised against mouse,), alpha-smooth muscle actin (SMA; 1:400; Sigma), cytokeratin 18 (CK18; 1:400; Sigma), von Willebrand Factor (vWF; 1:200; Sigma), and CD34 (human CD34 Class III; 1:100; DAKOCytomation, Carpinteria, CA). In addition, the following markers were tested on passage 11 postpartum cells: anti-human GRO alpha—PE (1:100; Becton Dickinson, Franklin Lakes, N.J.), anti-human GCP-2 (1:100; Santa Cruz Biotech, Santa Cruz, CA), anti-human oxidized LDL receptor 1 (ox-LDL R1; 1:100; Santa Cruz Biotech), and anti-human NOGA-A (1:100; Santa Cruz, Biotech).
  • Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma, St. Louis, Mo.) for 30 minutes to access intracellular antigens. Where the epitope of interest was located on the cell surface (CD34, ox-LDL R1), Triton X-100 was omitted in all steps of the procedure in order to prevent epitope loss. Furthermore, in instances where the primary antibody was raised against goat (GCP-2, ox-LDL R1, NOGO-A), 3% (v/v) donkey serum was used in place of goat serum throughout. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. The primary antibody solutions were removed and the cultures were washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG—Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and/or goat anti-rabbit IgG—Alexa 488 (1:250; Molecular Probes) or donkey anti-goat IgG—FITC (1:150, Santa Cruz Biotech). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using an appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Preparation of cells for FACS analysis. Adherent cells in flasks were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) and detached with Trypsin/EDTA (Gibco, Carlsbad, Calif.). Cells were harvested, centrifuged, and re-suspended 3% (v/v) FBS in PBS at a cell concentration of 1×107 per milliliter. One hundred microliter aliquots were delivered to conical tubes. Cells stained for intracellular antigens were permeablized with Perm/Wash buffer (BD Pharmingen, San Diego, Calif.). Antibody was added to aliquots as per manufactures specifications and the cells were incubated for in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess antibody. Cells requiring a secondary antibody were resuspended in 100 microliters of 3% FBS. Secondary antibody was added as per manufactures specification and the cells were incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove excess secondary antibody. Washed cells were resuspended in 0.5 milliliters PBS and analyzed by flow cytometry. The following antibodies were used: oxidized LDL receptor 1 (sc-5813; Santa Cruz, Biotech), GROa (555042; BD Pharmingen, Bedford, Mass.), Mouse IgG1 kappa, (P-4685 and M-5284; Sigma), Donkey against Goat IgG (sc-3743; Santa Cruz, Biotech.). Flow cytometry analysis was performed with FACScalibur (Becton Dickinson San Jose, Calif.).
  • Results
  • Results of real-time PCR for selected “signature” genes performed on cDNA from cells derived from human placentae, adult and neonatal fibroblasts and Mesenchymal Stem Cells (MSCs) indicate that both oxidized LDL receptor and rennin were expressed at higher level in the placenta-derived cells as compared to other cells. The data obtained from real-time PCR were analyzed by the ΔΔCT method and expressed on a logarithmic scale. Levels of reticulon and oxidized LDL receptor expression were higher in umbilicus-derived cells as compared to other cells. No significant difference in the expression levels of CXC ligand 3 and GCP-2 were found between postpartum-derived cells and controls. The results of real-time PCR were confirmed by conventional PCR. Sequencing of PCR products further validated these observations. No significant difference in the expression level of CXC ligand 3 was found between postpartum-derived cells and controls using conventional PCR CXC ligand 3 primers listed above.
  • The production of the cytokine, IL-8 in postpartum was elevated in both Growth Medium-cultured and serum-starved postpartum-derived cells. All real-time PCR data was validated with conventional PCR and by sequencing PCR products.
  • When supernatants of cells grown in serum-free medium were examined for the presence of IL-8, the highest amounts were detected in media derived from umbilical cells and some isolates of placenta cells (Table 10-1). No IL-8 was detected in medium derived from human dermal fibroblasts.
    TABLE 10-1
    IL-8 protein amount measured by ELISA
    Cell type IL-8
    hFibro ND
    Placenta Isolate 1 ND
    Umb Isolate 1 2058.42 ± 144.67 
    Placenta Isolate 2 ND
    Umb Isolate 2 2368.86 ± 22.73 
    Placenta Isolate3 (normal O2) 17.27 ± 8.63 
    Placenta Isolate 3 (lowO2, W/O 264.92 ± 9.88 
    BME)

    Results of the ELISA assay for interleukin-8 (IL-8) performed on placenta-and umbilicus-derived cells as well as human skin fibroblasts. Values are presented here are picograms/million cells, n = 2, sem.

    ND: Not Detected
  • Placenta-derived cells were also examined for the production of oxidized LDL receptor, GCP-2 and GROalpha by FACS analysis. Cells tested positive for GCP-2. Oxidized LDL receptor and GRO were not detected by this method.
  • Placenta-derived cells were also tested for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells derived from the human placenta were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Cells stained positive for both alpha-smooth muscle actin and vimentin. This pattern was preserved through passage 11. Only a few cells (<5%) at passage 0 stained positive for cytokeratin 18.
  • Cells derived from the human umbilical cord at passage 0 were probed for the production of selected proteins by immunocytochemical analysis. Immediately after isolation (passage 0), cells were fixed with 4% paraformaldehyde and exposed to antibodies for six proteins: von Willebrand Factor, CD34, cytokeratin 18, desmin, alpha-smooth muscle actin, and vimentin. Umbilicus-derived cells were positive for alpha-smooth muscle actin and vimentin, with the staining pattern consistent through passage 11.
  • Summary. Concordance between gene expression levels measured by microarray and PCR (both real-time and conventional) has been established for four genes: oxidized LDL receptor 1, rennin, reticulon, and IL-8. The expression of these genes was differentially regulated at the mRNA level in PPDCs, with IL-8 also differentially regulated at the protein level. The presence of oxidized LDL receptor was not detected at the protein level by FACS analysis in cells derived from the placenta. Differential expression of GCP-2 and CXC ligand 3 was not confirmed at the mRNA level, however GCP-2 was detected at the protein level by FACS analysis in the placenta-derived cells. Although this result is not reflected by data originally obtained from the microarray experiment, this may be due to a difference in the sensitivity of the methodologies.
  • Immediately after isolation (passage 0), cells derived from the human placenta stained positive for both alpha-smooth muscle actin and vimentin. This pattern was also observed in cells at passage 11. These results suggest that vimentin and alpha-smooth muscle actin expression may be preserved in cells with passaging, in the Growth Medium and under the conditions utilized in these procedures. Cells derived from the human umbilical cord at passage 0 were probed for the expression of alpha-smooth muscle actin and vimentin, and were positive for both. The staining pattern was preserved through passage 11.
  • EXAMPLE 11 In Vitro Immunological Evaluation of Postpartum-Derived Cells
  • Postpartum-derived cells (PPDCs) were evaluated in vitro for their immunological characteristics in an effort to predict the immunological response, if any, these cells would elicit upon in vivo transplantation. PPDCs were assayed by flow cytometry for the presence of HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2. These proteins are expressed by antigen-presenting cells (APC) and are required for the direct stimulation of naïve CD4+ T cells (Abbas & Lichtman, CELLULAR AND MOLECULAR IMMUNOLOGY, 5th Ed. (2003) Saunders, Philadelphia, p. 171). The cell lines were also analyzed by flow cytometry for the expression of HLA-G (Abbas & Lichtman, 2003, supra), CD 178 (Coumans, et al., (1999) Journal of Immunological Methods 224, 185-196), and PD-L2 (Abbas & Lichtman, 2003, supra; Brown, et. al. (2003) The Journal of Immunology 170, 1257-1266). The expression of these proteins by cells residing in placental tissues is thought to mediate the immuno-privileged status of placental tissues in utero. To predict the extent to which placenta- and umbilicus-derived cell lines elicit an immune response in vivo, the cell lines were tested in a one-way mixed lymphocyte reaction (MLR).
  • Materials and Methods
  • Cell culture. Cells were cultured to confluence in Growth Medium containing penicillin/streptomycin in T75 flasks (Corning, Corning, N.Y.) coated with 2% gelatin (Sigma, St. Louis, Mo.).
  • Antibody Staining. Cells were washed in phosphate buffered saline (PBS) (Gibco, Carlsbad, Calif.) and detached with Trypsin/EDTA (Gibco, Carlsbad, MO). Cells were harvested, centrifuged, and re-suspended in 3% (v/v) FBS in PBS at a cell concentration of 1×107 per milliliter. Antibody (Table 11-1) was added to one hundred microliters of cell suspension as per manufacturer's specifications and incubated in the dark for 30 minutes at 4° C. After incubation, cells were washed with PBS and centrifuged to remove unbound antibody. Cells were re-suspended in five hundred microliters of PBS and analyzed by flow cytometry using a FACSCalibur instrument (Becton Dickinson, San Jose, Calif.).
    TABLE 11-1
    Antibodies
    Antibody Manufacturer Catalog Number
    HLA-DRDPDQ BD Pharmingen (San Diego, CA) 555558
    CD80 BD Pharmingen (San Diego, CA) 557227
    CD86 BD Pharmingen (San Diego, CA) 555665
    B7-H2 BD Pharmingen (San Diego, CA) 552502
    HLA-G Abcam (Cambridgeshire, UK) ab 7904-100
    CD 178 Santa Cruz (San Cruz, CA) sc-19681
    PD-L2 BD Pharmingen (San Diego, CA) 557846
    Mouse IgG2a Sigma (St. Louis, MO) F-6522
    Mouse Sigma (St. Louis, MO) P-4685
    IgG1kappa
  • Mixed Lymphocyte Reaction. Cryopreserved vials of passage 10 umbilicus-derived cells labeled as cell line A and passage 11 placenta-derived cells labeled as cell line B were sent on dry ice to CTBR (Senneville, Quebec) to conduct a mixed lymphocyte reaction using CTBR SOP No. CAC-031. Peripheral blood mononuclear cells (PBMCs) were collected from multiple male and female volunteer donors. Stimulator (donor) allogeneic PBMC, autologous PBMC, and postpartum cell lines were treated with mitomycin C. Autologous and mitomycin C-treated stimulator cells were added to responder (recipient) PBMCs and cultured for 4 days. After incubation, [3H]thymidine was added to each sample and cultured for 18 hours. Following harvest of the cells, radiolabeled DNA was extracted, and [3H]-thymidine incorporation was measured using a scintillation counter.
  • The stimulation index for the allogeneic donor (SIAD) was calculated as the mean proliferation of the receiver plus mitomycin C-treated allogeneic donor divided by the baseline proliferation of the receiver. The stimulation index of the PPDCs was calculated as the mean proliferation of the receiver plus mitomycin C-treated postpartum cell line divided by the baseline proliferation of the receiver.
  • Results
  • Mixed lymphocyte reaction—placenta-derived cells. Seven human volunteer blood donors were screened to identify a single allogeneic donor that would exhibit a robust proliferation response in a mixed lymphocyte reaction with the other six blood donors. This donor was selected as the allogeneic positive control donor. The remaining six blood donors were selected as recipients. The allogeneic positive control donor and placenta-derived cell lines were treated with mitomycin C and cultured in a mixed lymphocyte reaction with the six individual allogeneic receivers. Reactions were performed in triplicate using two cell culture plates with three receivers per plate (Table 11-2). The average stimulation index ranged from 1.3 (plate 2) to 3 (plate 1) and the allogeneic donor positive controls ranged from 46.25 (plate 2) to 279 (plate 1) (Table 11-3).
    TABLE 11-2
    Mixed Lymphocyte Reaction Data - Cell Line B (Placenta)
    DPM for Proliferation Assay
    Analytical Culture Replicates
    number System 1 2 3 Mean SD CV
    Plate ID: Plate1
    IM03-7769 Proliferation baseline of receiver 79 119 138 112.0 30.12 26.9
    Control of autostimulation (Mitomycin C treated autologous cells) 241 272 175 229.3 49.54 21.6
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 23971 22352 20921 22414.7 1525.97 6.8
    MLR with cell line (Mitomycin C treated cell type B) 664 559 1090 771.0 281.21 36.5
    SI (donor) 200
    SI (cell line) 7
    IM03-7770 Proliferation baseline of receiver 206 134 262 200.7 64.17 32.0
    Control of autostimulation (Mitomycin C treated autologous cells) 1091 602 524 739.0 307.33 41.6
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 45005 43729 44071 44268.3 660.49 1.5
    MLR with cell line (Mitomycin C treated cell type B) 533 2582 2376 1830.3 1128.24 61.6
    SI (donor) 221
    SI (cell line) 9
    IM03-7771 Proliferation baseline of receiver 157 87 128 124.0 35.17 28.4
    Control of autostimulation (Mitomycin C treated autologous cells) 293 138 508 313.0 185.81 59.4
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 24497 34348 31388 30077.7 5054.53 16.8
    MLR with cell line (Mitomycin C treated cell type B) 601 643 a 622.0 29.70 4.8
    SI (donor) 243
    SI (cell line) 5
    IM03-7772 Proliferation baseline of receiver 56 98 51 68.3 25.81 37.8
    Control of autostimulation (Mitomycin C treated autologous cells) 133 120 213 155.3 50.36 32.4
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 14222 20076 22168 18822.0 4118.75 21.9
    MLR with cell line (Mitomycin C treated cell type B) a a a a a a
    SI (donor) 275
    SI (cell line) a
    IM03-7768 Proliferation baseline of receiver 84 242 208 178.0 83.16 46.7
    (allogenic Control of autostimulation (Mitomycin treated autologous cells) 361 617 304 427.3 166.71 39.0
    donor)
    Cell line Proliferation baseline of receiver 126 124 143 131.0 10.44 8.0
    type B Control of autostimulation (Mitomycin treated autologous cells) 822 1075 487 794.7 294.95 37.1
    Plate ID: Plate 2
    IM03-7773 Proliferation baseline of receiver 908 181 330 473.0 384.02 81.2
    Control of autostimulation (Mitomycin C treated autologous cells) 269 405 572 415.3 151.76 36.5
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 29151 28691 28315 28719.0 418.70 1.5
    MLR with cell line (Mitomycin C treated cell type B) 567 732 905 734.7 169.02 23.0
    SI (donor) 61
    SI (cell line) 2
    IM03-7774 Proliferation baseline of receiver 893 1376 185 818.0 599.03 73.2
    Control of autostimulation (Mitomycin C treated autologous cells) 261 381 568 403.3 154.71 38.4
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 53101 42839 48283 48074.3 5134.18 10.7
    MLR with cell line (Mitomycin C treated cell type B) 515 789 294 532.7 247.97 46.6
    SI (donor) 59
    SI (cell line) 1
    IM03-7775 Proliferation baseline of receiver 1272 300 544 705.3 505.69 71.7
    Control of autostimulation (Mitomycin C treated autologous cells) 232 199 484 305.0 155.89 51.1
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 23554 10523 28965 21014.0 9479.74 45.1
    MLR with cell line (Mitomycin C treated cell type B) 768 924 563 751.7 181.05 24.1
    SI (donor) 30
    SI (cell line) 1
    IM03-7776 Proliferation baseline of receiver 1530 137 1046 904.3 707.22 78.2
    Control of autostimulation (Mitomycin C treated autologous cells) 420 218 394 344.0 109.89 31.9
    MLR allogenic donor IM03-7768 (Mitomycin C treated) 28893 32493 34746 32044.0 2952.22 9.2
    MLR with cell line (Mitomycin C treated cell type B) a a a a a a
    SI (donor) 35
    SI (cell line) a
  • TABLE 11-3
    Average stimulation index of placenta cells and an allogeneic donor
    in a mixed lymphocyte reaction with six individual allogeneic receivers
    Average Stimulation Index
    Recipient Placenta
    Plate 1 (receivers 1-3) 279 3
    Plate 2 (receivers 4-6) 46.25 1.3
  • Mixed lymphocyte reaction—umbilicus-derived cells. Six human volunteer blood donors were screened to identify a single allogeneic donor that will exhibit a robust proliferation response in a mixed lymphocyte reaction with the other five blood donors. This donor was selected as the allogeneic positive control donor. The remaining five blood donors were selected as recipients. The allogeneic positive control donor and placenta cell lines were mitomycin C-treated and cultured in a mixed lymphocyte reaction with the five individual allogeneic receivers. Reactions were performed in triplicate using two cell culture plates with three receivers per plate (Table 11-4). The average stimulation index ranged from 6.5 (plate 1) to 9 (plate 2) and the allogeneic donor positive controls ranged from 42.75 (plate 1) to 70 (plate 2) (Table 11-5).
    TABLE 11-4
    Mixed Lymphocyte Reaction Data-Cell Line A (Umbilicus)
    DPM for Proliferation Assay
    Analytical Culture Replicates
    number System 1 2 3 Mean SD CV
    Plate ID: Plate1
    IM04-2478 Proliferation baseline of receiver 1074 406 391 623.7 390.07 62.5
    Control of autostimulation (Mitomycin C treated autologous cells) 672 510 1402 861.3 475.19 55.2
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 43777 48391 38231 43466.3 5087.12 11.7
    MLR with cell line (Mitomycin C treated cell type A) 2914 5622 6109 4881.7 1721.36 35.3
    SI (donor) 70
    SI (cell line) 8
    IM04-2479 Proliferation baseline of receiver 530 508 527 521.7 11.93 2.3
    control of autostimulation (Mitomycin C treated autologous cells) 701 567 1111 793.0 283.43 35.7
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 25593 24732 22707 24344.0 1481.61 6.1
    MLR with cell line (Mitomycin C treated cell type A) 5086 3932 1497 3505.0 1832.21 52.3
    SI (donor) 47
    SI (cell line) 7
    IM04-2480 Proliferation baseline of receiver 1192 854 1330 1125.3 244.90 21.8
    Control of autostimulation (Mitomycin C treated autologous cells) 2963 993 2197 2051.0 993.08 48.4
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 25416 29721 23757 26298.0 3078.27 11.7
    MLR with cell line (Mitomycin C treated cell type A) 2596 5076 3426 3699.3 1262.39 34.1
    SI (donor) 23
    SI (cell line) 3
    IM04-2481 Proliferation baseline of receiver 695 451 555 567.0 122.44 21.6
    Control of autostimulation (Mitomycin C treated autologous cells) 738 1252 464 818.0 400.04 48.9
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 13177 24885 15444 17835.3 6209.52 34.8
    MLR with cell line (Mitomycin C treated cell type A) 4495 3671 4674 4280.0 534.95 12.5
    SI (donor) 31
    SI (cell line) 8
    Plate ID: Plate 2
    IM04-2482 Proliferation baseline of receiver 432 533 274 413.0 130.54 31.6
    Control of autostimulation (Mitomycin C treated autologous cells) 1459 633 598 896.7 487.31 54.3
    MLR allogenic donor IM04-2477 (Mitomycin C treated) 24286 30823 31346 28818.3 3933.82 13.7
    MLR with cell line (Mitomycin C treated cell type A) 2762 1502 6723 3662.3 2724.46 74.4
    SI (donor) 70
    SI (cell line) 9
    IM04-2477 Proliferation baseline of receiver 312 419 349 360.0 54.34 15.1
    (allogenic Control of autostimulation (Mitomycin treated autologous cells) 567 604 374 515.0 123.50 24.0
    donor)
    Cell line Proliferation baseline of receiver 5101 3735 2973 3936.3 1078.19 27.4
    type A Control of autostimulation (Mitomycin treated autologous cells) 1924 4570 2153 2882.3 1466.04 50.9
  • TABLE 11-5
    Average stimulation index of umbilicus-derived cells and an
    allogeneic donor in a mixed lymphocyte reaction with five
    individual allogeneic receivers.
    Average Stimulation Index
    Recipient Umbilicus
    Plate 1 (receivers 1-4) 42.75 6.5
    Plate 2 (receiver 5) 70 9
  • Antigen presenting cell markers—placenta-derived cells. Histograms of placenta-derived cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, as noted by fluorescence value consistent with the IgG control, indicating that placental cell lines lack the cell surface molecules required to directly stimulate CD4+ T cells.
  • Immunomodulating markers—placenta-derived cells. Histograms of placenta-derived cells analyzed by flow cytometry show positive expression of PD-L2, as noted by the increased value of fluorescence relative to the IgG control, and negative expression of CD178 and HLA-G, as noted by fluorescence value consistent with the IgG control.
  • Antigen presenting cell markers—umbilicus-derived cells. Histograms of umbilicus-derived cells analyzed by flow cytometry show negative expression of HLA-DR, DP, DQ, CD80, CD86, and B7-H2, as noted by fluorescence value consistent with the IgG control, indicating that umbilical cell lines lack the cell surface molecules required to directly stimulate CD4+ T cells.
  • Immunomodulating cell markers—umbilicus-derived cells. Histograms of umbilicus-derived cells analyzed by flow cytometry show positive expression of PD-L2, as noted by the increased value of fluorescence relative to the IgG control, and negative expression of CD178 and HLA-G, as noted by fluorescence value consistent with the IgG control.
  • Summary. In the mixed lymphocyte reactions conducted with placenta-derived cell lines, the average stimulation index ranged from 1.3 to 3, and that of the allogeneic positive controls ranged from 46.25 to 279. In the mixed lymphocyte reactions conducted with umbilicus-derived cell lines the average stimulation index ranged from 6.5 to 9, and that of the allogeneic positive controls ranged from 42.75 to 70. Placenta- and umbilicus-derived cell lines were negative for the expression of the stimulating proteins HLA-DR, HLA-DP, HLA-DQ, CD80, CD86, and B7-H2, as measured by flow cytometry. Placenta- and umbilicus-derived cell lines were negative for the expression of immuno-modulating proteins HLA-G and CD178 and positive for the expression of PD-L2, as measured by flow cytometry. Allogeneic donor PBMCs contain antigen-presenting cells expressing HLA-DR, DQ, CD8, CD86, and B7-H2, thereby allowing for the stimulation of naïve CD4+ T cells. The absence of antigen-presenting cell surface molecules on placenta- and umbilicus-derived cells required for the direct stimulation of naïve CD4+ T cells and the presence of PD-L2, an immunomodulating protein, may account for the low stimulation index exhibited by these cells in a MLR as compared to allogeneic controls.
  • EXAMPLE 12 Secretion of Trophic Factors by Postpartum-Derived Cells
  • The secretion of selected trophic factors from placenta- and umbilicus-derived cells was measured. Factors selected for detection included: (1) those known to have angiogenic activity, such as hepatocyte growth factor (HGF) (Rosen et al. (1997) Ciba Found. Symp. 212:215-26), monocyte chemotactic protein 1 (MCP-1) (Salcedo et al. (2000) Blood 96; 34-40), interleukin-8 (IL-8) (L1 et al. (2003) J. Immunol. 170:3369-76), keratinocyte growth factor (KGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF) (Hughes et al. (2004) Ann. Thorac. Surg. 77:812-8), matrix metalloproteinase 1 (TIMP1), angiopoietin 2 (ANG2), platelet derived growth factor (PDGF-bb), thrombopoietin (TPO), heparin-binding epidermal growth factor (HB-EGF), stromal-derived factor 1 alpha (SDF-1alpha); (2) those known to have neurotrophic/neuroprotective activity, such as brain-derived neurotrophic factor (BDNF) (Cheng et al. (2003) Dev. Biol. 258; 319-33), interleukin-6 (IL-6), granulocyte chemotactic protein-2 (GCP-2), transforming growth factor beta2 (TGFbeta2); and (3) those known to have chemokine activity, such as macrophage inflammatory protein 1 alpha (MIP1a), macrophage inflammatory protein 1beta (MIP1b), monocyte chemoattractant-1 (MCP-1), Rantes (regulated on activation, normal T cell expressed and secreted), 1309, thymus and activation-regulated chemokine (TARC), Eotaxin, macrophage-derived chemokine (MDC), IL-8).
  • Methods & Materials
  • Cell culture. PPDCs from placenta and umbilicus as well as human fibroblasts derived from human neonatal foreskin were cultured in Growth Medium with penicillin/streptomycin on gelatin-coated T75 flasks. Cells were cryopreserved at passage 11 and stored in liquid nitrogen. After thawing of the cells, Growth Medium was added to the cells followed by transfer to a 15 milliliter centrifuge tube and centrifugation of the cells at 150×g for 5 minutes. The supernatant was discarded. The cell pellet was resuspended in 4 milliliters Growth Medium, and cells were counted. Cells were seeded at 375,000 cells/75 cm2 flask containing 15 milliliters of Growth Medium and cultured for 24 hours. The medium was changed to a serum-free medium (DMEM-low glucose (Gibco), 0.1% (w/v) bovine serum albumin (Sigma), penicillin/streptomycin (Gibco)) for 8 hours. Conditioned serum-free medium was collected at the end of incubation by centrifugation at 14,000×g for 5 minutes and stored at −20° C. To estimate the number of cells in each flask, cells were washed with PBS and detached using 2 milliliters trypsin/EDTA. Trypsin activity was inhibited by addition of 8 milliliters Growth Medium. Cells were centrifuged at 150×g for 5 minutes. Supernatant was removed, and cells were resuspended in 1 milliliter Growth Medium. Cell number was estimated using a hemocytometer.
  • ELISA assay. Cells were grown at 37° C. in 5% carbon dioxide and atmospheric oxygen. Placenta-derived cells (batch 101503) also were grown in 5% oxygen or beta-mercaptoethanol (BME). The amount of MCP-1, IL-6, VEGF, SDF-1alpha, GCP-2, IL-8, and TGF-beta 2 produced by each cell sample was measured by an ELISA assay (R&D Systems, Minneapolis, Minn.). All assays were performed according to the manufacturer's instructions.
  • SearchLight multiplexed ELISA assay. Chemokines (MIP1a, MIP1b, MCP-1, Rantes, 1309, TARC, Eotaxin, MDC, IL8), BDNF, and angiogenic factors (HGF, KGF, bFGF, VEGF, TIMP1, ANG2, PDGF-bb, TPO, HB-EGF were measured using SearchLight Proteome Arrays (Pierce Biotechnology Inc.). The Proteome Arrays are multiplexed sandwich ELISAs for the quantitative measurement of two to 16 proteins per well. The arrays are produced by spotting a 2×2, 3×3, or 4×4 pattern of four to 16 different capture antibodies into each well of a 96-well plate. Following a sandwich ELISA procedure, the entire plate is imaged to capture chemiluminescent signal generated at each spot within each well of the plate. The amount of signal generated in each spot is proportional to the amount of target protein in the original standard or sample.
  • Results
  • ELISA assay. MCP-1 and IL-6 were secreted by placenta- and umbilicus-derived cells and dermal fibroblasts (Table 12-1). SDF-1 alpha was secreted by placenta-derived cells cultured in 5% O2 and by fibroblasts. GCP-2 and IL-8 were secreted by umbilicus-derived cells and by placenta-derived cells cultured in the presence of BME or 5% O2. GCP-2 also was secreted by human fibroblasts. TGF-beta2 was not detectable by ELISA assay.
    TABLE 12-1
    ELISA assay results
    (values presented are picograms/milliliter/million cells (n = 2, sem)
    TGF-
    MCP-1 IL-6 VEGF SDF-1α GCP-2 IL-8 β2
    Fibroblast 17 ± 1  61 ± 3 29 ± 2 19 ± 1 21 ± 1 ND ND
    Placenta (042303) 60 ± 3  41 ± 2 ND ND ND ND ND
    Umbilicus (022803) 1150 ± 74  4234 ± 289 ND ND 160 ± 11 2058 ± 145 ND
    Placenta (071003) 125 ± 16  10 ± 1 ND ND ND ND ND
    Umbilicus (071003) 2794 ± 84  1356 ± 43  ND ND 2184 ± 98  2369 ± 23  ND
    Placenta (101503) BME 21 ± 10 67 ± 3 ND ND 44 ± 9 17 ± 9 ND
    Placenta (101503) 5% O2, W/O 77 ± 16 339 ± 21 ND 1149 ± 137 54 ± 2 265 ± 10 ND
    BME

    Key:

    ND: Not Detected.
  • SearchLight multiplexed ELISA assay. TIMP1, TPO, KGF, HGF, FGF, HBEGF, BDNF, MIP1b, MCP1, RANTES, 1309, TARC, MDC, and IL-8 were secreted from umbilicus-derived cells (Tables 12-2 and 12-3). TIMP1, TPO, KGF, HGF, HBEGF, BDNF, MIP1a, MCP-1, RANTES, TARC, Eotaxin, and IL-8 were secreted from placenta-derived cells (Tables 12-2 and 12-3). No Ang2, VEGF, or PDGF-bb were detected.
    TABLE 12-2
    SearchLight Multiplexed ELISA assay results
    TIMP1 ANG2 PDGFbb TPO KGF HGF FGF VEGF HBEGF BDNF
    hFB 19306.3 ND ND 230.5 5.0 ND ND 27.9 1.3 ND
    P1 24299.5 ND ND 546.6 8.8 16.4 ND ND 3.81.3 ND
    U1 57718.4 ND ND 1240.0 5.8 559.3 148.7 ND 9.3 165.7
    P3 14176.8 ND ND 568.7 5.2 10.2 ND ND 1.9 33.6
    U3 21850.0 ND ND 1134.5 9.0 195.6 30.8 ND 5.4 388.6

    Key:

    hFB (human fibroblasts),

    P1 (placenta-derived cells (042303)),

    U1 (umbilicus-derived cells (022803)),

    P3 (placenta-derived cells(071003)),

    U3 (umbilicus-derived cells (071003)).

    ND: Not Detected.
  • TABLE 12-3
    SearchLight Multiplexed ELISA assay results
    MIP1a MIP1b MCP1 RANTES I309 TARC Eotaxin MDC IL8
    hFB ND ND 39.6 ND ND 0.1 ND ND 204.9
    P1 79.5 ND 228.4  4.1 ND 3.8 12.2 ND 413.5
    U1 ND 8.0 1694.2 ND 22.4 37.6 ND 18.9 51930.1
    P3 ND ND 102.7 ND ND 0.4 ND ND 63.8
    U3 ND 5.2 2018.7 41.5 11.6 21.4 ND  4.8 10515.9

    Key:

    hFB (human fibroblasts),

    P1 (placenta-derived PPDC (042303)),

    U1 (umbilicus-derived PPDC (022803)),

    P3 (placenta-derived PPDC (071003)),

    U3 (umbilicus-derived PPDC (071003)).

    ND: Not Detected.
  • Summary. Umbilicus- and placenta-derived cells secreted a number of trophic factors. Some of these trophic factors, such as HGF, bFGF, MCP-1 and IL-8, play important roles in angiogenesis. Other trophic factors, such as BDNF and IL-6, have important roles in neural regeneration.
  • EXAMPLE 13 Short-Term Neural Differentiation of Postpartum-Derived Cells
  • The ability of placenta- and umbilicus-derived cells (collectively postpartum-derived cells or PPDCs) to differentiate into neural lineage cells was examined.
  • Materials & Methods
  • Isolation and Expansion of Postpartum Cells. PPDCs from placental and umbilical tissues were isolated and expanded as described in Example 1.
  • Modified Woodbury-Black Protocol. (A) This assay was adapted from an assay originally performed to test the neural induction potential of bone marrow stromal cells (1). Umbilicus-derived cells (022803) P4 and placenta-derived cells (042203) P3 were thawed and culture expanded in Growth Media at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 6,000 cells per well of a Titretek II glass slide (VWR International, Bristol, CT). As controls, mesenchymal stem cells (P3; 1F2155; Cambrex, Walkersville, MD), osteoblasts (P5; CC2538; Cambrex), adipose-derived cells (Artecel, U.S. Pat. No. 6,555,374 B1) (P6; Donor 2) and neonatal human dermal fibroblasts (P6; CC2509; Cambrex) were also seeded under the same conditions.
  • All cells were initially expanded for 4 days in DMEM/F12 medium (Invitrogen, Carlsbad, Calif.) containing 15% (v/v) fetal bovine serum (FBS; Hyclone, Logan, Utah), basic fibroblast growth factor (bFGF; 20 nanograms/milliliter; Peprotech, Rocky Hill, N.J.), epidermal growth factor (EGF; 20 nanograms/milliliter; Peprotech) and penicillin/streptomycin (Invitrogen). After four days, cells were rinsed in phosphate-buffered saline (PBS; Invitrogen) and were subsequently cultured in DMEM/F12 medium+20% (v/v) FBS+penicillin/streptomycin for 24 hours. After 24 hours, cells were rinsed with PBS. Cells were then cultured for 1-6 hours in an induction medium which was comprised of DMEM/F12 (serum-free) containing 200 mM butylated hydroxyanisole, 10 μM potassium chloride, 5 milligram/milliliter insulin, 10 μM forskolin, 4 μM valproic acid, and 2 μM hydrocortisone (all chemicals from Sigma, St. Louis, Mo.). Cells were then fixed in 100% ice-cold methanol and immunocytochemistry was performed (see methods below) to assess human nestin protein expression.
  • (B) PPDCs (umbilicus (022803) P11; placenta (042203) P11) and adult human dermal fibroblasts (1F1853, P11) were thawed and culture expanded in Growth Medium at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at similar density as in (A), but onto (1) 24 well tissue culture-treated plates (TCP, Falcon brand, VWR International), (2) TCP wells+2% (w/v) gelatin adsorbed for 1 hour at room temperature, or (3) TCP wells+20 μg/milliliter adsorbed mouse laminin (adsorbed for a minimum of 2 hours at 37° C.; Invitrogen).
  • Exactly as in (A), cells were initially expanded and media switched at the aforementioned timeframes. One set of cultures was fixed, as before, at 5 days and six hours, this time with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. In the second set of cultures, medium was removed and switched to Neural Progenitor Expansion medium (NPE) consisting of Neurobasal-A medium (Invitrogen) containing B27 (B27 supplement; Invitrogen), L-glutamine (4 mM), and penicillin/streptomycin (Invitrogen). NPE medium was further supplemented with retinoic acid (RA; 1 μM; Sigma). This medium was removed 4 days later and cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for nestin, GFAP, and TuJ1 protein expression (see Table N1-1).
    TABLE 13-1
    Summary of Primary Antibodies Used
    Antibody Concentration Vendor
    Rat 401 (nestin) 1:200 Chemicon, Temecula,
    CA
    Human Nestin 1:100 Chemicon
    TuJ1 (BIII Tubulin) 1:500 Sigma, St. Louis, MO
    GFAP 1:2000 DakoCytomation,
    Carpinteria, CA
    Tyrosine hydroxylase (TH) 1:1000 Chemicon
    GABA 1:400 Chemicon
    Desmin (mouse) 1:300 Chemicon
    alpha - alpha-smooth muscle 1:400 Sigma
    actin
    Human nuclear protein (hNuc) 1:150 Chemicon
  • Two Stage Differentiation Protocol. PPDCs (umbilicus (042203) P11, placenta (022803) P11), adult human dermal fibroblasts (P11; 1F1853; Cambrex) were thawed and culture expanded in Growth Medium at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm2, but onto 24 well plates coated with laminin (BD Biosciences, Franklin Lakes, N.J.) in the presence of NPE media supplemented with bFGF (20 nanograms/milliliter; Peprotech, Rocky Hill, NJ) and EGF (20 nanograms/milliliter; Peprotech) [whole media composition further referred to as NPE+F+E]. At the same time, adult rat neural progenitors isolated from hippocampus (P4; (062603) were also plated onto 24 well laminin-coated plates in NPE+F+E media. All cultures were maintained in such conditions for a period of 6 days (cells were fed once during that time) at which time media was switched to the differentiation conditions listed in Table N 1-2 for an additional period of 7 days. Cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human or rat nestin, GFAP, and TuJ1 protein expression.
    TABLE 13-2
    Summary of Conditions for Two-Stage Differentiation Protocol
    A B
    COND. # PRE-DIFFERENTIATION 2nd STAGE DIFF
    1 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + SHH (200 ng/ml) + F8 (100 ng/ml)
    2 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + SHH (200 ng/ml) + F8 (100 ng/ml) + RA
    (1 μM)
    3 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + RA (1 μM)
    4 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + F (20 ng/ml) + E (20 ng/ml)
    5 NPE + F (20 ng/ml) + E (20 ng/ml) Growth Medium
    6 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 1B + MP52 (20 ng/ml)
    7 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 1B + BMP7 (20 ng/ml)
    8 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 1B + GDNF (20 ng/ml)
    9 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 2B + MP52 (20 ng/ml)
    10 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 2B + BMP7 (20 ng/ml)
    11 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 2B + GDNF (20 ng/ml)
    12 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 3B + MP52 (20 ng/ml)
    13 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 3B + BMP7 (20 ng/ml)
    14 NPE + F (20 ng/ml) + E (20 ng/ml) Condition 3B + GDNF (20 ng/ml)
    15 NPE + F (20 ng/ml) + E (20 ng/mL) NPE + MP52 (20 ng/ml)
    16 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + BMP7 (20 ng/ml)
    17 NPE + F (20 ng/ml) + E (20 ng/ml) NPE + GDNF (20 ng/ml)
  • Multiple growth factor protocol. Umbilicus-derived cells (P11; (042203)) were thawed and culture expanded in Growth Medium at 5,000 cells/cm2 until sub-confluence (75%) was reached. Cells were then trypsinized and seeded at 2,000 cells/cm2, onto 24 well laminin-coated plates (BD Biosciences) in the presence of NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter). In addition, some wells contained NPE+F+E+2% FBS or 10% FBS. After four days of “pre-differentiation” conditions, all media were removed and samples were switched to NPE medium supplemented with sonic hedgehog (SHH; 200 nanograms/milliliter; Sigma, St. Louis, Mo.), FGF8 (100 nanograms/milliliter; Peprotech), BDNF (40 nanograms/milliliter; Sigma), GDNF (20 nanograms/milliliter; Sigma), and retinoic acid (1 μM; Sigma). Seven days post medium change, cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nestin, GFAP, TuJ1, desmin, and alpha-smooth muscle actin expression.
  • Neural progenitor co-culture protocol. Adult rat hippocampal progenitors (062603) were plated as neurospheres or single cells (10,000 cells/well) onto laminin-coated 24 well dishes (BD Biosciences) in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter).
  • Separately, umbilicus-derived cells (042203) P11 and placenta-derived cells (022803) P11 were thawed and culture expanded in NPE+F (20 nanograms/milliliter)+E (20 nanograms/milliliter) at 5,000 cells/cm2 for a period of 48 hours. Cells were then trypsinized and seeded at 2,500 cells/well onto existing cultures of neural progenitors. At that time, existing medium was exchanged for fresh medium. Four days later, cultures were fixed with ice-cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature, and stained for human nuclear protein (hNuc; Chemicon) (Table NU1-1 above) to identify PPDCs.
  • Immunocytochemistry. Immunocytochemistry was performed using the antibodies listed in Table NU1-1. Cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti-mouse IgG—Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG—Alexa 488 (1:250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
  • Results
  • Woodbury-Black protocol. (A) Upon incubation in this neural induction composition, all cell types transformed into cells with bipolar morphologies and extended processes. Other larger non-bipolar morphologies were also observed. Furthermore, the induced cell populations stained positively for nestin, a marker of multipotent neural stem and progenitor cells.
  • (B) When repeated on tissue culture plastic (TCP) dishes, nestin expression was not observed unless laminin was pre-adsorbed to the culture surface. To further assess whether nestin-expressing cells could then go on to generate mature neurons, PPDCs and fibroblasts were exposed to NPE+RA (1 μM), a media composition known to induce the differentiation of neural stem and progenitor cells into such cells (2,3,4). Cells were stained for TuJ1, a marker for immature and mature neurons, GFAP, a marker of astrocytes, and nestin. Under no conditions was TuJ1 detected, nor were cells with neuronal morphology observed, suggesting that neurons were not generated in the short term. Furthermore, nestin and GFAP were no longer expressed by PPDCs, as determined by immunocytochemistry.
  • Two-stage differentiation. Umbilicus and placenta PPDC isolates (as well as human fibroblasts and rodent neural progenitors as negative and positive control cell types, respectively) were plated on laminin (neural promoting)-coated dishes and exposed to 13 different growth conditions (and two control conditions) known to promote differentiation of neural progenitors into neurons and astrocytes. In addition, two conditions were added to examine the influence of GDF5, and BMP7 on PPDC differentiation. Generally, a two-step differentiation approach was taken, where the cells were first placed in neural progenitor expansion conditions for a period of 6 days, followed by full differentiation conditions for 7 days. Morphologically, both umbilicus- and placenta-derived cells exhibited fundamental changes in cell morphology throughout the time-course of this procedure. However, neuronal or astrocytic-shaped cells were not observed except for in control, neural progenitor-plated conditions. Immunocytochemistry, negative for human nestin, TuJ1, and GFAP confirmed the morphological observations.
  • Multiple growth factors. Following one week's exposure to a variety of neural differentiation agents, cells were stained for markers indicative of neural progenitors (human nestin), neurons (TuJ1), and astrocytes (GFAP). Cells grown in the first stage in non-serum containing media had different morphologies than those cells in serum containing (2% or 10%) media, indicating potential neural differentiation. Specifically, following a two step procedure of exposing umbilicus-derived cells to EGF and bFGF, followed by SHH, FGF8, GDNF, BDNF, and retinoic acid, cells showed long extended processes similar to the morphology of cultured astrocytes. When 2% FBS or 10% FBS was included in the first stage of differentiation, cell number was increased and cell morphology was unchanged from control cultures at high density. Potential neural differentiation was not evidenced by immunocytochemical analysis for human nestin, TuJ1, or GFAP.
  • Neural progenitor and PPDC co-culture. PPDCs were plated onto cultures of rat neural progenitors seeded two days earlier in neural expansion conditions (NPE+F+E). While visual confirmation of plated PPDCs proved that these cells were plated as single cells, human-specific nuclear staining (hNuc) 4 days post-plating (6 days total) showed that they tended to ball up and avoid contact with the neural progenitors. Furthermore, where PPDCs attached, these cells spread out and appeared to be innervated by differentiated neurons that were of rat origin, suggesting that the PPDCs may have differentiated into muscle cells. This observation was based upon morphology under phase contrast microscopy. Another observation was that typically large cell bodies (larger than neural progenitors) possessed morphologies resembling neural progenitors, with thin processes spanning out in multiple directions. HNuc staining (found in one half of the cell's nucleus) suggested that in some cases these human cells may have fused with rat progenitors and assumed their phenotype. Control wells containing only neural progenitors had fewer total progenitors and apparent differentiated cells than did co-culture wells containing umbilicus or placenta PPDCs, further indicating that both umbilicus- and placenta-derived cells influenced the differentiation and behavior of neural progenitors, either by release of chemokines and cytokines, or by contact-mediated effects.
  • Summary. Multiple protocols were conducted to determine the short term potential of PPDCs to differentiate into neural lineage cells. These included phase contrast imaging of morphology in combination with immunocytochemistry for nestin, TuJ1, and GFAP, proteins associated with multipotent neural stem and progenitor cells, immature and mature neurons, and astrocytes, respectively. Evidence was observed to suggest that neural differentiation occurred in certain instances in these short-term protocols.
  • Several notable observations were made in co-cultures of PPDCs with neural progenitors. This approach, using human PPDCs along with a xenogeneic cell type allowed for absolute determination of the origin of each cell in these cultures. First, some cells were observed in these cultures where the cell cytoplasm was enlarged, with neurite-like processes extending away from the cell body, yet only half of the body labeled with hNuc protein. Those cells may have been human PPDCs that had differentiated into neural lineage cells or they may have been PPDCs that had fused with neural progenitors. Second, it appeared that neural progenitors extended neurites to PPDCs in a way that indicates the progenitors differentiated into neurons and innervated the PPDCs. Third, cultures of neural progenitors and PPDCs had more cells of rat origin and larger amounts of differentiation than control cultures of neural progenitors alone, further indicating that plated PPDCs provided soluble factors and or contact-dependent mechanisms that stimulated neural progenitor survival, proliferation, and/or differentiation.
  • References for Example 13
    • (1) Woodbury, D. et al. (2000). J. Neurosci. Research. 61(4): 364-70.
    • (2) Jang, Y. K. et al. (2004). J. Neurosci. Research. 75(4): 573-84.
    • (3) Jones-Villeneuve, E. M. et al. (1983). Mol Cel Biol. 3(12): 2271-9.
    • (4) Mayer-Proschel, M. et al. (1997). Neuron. 19(4): 773-85.
    EXAMPLE 14 Long-Term Neural Differentiation of Postpartum-Derived Cells
  • The ability of umbilicus and placenta-derived cells (collectively postpartum-derived cells or PPDCs) to undergo long-term differentiation into neural lineage cells was evaluated.
  • Materials & Methods
  • Isolation and Expansion of PPDCs. PPDCs were isolated and expanded as described in previous Examples.
  • PPDC Cell Thaw and Plating. Frozen aliquots of PPDCs (umbilicus (022803) P11; (042203) P11; (071003) P12; placenta (101503) P7) previously grown in Growth Medium were thawed and plated at 5,000 cells/cm2 in T-75 flasks coated with laminin (BD, Franklin Lakes, N.J.) in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement, Invitrogen), L-glutamine (4 mM), and Penicillin/Streptomycin (10 milliliters), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) media. NPE media was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ), herein referred to as NPE+bFGF+EGF.
  • Control Cell Plating. In addition, adult human dermal fibroblasts (P11, Cambrex, Walkersville, MD) and mesenchymal stem cells (P5, Cambrex) were thawed and plated at the same cell seeding density on laminin-coated T-75 flasks in NPE+bFGF+EGF. As a further control, fibroblasts, umbilicus, and placenta PPDCs were grown in Growth Medium for the period specified for all cultures.
  • Cell Expansion. Media from all cultures were replaced with fresh media once a week and cells observed for expansion. In general, each culture was passaged one time over a period of one month because of limited growth in NPE+bFGF+EGF.
  • Immunocytochemistry. After a period of one month, all flasks were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against TuJ1 (BIII Tubulin; 1:500; Sigma, St. Louis, Mo.) and GFAP (glial fibrillary acidic protein; 1:2000; DakoCytomation, Carpinteria, CA). Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing block along with goat anti-mouse IgG—Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG—Alexa 488 (1:250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
    TABLE 14-1
    Summary of Primary Antibodies Used
    Antibody Concentration Vendor
    TuJ1 (BIII Tubulin) 1:500 Sigma, St. Louis, MO
    GFAP 1:2000 DakoCytomation, Carpinteria, CA

    Results
  • NPE+bFGF+EGF media slows proliferation of PPDCs and alters their morphology. Immediately following plating, a subset of PPDCs attached to the culture flasks coated with laminin. This may have been due to cell death as a function of the freeze/thaw process or because of the new growth conditions. Cells that did attach adopted morphologies different from those observed in Growth Media.
  • Upon confluence, cultures were passaged and observed for growth. Very little expansion took place of those cells that survived passage. At this point, very small cells with no spread morphology and with phase-bright characteristics began to appear in cultures of umbilicus-derived cells. These areas of the flask were followed over time. From these small cells, bifurcating processes emerged with varicosities along their lengths, features very similar to previously described PSA-NCAM+neuronal progenitors and TuJ1+immature neurons derived from brain and spinal cord (1, 2). With time, these cells became more numerous, yet still were only found in clones.
  • Clones of umbilicus-derived cells express neuronal proteins. Cultures were fixed at one month post-thawing/plating and stained for the neuronal protein TuJ1 and GFAP, an intermediate filament found in astrocytes. While all control cultures grown in Growth Medium and human fibroblasts and MSCs grown in NPE+bFGF+EGF medium were found to be TuJ1-/GFAP-, TuJ1 was detected in the umbilicus and placenta PPDCs. Expression was observed in cells with and without neuronal-like morphologies. No expression of GFAP was observed in either culture. The percentage of cells expressing TuJ1 with neuronal-like morphologies was less than or equal to 1% of the total population (n=3 umbilicus-derived cell isolates tested). While not quantified, the percentage of TuJ1+cells without neuronal morphologies was higher in umbilicus-derived cell cultures than placenta-derived cell cultures. These results appeared specific as age-matched controls in Growth Medium did not express TuJ1.
  • Summary. Methods for generating differentiated neurons (based on TuJ1 expression and neuronal morphology) from umbilicus-derived cells were developed. While expression for TuJ1 was not examined earlier than one month in vitro, it is clear that at least a small population of umbilicus-derived cells can give rise to neurons either through default differentiation or through long-term induction following one month's exposure to a minimal media supplemented with L-glutamine, basic FGF, and EGF.
  • References for Example 14
    • (1) Mayer-Proschel, M. et al. (1997). Neuron. 19(4): 773-85.
    • (2) Yang, H. et al. (2000). PNAS. 97(24): 13366-71.
    EXAMPLE 15 PPDC Trophic Factors for Neural Progenitor Support
  • The influence of umbilicus- and placenta-derived cells (collectively postpartum-derived cells or PPDCs) on adult neural stem and progenitor cell survival and differentiation through non-contact dependent (trophic) mechanisms was examined.
  • Materials & Methods
  • Adult neural stem and progenitor cell isolation. Fisher 344 adult rats were sacrificed by CO2 asphyxiation followed by cervical dislocation. Whole brains were removed intact using bone rongeurs and hippocampus tissue dissected based on coronal incisions posterior to the motor and somatosensory regions of the brain (Paxinos, G. & Watson, C. 1997. The Rat Brain in Stereotaxic Coordinates). Tissue was washed in Neurobasal-A medium (Invitrogen, Carlsbad, Calif.) containing B27 (B27 supplement; Invitrogen), L-glutamine (4 mM; Invitrogen), and penicillin/streptomycin (Invitrogen), the combination of which is herein referred to as Neural Progenitor Expansion (NPE) medium. NPE medium was further supplemented with bFGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ) and EGF (20 nanograms/milliliter, Peprotech, Rocky Hill, NJ), herein referred to as NPE+bFGF+EGF.
  • Following wash, the overlying meninges were removed, and the tissue minced with a scalpel. Minced tissue was collected and trypsin/EDTA (Invitrogen) added as 75% of the total volume. DNAse (100 microliters per 8 milliliters total volume, Sigma, St. Louis, Mo.) was also added. Next, the tissue/media was sequentially passed through an 18 gauge needle, 20 gauge needle, and finally a 25 gauge needle one time each (all needles from Becton Dickinson, Franklin Lakes, N.J.). The mixture was centrifuged for 3 minutes at 250 g. Supernatant was removed, fresh NPE+bFGF+EGF was added and the pellet resuspended. The resultant cell suspension was passed through a 40 micrometer cell strainer (Becton Dickinson), plated on laminin-coated T-75 flasks (Becton Dickinson) or low cluster 24-well plates (Becton Dickinson), and grown in NPE+bFGF+EGF media until sufficient cell numbers were obtained for the studies outlined.
  • PPDC plating. Postpartum-derived cells (umbilicus (022803) P12, (042103) P12, (071003) P12; placenta (042203) P12) previously grown in Growth Medium were plated at 5,000 cells/transwell insert (sized for 24 well plate) and grown for a period of one week in Growth Medium in inserts to achieve confluence.
  • Adult neural progenitor plating. Neural progenitors, grown as neurospheres or as single cells, were seeded onto laminin-coated 24 well plates at an approximate density of 2,000 cells/well in NPE+bFGF+EGF for a period of one day to promote cellular attachment. One day later, transwell inserts containing postpartum cells were added according to the following scheme:
  • (1) Transwell (umbilicus-derived cells in Growth Media, 200 microliters)+neural progenitors (NPE+bFGF+EGF, 1 milliliter)
    • (2) Transwell (placenta-derived cells in Growth Media, 200 microliters)+neural progenitors (NPE+bFGF+EGF, 1 milliliter)
  • (3) Transwell (adult human dermal fibroblasts [1F1853; Cambrex, Walkersville, MD] P12 in Growth Media, 200 microliters)+neural progenitors (NPE+bFGF+EGF, 1 milliliter)
    • (4) Control: neural progenitors alone (NPE+bFGF+EGF, 1 milliliter)
    • (5) Control: neural progenitors alone (NPE only, 1 milliliter)
  • Immunocytochemistry. After 7 days in co-culture, all conditions were fixed with cold 4% (w/v) paraformaldehyde (Sigma) for a period of 10 minutes at room temperature. Immunocytochemistry was performed using antibodies directed against the epitopes listed in Table 15-1. Briefly, cultures were washed with phosphate-buffered saline (PBS) and exposed to a protein blocking solution containing PBS, 4% (v/v) goat serum (Chemicon, Temecula, CA), and 0.3% (v/v) Triton (Triton X-100; Sigma) for 30 minutes to access intracellular antigens. Primary antibodies, diluted in blocking solution, were then applied to the cultures for a period of 1 hour at room temperature. Next, primary antibodies solutions were removed and cultures washed with PBS prior to application of secondary antibody solutions (1 hour at room temperature) containing blocking solution along with goat anti-mouse IgG—Texas Red (1:250; Molecular Probes, Eugene, Oreg.) and goat anti-rabbit IgG—Alexa 488 (1:250; Molecular Probes). Cultures were then washed and 10 micromolar DAPI (Molecular Probes) applied for 10 minutes to visualize cell nuclei.
  • Following immunostaining, fluorescence was visualized using the appropriate fluorescence filter on an Olympus inverted epi-fluorescent microscope (Olympus, Melville, NY). In all cases, positive staining represented fluorescence signal above control staining where the entire procedure outlined above was followed with the exception of application of a primary antibody solution. Representative images were captured using a digital color videocamera and ImagePro software (Media Cybernetics, Carlsbad, Calif.). For triple-stained samples, each image was taken using only one emission filter at a time. Layered montages were then prepared using Adobe Photoshop software (Adobe, San Jose, Calif.).
    TABLE 15-1
    Summary of Primary Antibodies Used
    Antibody Concentration Vendor
    Rat 401 (nestin) 1:200 Chemicon, Temecula,
    CA
    TuJ1 (BIII Tubulin) 1:500 Sigma, St. Louis, MO
    Tyrosine hydroxylase (TH)  1:1000 Chemicon
    GABA 1:400 Chemicon
    GFAP  1:2000 DakoCytomation,
    Carpinteria, CA
    Myelin Basic Protein (MBP) 1:400 Chemicon
  • Quantitative analysis of neural progenitor differentiation. Quantification of hippocampal neural progenitor differentiation was examined. A minimum of 1000 cells were counted per condition or if less, the total number of cells observed in that condition. The percentage of cells positive for a given stain was assessed by dividing the number of positive cells by the total number of cells as determined by DAPI (nuclear) staining.
  • Mass spectrometry analysis & 2D gel electrophoresis. In order to identify unique, secreted factors as a result of co-culture, conditioned media samples taken prior to culture fixation were frozen down at −80° C. overnight. Samples were then applied to ultrafiltration spin devices (MW cutoff 30 kD). Retentate was applied to immunoaffinity chromatography (anti-Hu-albumin; IgY) (immunoaffinity did not remove albumin from the samples). Filtrate was analyzed by MALDI. The pass through was applied to Cibachron Blue affinity chromatography. Samples were analyzed by SDS-PAGE and 2D gel electrophoresis.
  • Results
  • PPDC co-culture stimulates adult neural progenitor differentiation. Following culture with umbilicus- or placenta-derived cells, co-cultured neural progenitor cells derived from adult rat hippocampus exhibited significant differentiation along all three major lineages in the central nervous system. This effect was clearly observed after five days in co-culture, with numerous cells elaborating complex processes and losing their phase bright features characteristic of dividing progenitor cells. Conversely, neural progenitors grown alone in the absence of bFGF and EGF appeared unhealthy and survival was limited.
  • After completion of the procedure, cultures were stained for markers indicative of undifferentiated stem and progenitor cells (nestin), immature and mature neurons (TuJ1), astrocytes (GFAP), and mature oligodendrocytes (MBP). Differentiation along all three lineages was confirmed while control conditions did not exhibit significant differentiation as evidenced by retention of nestin-positive staining amongst the majority of cells. While both umbilicus- and placenta-derived cells induced cell differentiation, the degree of differentiation for all three lineages was less in co-cultures with placenta-derived cells than in co-cultures with umbilicus-derived cells.
  • The percentage of differentiated neural progenitors following co-culture with umbilicus-derived cells was quantified (Table 15-2). Umbilicus-derived cells significantly enhanced the number of mature oligodendrocytes (MBP) (24.0% vs 0% in both control conditions). Furthermore, co-culture enhanced the number of GFAP+astrocytes and TuJ1+neurons in culture (47.2% and 8.7% respectively). These results were confirmed by nestin staining indicating that progenitor status was lost following co-culture (13.4% vs 71.4% in control condition 4).
  • Though differentiation also appeared to be influenced by adult human fibroblasts, such cells were not able to promote the differentiation of mature oligodendrocytes nor were they able to generate an appreciable quantity of neurons. Though not quantified, fibroblasts did, however, appear to enhance the survival of neural progenitors.
    TABLE 15-2
    Quantification of progenitor differentiation in control vs transwell
    co-culture with umbilical-derived cells (E = EGF, F = bFGF)
    F + E/Umb F + E/F + E F + E/removed
    Antibody [Cond. 1] [Cond. 4] [Cond. 5]
    TuJ1  8.7%  2.3%  3.6%
    GFAP 47.2% 30.2% 10.9%
    MBP 23.0%   0%   0%
    Nestin 13.4% 71.4% 39.4%
  • Identification of unique compounds. Conditioned media from umbilicus- and placenta-derived co-cultures, along with the appropriate controls (NPE media±1.7% serum, media from co-culture with fibroblasts), were examined for differences. Potentially unique compounds were identified and excised from their respective 2D gels.
  • Summary. Co-culture of adult neural progenitor cells with umbilicus or placenta PPDCs results in differentiation of those cells. Results presented in this example indicate that the differentiation of adult neural progenitor cells following co-culture with umbilicus-derived cells is particularly profound. Specifically, a significant percentage of mature oligodendrocytes was generated in co-cultures of umbilicus-derived cells. In view of the lack of contact between the umbilicus-derived cells and the neural progenitors, this result appears to be a function of soluble factors released from the umbilicus-derived cells (trophic effect).
  • Several other observations were made. First, there were very few cells in the control condition where EGF and bFGF were removed. Most cells died and on average, there were about 100 cells or fewer per well. Second, it is to be expected that there would be very little differentiation in the control condition where EGF and bFGF was retained in the medium throughout, since this is normally an expansion medium. While approximately 70% of the cells were observed to retain their progenitor status (nestin+), about 30% were GFAP+(indicative of astrocytes). This may be due to the fact that such significant expansion occurred throughout the course of the procedure that contact between progenitors induced this differentiation (Song, H. et al. 2002. Nature 417: 29-32).
  • EXAMPLE 16 Transplantation of Postpartum-Derived Cells
  • Cells derived from the postpartum umbilicus and placenta are useful for regenerative therapies. The tissue produced by postpartum-derived cells (PPDCs) transplanted into SCID mice with a biodegradable material was evaluated. The materials evaluated were Vicryl non-woven, 35/65 PCL/PGA foam, and RAD 16 self-assembling peptide hydrogel.
  • Methods & Materials
  • Cell Culture. Placenta- and umbilicus-derived cells were grown in Growth Medium (DMEM-low glucose (Gibco, Carlsbad Calif.), 15% (v/v) fetal bovine serum (Cat. #SH30070.03; Hyclone, Logan, Utah), 0.001% (v/v) betamercaptoethanol (Sigma, St. Louis, Mo.), penicillin/streptomycin (Gibco)) in a gelatin-coated flasks.
  • Sample Preparation. One million viable cells were seeded in 15 microliters Growth Medium onto 5 mm diameter, 2.25 mm thick Vicryl non-woven scaffolds (64.33 milligrams/cc; Lot#3547-47-1) or 5 mm diameter 35/65 PCL/PGA foam (Lot# 3415-53). Cells were allowed to attach for two hours before adding more Growth Medium to cover the scaffolds. Cells were grown on scaffolds overnight. Scaffolds without cells were also incubated in medium.
  • RAD16 self-assembling peptides (3D Matrix, Cambridge, Mass. under a material transfer agreement) was obtained as a sterile 1% (w/v) solution in water, which was mixed 1:1 with 1×106 cells in 10% (w/v) sucrose (Sigma, St Louis, Mo.), 10 mM HEPES in Dulbecco's modified medium (DMEM; Gibco) immediately before use. The final concentration of cells in RAD16 hydrogel was 1×106 cells/100 microliters.
  • TEST MATERIAL (N=4/Rx)
  • 1. Vicryl non-woven+1×106 umbilicus-derived cells
  • 2. 35/65 PCL/PGA foam+1×106 umbilicus-derived cells
  • 3. RAD 16 self-assembling peptide+1×106 umbilicus-derived cells
  • 4. Vicryl non-woven+1×106 placenta-derived cells
  • 5. 35/65 PCL/PGA foam+1×106 placenta-derived cells
  • 6. RAD 16 self-assembling peptide+1×106 placenta-derived cells
  • 7. 35/65 PCL/PGA foam
  • 8. Vicryl non-woven
  • Animal Preparation. The animals were handled and maintained in accordance with the current requirements of the Animal Welfare Act. Compliance with the above Public Laws were accomplished by adhering to the Animal Welfare regulations (9 CFR) and conforming to the current standards promulgated in the Guide for the Care and Use of Laboratory Animals, 7th edition.
  • Mice (Mus Musculus)/Fox Chase SCID/Male (Harlan Sprague Dawley, Inc., Indianapolis, Ind.), 5 weeks of age. All handling of the SCID mice took place under a hood. The mice were individually weighed and anesthetized with an intraperitoneal injection of a mixture of 60 milligrams/kg KETASET (ketamine hydrochloride, Aveco Co., Inc., Fort Dodge, Iowa) and 10 milligrams/kg ROMPUN (xylazine, Mobay Corp., Shawnee, Kansas) and saline. After induction of anesthesia, the entire back of the animal from the dorsal cervical area to the dorsal lumbosacral area was clipped free of hair using electric animal clippers. The area was then scrubbed with chlorhexidine diacetate, rinsed with alcohol, dried, and painted with an aqueous iodophor solution of 1% available iodine. Ophthalmic ointment was applied to the eyes to prevent drying of the tissue during the anesthetic period.
  • Subcutaneous Implantation Technique. Four skin incisions, each approximately 1.0 cm in length, were made on the dorsum of the mice. Two cranial sites were located transversely over the dorsal lateral thoracic region, about 5-mm caudal to the palpated inferior edge of the scapula, with one to the left and one to the right of the vertebral column. Another two were placed transversely over the gluteal muscle area at the caudal sacro-lumbar level, about 5-mm caudal to the palpated iliac crest, with one on either side of the midline. Implants were randomly placed in these sites in accordance with the experimental design. The skin was separated from the underlying connective tissue to make a small pocket and the implant placed (or injected for RAD16) about 1-cm caudal to the incision. The appropriate test material was implanted into the subcutaneous space. The skin incision was closed with metal clips.
  • Animal Housing. Mice were individually housed in microisolator cages throughout the course of the study within a temperature range of 64° F.-79° F. and relative humidity of 30% to 70%, and maintained on an approximate 12 hour light/12 hour dark cycle. The temperature and relative humidity were maintained within the stated ranges to the greatest extent possible. Diet consisted of Irradiated Pico Mouse Chow 5058 (Purina Co.) and water fed ad libitum.
  • Mice were euthanized at their designated intervals by carbon dioxide inhalation. The subcutaneous implantation sites with their overlying skin were excised and frozen for histology.
  • Histology. Excised skin with implant was fixed with 10% neutral buffered formalin (Richard-Allan Kalamazoo, Mich.). Samples with overlying and adjacent tissue were centrally bisected, paraffin-processed, and embedded on cut surface using routine methods. Five-micron tissue sections were obtained by microtome and stained with hematoxylin and eosin (Poly Scientific Bay Shore, NY) using routine methods.
  • Results
  • There was minimal ingrowth of tissue into foams (without cells) implanted subcutaneously in SCID mice after 30 days. In contrast there was extensive tissue fill in foams implanted with umbilical-derived cells or placenta-derived cells. Some tissue ingrowth was observed in Vicryl non-woven scaffolds. Non-woven scaffolds seeded with umbilicus- or placenta-derived cells showed increased matrix deposition and mature blood vessels.
  • Summary. Synthetic absorbable non-woven/foam discs (5.0 mm diameter×1.0 mm thick) or self-assembling peptide hydrogel were seeded with either cells derived from human umbilicus or placenta and implanted subcutaneously bilaterally in the dorsal spine region of SCID mice. The results demonstrated that postpartum-derived cells could dramatically increase good quality tissue formation in biodegradable scaffolds.
  • EXAMPLE 17 Use of Postpartum-Derived Cells in Nerve Repair
  • Retinal ganglion cell (RGC) lesions have been extensively used as models for various repair strategies in the adult mammalian CNS. It has been demonstrated that retrobulbar section of adult rodent RGC axons results in abortive sprouting (Zeng et al., 1995) and progressive death of the parent cell population (Villegas-Perez et al., 1993). Numerous studies have demonstrated the stimulatory effects of various exogenous and endogenous factors on the survival of axotomized RGC's and regeneration of their axons (Yip and So, 2000; Fischer et al., 2001). Furthermore, other studies have demonstrated that cell transplants can be used to promote regeneration of severed nerve axons (L1 et al., 2003; Ramon-Cueto et al., 2000). Thus, these and other studies have demonstrated that cell based therapy can be utilized for the treatment of neural disorders that affect the spinal cord, peripheral nerves, pudendal nerves, optic nerves or other diseases/trauma due to injury in which nervous damage can occur.
  • Self-assembling peptides (PuraMatrix™, U.S. Pat. Nos. 5,670,483, 5,955,343, US/PCT applications US2002/0160471, WO02/062969) have been developed to act as a scaffold for cell-attachment to encapsulate cells in 3-D, plate cells in 2-D coatings, or as microcarriers in suspension cultures. Three-dimensional cell culture has required either animal-derived materials (mouse sarcoma extract), with their inherent reproducibility and cell signaling issues, or much larger synthetic scaffolds, which fail to approximate the physical nanometer-scale and chemical attributes of native ECM. RAD 16 (NH2-(RADA)3-COOH) and KLD (NH2-(KLDL)3-COOH) are synthesized in small (RAD16 is 5 nanometers) oligopeptide fragments that self-assemble into nanofibers on a scale similar to the in vivo extracellular matrix (ECM) (3D Matrix, Inc Cambridge, Mass.). The self-assembly is initiated by mono- or di-valent cations found in culture media or the physiological environment. In the protocols described in this example, RAD 16 was used as a microcarrier for the implantation of postpartum cells into the ocular defect. In this example, it is demonstrated that transplants of postpartum-derived cells PPDCs) can provide efficacy in an adult rat optic nerve axonal regeneration model.
  • Methods & Materials
  • Cells. Cultures of human adult PPDCs (umbilicus and placenta) and fibroblast cells (passage 10) were expanded for 1 passage. All cells were initially seeded at 5,000 cells/cm2 on gelatin-coated T75 flasks in Growth Medium with 100 Units per milliliter penicillin, 100 micrograms per milliliter streptomycin, 0.25 micrograms per milliliter amphotericin B (Invitrogen, Carlsbad, Calif.). At passage 11 cells were trypsinized and viability was determined using trypan blue staining. Briefly, 50 microliters of cell suspension was combined with 50 microliters of 0.04% w/v trypan blue (Sigma, St. Louis Mo.) and the viable cell number, was estimated using a hemocytometer. Cells were then washed three times in supplement free-Leibovitz's L-15 medium (Invitrogen, Carlsbad, Calif.). Cells were then suspended at a concentration of 200,000 cells in 25 microliters of RAD-16 (3DM Inc., Cambridge, Mass.) which was buffered and made isotonic as per manufacturer's recommendations. One hundred microliters of supplement free Leibovitz's L-15 medium was added above the cell/matrix suspension to keep it wet till use. These cell/matrix cultures were maintained under standard atmospheric conditions until transplantation occurred. At the point of transplantation the excess medium was removed.
  • Animals and Surgery. Long Evans female rats (220-240 gram body weight) were used. Under intraperitoneal tribromoethanol anesthesia (20 milligram/100 grams body weight), the optic nerve was exposed, and the optic sheath was incised intraorbitally at approximately 2 millimeters from the optic disc, the nerve was lifted from the sheath to allow complete transsection with fine scissors (L1 et al., 2003). The completeness of transsection was confirmed by visually observing complete separation of the proximal and distal stumps. The control group consisted of lesioned rats without transplants. In transplant rats cultured postpartum cells seeded in RAD-16 were inserted between the proximal and distal stumps using a pair of microforceps. Approximately 75,000 cells in RAD-16 were implanted into the severed optic nerve. Cell/matrix was smeared into the severed cut using a pair of fine microforceps. The severed optic nerve sheath was closed with 10/0 black monofilament nylon (Ethicon, Inc., Edinburgh, UK). Thus, the gap was closed by drawing the cut proximal and distal ends of the nerve in proximity with each other.
  • After cell injections were performed, animals were injected with dexamethasone (2 milligrams/kilogram) for 10 days post transplantation. For the duration of the study, animals were maintained on oral cyclosporine A (210 milligrams/liter of drinking water; resulting blood concentration: 250-300 micrograms/liter) (Bedford Labs, Bedford, Ohio) from 2 days pre-transplantation until end of the study. Food and water were available ad libitum. Animals were sacrificed at either 30 or 60 days posttransplantation.
  • CTB Application. Three days before animals were sacrificed, under anesthesia, a glass micropipette with a 30-50 millimeter tip was inserted tangentially through the sclera behind the lens, and two 4-5 microliter aliquots of a 1% retrograde tracer-cholera toxin B (CTB) aqueous solution (List Biologic, Campbell, CA) was injected into the vitreous. Animals were perfused with fixative and optic nerves were collected in the same fixative for 1 hour. The optic nerves were transferred into sucrose overnight. Twenty micrometer cryostat sections were incubated in 0.1 molar glycine for 30 minutes and blocked in a PBS solution containing 2.5% bovine serum albumin (BSA) (Boeringer Mannheim, Mannheim, Germany) and 0.5% triton X-100 (Sigma, St. Louis, Mo.), followed by a solution containing goat anti-CTB antibody (List Biologic, Campbell, CA) diluted 1:4000 in a PBS containing 2% normal rabbit serum (NRS) (Invitrogen, Carlsbad, Calif.), 2.5% BSA, and 2% Triton X-100 (Sigma, St. Louis, Mo.) in PBS, and incubated in biotinylated rabbit anti-goat IgG antibody (Vector Laboratories, Burlinghame, CA) diluted 1:200 in 2% Triton-X100 in PBS for 2 hours at room temperature. This was followed by staining in 1:200 streptavidin-green (Alexa Flour 438;Molecular Probes, Eugene, Oreg.) in PBS for 2 hours at room temperature. Stained sections were then washed in PBS and counterstained with propidium iodide for confocal microscopy.
  • Histology Preparation. Briefly, 5 days after CTB injection, rats were perfused with 4% paraformaldehyde. Rats were given 4 cubic centimeters of urethane and were then perfused with PBS (0.1 molar) then with 4% Para formaldehyde. The spinal cord was cut and the bone removed from the head to expose the colliculus. The colliculus was then removed and placed in 4% paraformaldehyde. The eye was removed by cutting around the outside of the eye and going as far back as possible. Care was given not to cut the optic nerve that lies on the underside of the eye. The eye was removed and the muscles were cut exposing the optic nerve this was then placed in 4% paraformaldehyde.
  • Results
  • Lesions alone. One month after retrotubular section of the optic nerve, a number of CTB-labeled axons were identified in the nerve segment attached to the retina. In the 200 micrometers nearest the cut, axons were seen to emit a number of collaterals at right angles to the main axis and terminate as a neuromatous tangle at the cut surface. In this cut between the proximal and distal stumps, the gap was observed to be progressively bridged by a 2-3 millimeter segment of vascularized connective tissue; however, no axons were seen to advance into this bridged area. Thus, in animals that received lesion alone no axonal growth was observed to reach the distal stump.
  • RAD-16 transplantation. Following transplantation of RAD-16 into the cut, visible ingrowth of vascularized connective tissue was observed. However, no axonal in growth was observed between the proximal and distal stumps. The results demonstrate that application of RAD-16 alone is not sufficient for inducing axonal regeneration in this situation.
  • Transplantation of postpartum-derived cells. Transplantation of postpartum-derived cells into the severed optic nerve stimulated optic nerve regrowth. Some regrowth was also observed in conditions in which fibroblast cells were implanted, although this was minimal as compared with the regrowth observed with the transplanted placenta-derived cells. Optic nerve regrowth was observed in 4/5 animals transplanted with placenta-derived cells, 3/6 animals transplanted with adult dermal fibroblasts and in ¼ animals transplanted with umbilicus-derived cells. In situations where regrowth was observed, CTB labeling confirmed regeneration of retinal ganglion cell axons, which were demonstrated to penetrate through the transplant area. GFAP labeling was also performed to determine the level of glial scarring. The GFAP expression was intensified at the proximal stump with some immunostaining being observed through the reinervated graft.
  • Summary. These results demonstrate that transplanted human adult postpartum-derived cells are able to stimulate and guide regeneration of cut retinal ganglion cell axons.
  • References for Example 17
    • 1) Zeng B Y, Anderson P N, Campbell G, Lieberman A R. 1995. J. Anat.186:495-508.
    • 2) Villegas-Perez M P, Vidal-Sanz M, Bray G M, Aguayo A J. 1988. J. Neurosci.8:265-80.
    • 3) Yip H K, So K F. 2000. Prog Retin Eye Res. 19: 559-75.
    • 4) Fischer D, Heiduschka P, Thanos S. 2001. Exp Neurol. 172: 257-72.
    • 5) Ramon-Cueto A, Cordero M I, Santos-Benito F F, Avila J. 2000. Neuron 25: 425-35.
    EXAMPLE 18 Use of Postpartum-Derived Cells in the Treatment of Retinitis Pigmentosa
  • Currently no real treatment exists for blinding disorders that stem from the degeneration of cells in the retina. Loss of photoreceptors as a result of apoptosis or secondary degeneration lead to progressive deterioration of vision, and ultimately to blindness. Diseases in which this occurs include age-related macular degeneration (AMD) and retinitis pigmentosa (RP). RP is most commonly associated with a single gene mutation, which contributes to photoreceptor cell death.
  • The retinal photoreceptors and adjacent retinal pigment epithelium form a functional unit. The Royal College of Surgeons (RCS) rat presents with a tyrosine receptor kinase (Merkt) defect affecting outer segment phagocytosis, leading to photoreceptor cell death (1). Transplantation of retinal pigment epithelial (RPE) cells into the subretinal space of RCS rats was found to limit the progress of photoreceptor loss and preserve visual function (2). In this example, it is demonstrated that postpartum-derived cells can be used to promote photoreceptor rescue and thus preserve photoreceptors in an RCS model.
  • Methods & Materials
  • Cell transplants. Cultures of human adult umbilical, placental and fibroblast cells (passage 10) were expanded for 1 passage. All cells were initially seeded at 5,000 cells/cm2 on gelatin-coated T75 flasks in Growth Medium. For subsequent passages, all cells were treated as follows. After trypsinization, viable cells were counted after trypan blue staining. Briefly, 50 microliters of cell suspension was combined with 50 microliters of 0.04% w/v trypan blue (Sigma, St. Louis Mo.) and the viable cell number, was estimated using a heamocytometer. Cells were trypsinized and washed three times in supplement free—DMEM:Low glucose medium (Invitrogen, Carlsbad, Calif.). Cultures of human umbilical placental and fibroblast cells at passage 11 were trypsinized and washed twice in Leibovitz's L-15 medium (Invitrogen, Carlsbad, Calif.). For the transplantation procedure, dystrophic RCS rats were anesthetized with xylazine-ketamine (1 mg/kg i.p. of the following mixture: 2.5 ml xylazine at 20 mg/ml, 5 ml ketamine at 100 mg/ml, and 0.5 ml distilled water) and their heads secured by a nose bar. Cells devoid of serum were resuspended (2×105 cells per injection) in 2 microliters of Leibovitz, L-15 medium (Invitrogen, Carlsbad, Calif.) and transplanted using a fine glass pipette (internal diameter 75-150 micrometers) trans-scerally. Cells were delivered into the dorso-temporal subretinal space of anesthetized 3 week old dystrophic-pigmented RCS rats (total N=10/cell type).
  • Cells were injected unilaterally into the right eye, while the left eye was injected with carrier medium alone (Sham control; Leibovitz's L-15 medium). Viability of residual transplant cells remained at greater than 95% as assessed by trypan blue exclusion at the end of the transplant session. After cell injections were performed, animals were injected with dexamethasone (2 mg/kg) for 10 days post transplantation. For the duration of the study, animals were maintained on oral cyclosporine A (210 mg/L of drinking water; resulting blood concentration: 250-300 micrograms/L) (Bedford Labs, Bedford, Ohio) from 2 days pre-transplantation until end of the study. Food and water were available ad libitum. Animals were sacrificed at 60 or 90 days postoperatively, with some animals being sacrificed at earlier timepoints for histological assessment of short-term changes associated with cell transplantation.
  • ERG recordings. Following overnight dark adaptation, animals were prepared for ERG recording under dim red light, as previously described (3). In brief, under anesthesia (with a mixture of 150 mg/kg i.p ketamine, and 10 mg/kg i.p. xylazine) the head was secured with a stereotaxic head holder and the body temperature monitored through a rectal thermometer and maintained at 38° C. using a homeothermic blanket. Pupils were dilated using equal parts of topical 2.5% phenylephrine and 1% tropicamide. Topical anesthesia with 0.75% bupivacaine was used to prevent any corneal reflexes and a drop of 0.9% saline was frequently applied on the cornea to prevent its dehydration and allow electrical contact with the recording electrode (gold wire loop). A 25-gauge needle inserted under the scalp, between the two eyes, served as the reference electrode. Amplification (at 1-1000 Hz bandpass, without notch filtering), stimulus presentation, and data acquisition were provided by the UTAS-3000 system from LKC Technologies (Gaithersburg, Md.). ERGs were recorded at 60 and 90 days of age in the umbilical cell groups and at 60 days only in the placental and fibroblast groups.
  • Mixed a- and b-wave recording. For the quantification of dark-adapted b-waves, recordings consisted of single flash presentations (10 lsec duration), repeated 3 to 5 times to verify the response reliability and improve the signal-to-noise ratio, if required. Stimuli were presented at six increasing intensities in one log unit steps varying from −3.6 to 1.4 log candila/m2 in luminance. To minimize the potential bleaching of rods, inter-stimulus intervals were increased as the stimulus luminance was elevated from 10 sec at lowest stimulus intensity to 2 minutes at highest stimulus intensity. The maximum b-wave amplitude was defined as that obtained from the flash intensity series, regardless of the stimulus intensity. The true Vmax from fitting the data with a Naka-Rushton curve was not used because ERG responses were often erratic at higher luminance levels in dystrophic animals and showed tendencies for depressed responses around 0.4 and 1.4 log candila/m2. In order to determine the age at which ERG components were obtained or lost, criterion amplitudes were used: 20 μV for a- and b-waves, and 10 μV for STR-like responses. The amplitude of the b-wave was measured from the a-wave negative peak up to the b-wave positive apex, and not up to the peak of oscillations, which can exceed the b-wave apex (4).
  • Isolation of rod and cone responses. The double flash protocol was used to determine the isolation of rod and cone responses (5). A probe flash was presented 1 sec after a conditioning flash, using a specific feature of the UTAS-3000 system (LKC Technologies) with calibrated ganzfeld; assuring complete recharge of the stimulator under the conditions used. The role of the conditioning flash in the procedure was to transiently saturate rods so that they were rendered unresponsive to the probe flash. Response to the probe flash was taken as reflecting cone-driven activity. A rod-driven b-wave was obtained by subtracting the cone-driven response from the mixed response (obtained following presentation of a probe flash alone, i.e. not preceded by any conditioning flash).
  • Functional Assessment. Physiological retinal sensitivity testing was performed to demonstrate retinal response to dim light. Animals were anesthetized with a recovery dose of urethane at 1.25 g/kg i.p. Physiological assessment in the animals was tested post graft in animals at 90 days by recording multiunit extracellular activity in the superior colliculus to illumination of respective visual receptive fields (6). This procedure was repeated for 20 independent points (spaced 200 mm apart, with each step corresponding to approximately 10-150 displacements in the visual field), covering the visual field. Visual thresholds were measured as the increase in intensity over background and maintained at 0.02 candila/m2 (luminescence unit) [at least 2.6 logarithm units below rod saturation (7)], required for activating units in the superficial 200 μm of the superior colliculus. Response parameters were compared between transplanted and sham control eyes that received vehicle alone.
  • Histology. Animals were sacrificed with an overdose of urethane (12.5 g/kg). The orientation of the eye was maintained by placing a 6.0 suture through the superior rectus muscle prior to enucleation. After making a corneal incision, the eyes were fixed with 2.5% parafomaldehyde, 2.5% glutaraldehyde, 0.01% picric acid in 0.1 M cacodylate buffer (pH7.4) (Sigma, St. Louis, Mo.). After fixation, the cornea and lens were removed by cutting around the ciliary body. A small nick was made in the periphery of the dorsal retina prior to removal of the superior rectus to assist in maintaining orientation. The retinas were then post-fixed in 1% osmium tetroxide for 1 h. After dehydration through a series of alcohols to epoxypropane, the retinas were embedded in TAAB embedding resin (TAAB Laboratories, Aldemarston, UK). Semi-thin sections were stained with 1% toluidine Blue in 1% borate buffer and the ultra thin sections were contrasted with uranyl acetate and lead citrate.
  • For Nissl staining, sections were stained with 0.75% cresyl violet (Sigma, St. Louis, Mo.) after which they were dehydrated through graded alcohols at 70, 95 and 100% twice. They were then placed in xylene (Sigma, St. Louis, Mo.), rinsed with PBS (pH 7.4) (Invitrogen, Carlsbad, Calif.), coverslipped and mounted with DPX mountant (Sigma, St. Louis, Mo.).
  • Results
  • ERG Recordings. Animals that received umbilical cell injections exhibited relative preservation of visual response properties 60 and 90 days post-operatively (Table 18-1). The response observed in these animals was greater than that seen with placental, fibroblast or sham treated animals. Placental cell transplants (n=4) at 60 days showed no improvement in a-wave (20±20) versus sham controls (0), but showed some improvement in mixed b-wave (81±72) versus sham controls (1.5±2), and good improvement in cone-b-wave (50±19) versus sham controls (7±7), and in rod contribution (30%) versus sham controls (O). These results indicated some improvement in visual responsiveness when compared to sham controls.
  • Umbilical cell-transplanted animals (n=6) demonstrated good improvement in all outcome measures tested at 60 days (Table N5-1), a-wave (27±11) versus sham controls (O), mixed b-wave (117±67) versus sham controls (18±13), cone-b-wave (55±25) versus sham controls (28±11), and in rod contribution (49±16%) versus sham controls (6±7%). Furthermore, at 90 days, improved responses were measured in two animals tested, with measures including: a-wave (15±7) versus sham controls (O), mixed b-wave (37±15) versus sham controls (O), cone-b-wave (16±11) versus sham controls (7±5), and in rod contribution (58±39%) versus sham controls (0%). These results indicate that visual responsiveness was improved in umbilical cell transplanted animals with evidence for photoreceptor preservation in the RCS model. Although a diminution in responsiveness to ERG was observed in the 90 day animals tested, their preservation of visual function in comparison to sham-treated controls was good.
  • In contrast to either umbilical or placental cells, fibroblast transplantations showed no improvement in any of the parameters tested, with values less than or equal to sham-treated controls.
    TABLE 18-1
    ERG data
    a-wave mixed b-wave cone b-wave % rod contribution
    Group Untreated Treated Untreated Treated Untreated Treated Untreated Treated
    Sham 60 d 0 0 7 ± 9 0 23 ± 5  12 ± 16 N/A N/A
    1 (n = 4) 60 d 0 20 ± 20 1.5 ± 2   81 ± 72 7 ± 7 50 ± 19 N/A 30
    3 (n = 6) 60 d 0 27 ± 11 18 ± 13 117 ± 67  28 ± 11 55 ± 25 6 ± 7 49 ± 16
    3 (n = 6) 90 d 0 15 ± 7  0 37 ± 15 7 ± 5 16 ± 11 0 58 ± 39

    N.B. Sham = control (medium only),

    1 = Placental cell transplant,

    3 = Umbilical cell transplant
  • Histology. Following transplantation, there was no histological evidence of an inflammatory reaction and infiltrating immune cells were not observed in Nissl-stained sections in the postpartum cell groups. However, fibroblast implantations resulted in animal death (n=7) and indications of early stage inflammatory responses. Histologically at the 90 day time point in the umbilical transplanted animals anatomical rescue of photoreceptors was clearly demonstrated. The photoreceptors formed a thick layer separated by a gap from the inner nuclear layer, made up of other retinal cells. By comparison, the width of the outer layer in the sham control was, at best, a discontinuous single layer as opposed to around 5 cells thick in the grafted eye. In comparison to a normal animal this is marginally more than half the thickness of photoreceptor cell layers normally observed.
  • Functional Assessment. Efficacy of transplants in preventing visual loss was monitored by assessment of electrophysiological responsiveness in two animals. The threshold sensitivity response to light was used to define the area of visual field rescue in sham-injected control eyes versus eyes transplanted with umbilical cells. In nondystrophic rats, visual thresholds never exceeded 0.5 log candila/m2 above background. In non-operated dystrophic rats, the thresholds are usually in the magnitude of 4 log candila/m2 units (8). By contrast, in non-operated sham injected dystrophic rats, the thresholds were in the order of 2.9-4.9 log candila/m2 units with an average threshold of 4.0 log candila/m2 units, in some instances no recording could be attained. Thus, the sham-injected rats showed some highly localized functional rescue in the temporal retina. However, the human umbilical transplanted rats exhibited substantially greater levels of visual preservation with thresholds ranging from 0.8 to 2.1 log candila/m2 units, with an average threshold of 1.3 log candila/m2 units.
  • Summary. Transplantation of umbilical cells into dystrophic RCS rats was shown to preserve photoreceptors. To a lesser degree this response was also seen following transplantation with placental cells although improvement was seen in a-wave responsiveness was not observed. In this degenerative model, one would expect the a-wave to disappear within 30 to 60 days and the b-wave to disappear within 3 months. Thus, basically retained a-wave indicates that real and normal rod function is preserved. Rod contribution to b-wave suggests abnormal rod function is still possible. The sustained non-rod b-wave is the measure of how much cone function is maintained, which is a real measure of vision. Thus, the level of improvement assessed both physiologically and anatomically following umbilical transplantation is well defined here. ERG measurements provide an assessment of visual function after photoreceptor loss, indicating changes in electrical activity in the retina. However, ERG does not provide direct information as to image forming capability. The measurement of collicular threshold sensitivity used in this study provides an indication of relative preservation of visual fields. The importance of this measure is based on a correlation between the amounts of functional rescue and anatomical preservation and that the data collected compares with visual field perimetry testing in humans (9). The transplantation has demonstrated a retardation of the disease process in the test animals. Thus, the results presented herein demonstrate clear evidence of functional efficacy of grafting human PPDCs into the subretinal space, and that rescue occurs in the general region in which the grafted cells are located.
  • References for Example 18
    • 1. D'Cruz P. M. et al., 2000, Hum Mol Genet. 9: 645-651.
    • 2. Li, L. X. & Turner, J. E., 1988, Exp Eye Res. 47: 911-917.
    • 3. Sauve, Y. et al., 2004, Vision Res. 44: 9-18
    • 4. Nusinowitz, S. et al., 1999, Ridder, Invest Ophthalmol Vis Sci. 40: 2848-2858.
    • 5. Nixon, P. J. et al., 2001, Clin Experiment Ophthalmol. 29:193-196.
    • 6. Lund, R. D. et al., 2001, Proc. Natl. Acad. Sci. USA. 98: 9942-9947.
    • 7. Siminoff, R. & Kruger, L., 1968, Exp Neurol. 20: 403-414.
    • 8. Blakema, G. W. & Drager, U. C., 1991. Visual Neuroscience. 6: 577-585.
    • 9. Beck, R. W. et al., 1985, Ophthalmology 92: 77-82.
  • The present invention is not limited to the embodiments described and exemplified above. It is capable of variation and modification within the scope of the appended claims.

Claims (48)

1. An isolated postpartum-derived cell comprising a cell derived from human placental or umbilical cord tissue substantially free of blood, wherein the cell is capable of self-renewal and expansion in culture and has the potential to differentiate into a cell of a neural phenotype; wherein the cell requires L-valine for growth and is capable of growth in at least about 5% oxygen; wherein the cell further comprises at least one of the following characteristics:
a) potential for at least about 40 doublings in culture;
b) attachment and expansion on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin, or fibronectin;
c) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; d) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
e) lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry; f) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of a gene encoding: interleukin 8; reticulon 1; chemokine (C—X—C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C—X—C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C—X—C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547k1113.
g) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of a gene encoding: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMAI001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); similar to neuralin 1; B cell translocation gene 1; hypothetical protein FLJ23191; and DKFZp586L151;
h) secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and
i) lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
2. The postpartum-derived cell of claim 1 isolated in the presence of one or more enzyme activities comprising metalloprotease activity, mucolytic activity and neutral protease activity.
3. The postpartum-derived cell of claim 6 comprising a normal karyotype.
4. The postpartum-derived cell of claim 9 wherein the cell comprises each of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, and HLA-A,B,C and does not comprise any of CD31, CD34, CD45, CD117, CD141, or HLA-DR,DP,DQ, as detected by flow cytometry.
5. A cell population comprising the postpartum-derived cell of claim 1.
6. The cell population of claim 5, which is a substantially homogeneous population of the postpartum-derived cells.
7. The cell population of claim 6 comprising a clonal cell line of postpartum-derived cells.
8. The cell population of claim 5, which is a heterogeneous population comprising the postpartum-derived cells and at least one other cell type.
9. The cell population of claim 8, wherein the at least one other cell type is an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell or other multipotent or pluripotent stem cell.
10. A cell lysate prepared from the cell population of claim 5.
11. A soluble cell fraction prepared from the cell lysate of claim 10.
12. An extracellular matrix prepared from the cell population of claim 5.
13. The cell population of claim 5, cultured in contact with one or more factors that stimulate stem cell differentiation toward a neural or epithelial lineage.
14-29. (canceled)
30. A pharmaceutical composition for treating a patient having an ocular degenerative condition, comprising a pharmaceutically acceptable carrier and multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord in an amount effective to treat the ocular degenerative condition.
31. The pharmaceutical composition of claim 30, wherein the ocular degenerative condition is an acute ocular degenerative condition.
32. The pharmaceutical composition of claim 30, wherein the ocular degenerative condition is a chronic or progressive degenerative condition.
33. The pharmaceutical composition of claim 30, wherein the multipotent or pluripotent cells are isolated postpartum-derived cells derived from human placental or umbilical cord tissue substantially free of blood, wherein the cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of at least a neural phenotype; wherein the cells require L-valine for growth and can grow in at least about 5% oxygen; wherein the cells further comprise at least one of the following characteristics:
a) potential for at least about 40 doublings in culture;
b) attachment and expansion on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyornithine, vitronectin, or fibronectin;
c) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin; d) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
e) lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry;
f) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of a gene encoding: interleukin 8; reticulon 1; chemokine (C—X—C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C—X—C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C—X—C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547k1113.
g) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of a gene encoding: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); similar to neuralin 1; B cell translocation gene 1; hypothetical protein FLJ23191; and DKFZp586L151;
h) secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and
i) lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
34. The pharmaceutical composition of claim 30, wherein the cells are induced in vitro to differentiate into a neural or epithelial lineage cells prior to formulation of the composition.
35. The pharmaceutical composition of claim 30, comprising at least one other cell type.
36. The pharmaceutical composition of claim 35, wherein the other cell type is an astrocyte, oligodendrocyte, neuron, neural progenitor, neural stem cell, retinal epithelial stem cell, corneal epithelial stem cell, or other multipotent or pluripotent stem cell.
37. The pharmaceutical composition of claim 30, comprising at least one other agent.
38. The pharmaceutical composition of claim 37, wherein the other agent is a drug for treating the ocular degenerative disorder.
39. The pharmaceutical composition of claim 30, formulated for administration to the surface of an eye.
40. The pharmaceutical composition of claim 30, formulated for administration to the interior of an eye.
41. The pharmaceutical composition of claim 30, formulated as a matrix or scaffold containing the cells.
42. A kit for treating a patient having an ocular degenerative condition, the kit comprising a pharmaceutically acceptable carrier, a population of multipotent or pluripotent cells isolated from postpartum placenta or umbilicus, and instructions for using the kit in a method of treating the patient.
43. The kit of claim 42, which further comprises at least one reagent and instructions for culturing the cells.
44. The kit of claim 42, which further comprises a population of at least one other cell type.
45. The kit of claim 42, which further comprises at least one other agent for treating an ocular degenerative condition.
46. (canceled)
47. A pharmaceutical composition for treating a patient having an ocular degenerative condition, which comprises a pharmaceutically acceptable carrier and a preparation made from multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord, in an amount effective to treat the ocular degenerative condition, wherein the preparation comprises a cell lysate of the cells, or a conditioned medium in which the cells were grown or an extracellular matrix of the cells.
48. A kit for treating a patient having an ocular degenerative condition, which comprises a pharmaceutically acceptable carrier, a preparation made from multipotent or pluripotent cells isolated from a postpartum placenta or umbilical cord, wherein the preparation comprises a cell lysate of the cells, or a conditioned medium in which the cells were grown or an extracellular matrix of the cells, and instructions for using the kit components for treatment of the ocular degenerative condition.
49. (canceled)
50. A kit for increasing the survival, growth or activity of cells for transplantation to treat an ocular degenerative disorder, comprising cultured cells derived from postpartum placental or umbilical tissue and instructions for co-culturing the cells for transplantation with the postpartum cells under conditions effective to increase the survival, growth or activity of the cells for transplantation.
51. The pharmaceutical composition of claim 47, wherein the ocular degenerative condition is an acute ocular degenerative condition.
52. The pharmaceutical composition of claim 47, wherein the ocular degenerative condition is a chronic or progressive degenerative condition.
53. The pharmaceutical composition of claim 47, wherein the multipotent or pluripotent cells are isolated postpartum-derived cells derived from human placental or umbilical cord tissue substantially free of blood, wherein the cells are capable of self-renewal and expansion in culture and have the potential to differentiate into cells of at least a neural phenotype; wherein the cells require L-valine for growth and can grow in at least about 5% oxygen; wherein the cells further comprise at least one of the following characteristics:
a) potential for at least about 40 doublings in culture;
b) attachment and expansion on a coated or uncoated tissue culture vessel, wherein the coated tissue culture vessel comprises a coating of gelatin, laminin, collagen, polyomithine, vitronectin, or fibronectin;
c) production of at least one of tissue factor, vimentin, and alpha-smooth muscle actin;
d) production of at least one of CD10, CD13, CD44, CD73, CD90, PDGFr-alpha, PD-L2 and HLA-A,B,C;
e) lack of production of at least one of CD31, CD34, CD45, CD80, CD86, CD117, CD141, CD178, B7-H2, HLA-G, and HLA-DR,DP,DQ, as detected by flow cytometry;
f) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is increased for at least one of a gene encoding: interleukin 8; reticulon 1; chemokine (C—X—C motif) ligand 1 (melonoma growth stimulating activity, alpha); chemokine (C—X—C motif) ligand 6 (granulocyte chemotactic protein 2); chemokine (C—X—C motif) ligand 3; tumor necrosis factor, alpha-induced protein 3; C-type lectin superfamily member 2; Wilms tumor 1; aldehyde dehydrogenase 1 family member A2; renin; oxidized low density lipoprotein receptor 1; Homo sapiens clone IMAGE:4179671; protein kinase C zeta; hypothetical protein DKFZp564F013; downregulated in ovarian cancer 1; and Homo sapiens gene from clone DKFZp547k1113.
g) expression of a gene, which relative to a human cell that is a fibroblast, a mesenchymal stem cell, or an ileac crest bone marrow cell, is reduced for at least one of a gene encoding: short stature homeobox 2; heat shock 27 kDa protein 2; chemokine (C—X—C motif) ligand 12 (stromal cell-derived factor 1); elastin (supravalvular aortic stenosis, Williams-Beuren syndrome); Homo sapiens mRNA; cDNA DKFZp586M2022 (from clone DKFZp586M2022); mesenchyme homeo box 2 (growth arrest-specific homeo box); sine oculis homeobox homolog 1 (Drosophila); crystallin, alpha B; disheveled associated activator of morphogenesis 2; DKFZP586B2420 protein; similar to neuralin 1; tetranectin (plasminogen binding protein); src homology three (SH3) and cysteine rich domain; cholesterol 25-hydroxylase; runt-related transcription factor 3; interleukin 11 receptor, alpha; procollagen C-endopeptidase enhancer; frizzled homolog 7 (Drosophila); hypothetical gene BC008967; collagen, type VIII, alpha 1; tenascin C (hexabrachion); iroquois homeobox protein 5; hephaestin; integrin, beta 8; synaptic vesicle glycoprotein 2; neuroblastoma, suppression of tumorigenicity 1; insulin-like growth factor binding protein 2, 36 kDa; Homo sapiens cDNA FLJ12280 fis, clone MAMMA1001744; cytokine receptor-like factor 1; potassium intermediate/small conductance calcium-activated channel, subfamily N, member 4; integrin, beta 7; transcriptional co-activator with PDZ-binding motif (TAZ); sine oculis homeobox homolog 2 (Drosophila); KIAA1034 protein; vesicle-associated membrane protein 5 (myobrevin); EGF-containing fibulin-like extracellular matrix protein 1; early growth response 3; distal-less homeo box 5; hypothetical protein FLJ20373; aldo-keto reductase family 1, member C3 (3-alpha hydroxysteroid dehydrogenase, type II); biglycan; transcriptional co-activator with PDZ-binding motif (TAZ); fibronectin 1; proenkephalin; integrin, beta-like 1 (with EGF-like repeat domains); Homo sapiens mRNA full length insert cDNA clone EUROIMAGE 1968422; EphA3; KIAA0367 protein; natriuretic peptide receptor C/guanylate cyclase C (atrionatriuretic peptide receptor C); hypothetical protein FLJ14054; Homo sapiens mRNA; cDNA DKFZp564B222 (from clone DKFZp564B222); BCL2/adenovirus E1B 19 kDa interacting protein 3-like; AE binding protein 1; cytochrome c oxidase subunit VIIa polypeptide 1 (muscle); similar to neuralin 1; B cell translocation gene 1; hypothetical protein FLJ23191; and DKFZp586L151;
h) secretion of at least one of MCP-1, IL-6, IL-8, GCP-2, HGF, KGF, FGF, HB-EGF, BDNF, TPO, MIP1a, RANTES, and TIMP1; and
i) lack of secretion of at least one of TGF-beta2, ANG2, PDGFbb, MIP1b, 1309, MDC, and VEGF, as detected by ELISA.
54. The pharmaceutical composition of claim 47, wherein the cells are induced in vitro to differentiate into a neural or epithelial lineage cells prior to making the preparation.
55. The pharmaceutical composition of claim 47, comprising at least one other agent.
56. The pharmaceutical composition of claim 55, wherein the other agent is a drug for treating the ocular degenerative disorder.
57. The pharmaceutical composition of claim 47, formulated for administration to the surface of an eye.
58. The pharmaceutical composition of claim 47, formulated for administration to the interior of an eye.
59. The pharmaceutical composition of claim 47, formulated as a matrix or scaffold containing the preparation.
60. The kit of claim 48, which further comprises at least one other agent for treating an ocular degenerative condition.
61. The kit of claim 50, which further comprises at least one reagent and instructions for culturing the cells.
62. The kit of claim 50, which further comprises a population of at least one other cell type.
63. The kit of claim 50, which further comprises at least one other agent for treating an ocular degenerative condition.
US11/317,574 2003-06-27 2005-12-23 Repair and regeneration of ocular tissue using postpartum-derived cells Abandoned US20060234376A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/317,574 US20060234376A1 (en) 2003-06-27 2005-12-23 Repair and regeneration of ocular tissue using postpartum-derived cells

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US48326403P 2003-06-27 2003-06-27
US10/877,541 US7413734B2 (en) 2003-06-27 2004-06-25 Treatment of retinitis pigmentosa with human umbilical cord cells
US11/317,574 US20060234376A1 (en) 2003-06-27 2005-12-23 Repair and regeneration of ocular tissue using postpartum-derived cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/877,541 Division US7413734B2 (en) 2003-06-27 2004-06-25 Treatment of retinitis pigmentosa with human umbilical cord cells

Publications (1)

Publication Number Publication Date
US20060234376A1 true US20060234376A1 (en) 2006-10-19

Family

ID=38456561

Family Applications (28)

Application Number Title Priority Date Filing Date
US10/877,541 Active US7413734B2 (en) 2003-06-27 2004-06-25 Treatment of retinitis pigmentosa with human umbilical cord cells
US10/877,446 Active 2027-11-10 US9579351B2 (en) 2003-06-27 2004-06-25 Postpartum cells derived from placental tissue, and methods of making and using the same
US10/877,445 Active 2026-05-22 US8703121B2 (en) 2003-06-27 2004-06-25 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US10/876,998 Active 2024-11-21 US11191789B2 (en) 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells
US10/877,009 Active 2025-06-19 US7560276B2 (en) 2003-01-07 2004-06-25 Soft tissue repair and regeneration using postpartum-derived cells
US10/877,012 Active 2024-11-26 US7510873B2 (en) 2003-06-27 2004-06-25 Postpartum cells isolated from umbilical cord tissue, and methods of making and using the same
US10/877,269 Active 2025-09-28 US7524489B2 (en) 2003-06-27 2004-06-25 Regeneration and repair of neural tissue using postpartum-derived cells
US11/316,104 Active 2027-08-17 US9504719B2 (en) 2003-01-07 2005-12-22 Soft tissue repair and regeneration using postpartum-derived cells and cell products
US11/315,969 Active 2025-07-01 US11179422B2 (en) 2003-06-27 2005-12-22 Method of differentiating umbilical cord tissue into a chondrogenic phenotype
US11/317,574 Abandoned US20060234376A1 (en) 2003-06-27 2005-12-23 Repair and regeneration of ocular tissue using postpartum-derived cells
US11/321,863 Abandoned US20060153817A1 (en) 2003-06-27 2005-12-29 Cartilage and bone repair and regeneration using postpartum-derived cells
US11/321,864 Abandoned US20060153818A1 (en) 2003-06-27 2005-12-29 Cartilage and bone repair and regeneration using postpartum-derived cells
US11/322,003 Abandoned US20060154367A1 (en) 2003-06-27 2005-12-29 Cartilage and bone repair and regeneration using postpartum-derived cells
US11/322,372 Abandoned US20060188983A1 (en) 2003-06-27 2005-12-30 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US11/481,481 Abandoned US20070014771A1 (en) 2003-06-27 2006-07-06 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US11/481,456 Active 2025-12-28 US8318483B2 (en) 2003-06-27 2006-07-06 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US11/481,480 Abandoned US20070009494A1 (en) 2003-06-27 2006-07-06 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US12/389,305 Active 2025-11-18 US9498501B2 (en) 2003-06-27 2009-02-19 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US12/429,849 Active 2025-08-13 US8277796B2 (en) 2003-06-27 2009-04-24 Regeneration and repair of neural tissue using postpartum-derived cells
US13/605,716 Active US8658152B2 (en) 2003-06-27 2012-09-06 Regeneration and repair of neural tissue using postpartum-derived cells
US14/018,842 Active 2026-02-17 US9717763B2 (en) 2003-06-27 2013-09-05 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US15/362,453 Active 2024-08-28 US10039793B2 (en) 2003-06-27 2016-11-28 Soft tissue repair and regeneration using postpartum-derived cells and cell products
US15/443,602 Active US10220059B2 (en) 2003-06-27 2017-02-27 Postpartum cells derived from placental tissue, and methods of making and using the same
US15/443,733 Expired - Fee Related US10383898B2 (en) 2003-06-27 2017-02-27 Postpartum cells derived from placental tissue, and methods of making and using the same
US15/443,718 Active US10195233B2 (en) 2003-06-27 2017-02-27 Postpartum cells derived from placental tissue, and methods of making and using the same
US15/661,878 Expired - Fee Related US10500234B2 (en) 2003-06-27 2017-07-27 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US16/056,146 Active 2024-09-27 US10758576B2 (en) 2003-06-27 2018-08-06 Soft tissue repair and regeneration using postpartum-derived cells and cell products
US16/291,784 Active 2024-06-27 US11000554B2 (en) 2003-06-27 2019-03-04 Postpartum cells derived from placental tissue, and methods of making and using the same

Family Applications Before (9)

Application Number Title Priority Date Filing Date
US10/877,541 Active US7413734B2 (en) 2003-06-27 2004-06-25 Treatment of retinitis pigmentosa with human umbilical cord cells
US10/877,446 Active 2027-11-10 US9579351B2 (en) 2003-06-27 2004-06-25 Postpartum cells derived from placental tissue, and methods of making and using the same
US10/877,445 Active 2026-05-22 US8703121B2 (en) 2003-06-27 2004-06-25 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US10/876,998 Active 2024-11-21 US11191789B2 (en) 2003-06-27 2004-06-25 Cartilage and bone repair and regeneration using postpartum-derived cells
US10/877,009 Active 2025-06-19 US7560276B2 (en) 2003-01-07 2004-06-25 Soft tissue repair and regeneration using postpartum-derived cells
US10/877,012 Active 2024-11-26 US7510873B2 (en) 2003-06-27 2004-06-25 Postpartum cells isolated from umbilical cord tissue, and methods of making and using the same
US10/877,269 Active 2025-09-28 US7524489B2 (en) 2003-06-27 2004-06-25 Regeneration and repair of neural tissue using postpartum-derived cells
US11/316,104 Active 2027-08-17 US9504719B2 (en) 2003-01-07 2005-12-22 Soft tissue repair and regeneration using postpartum-derived cells and cell products
US11/315,969 Active 2025-07-01 US11179422B2 (en) 2003-06-27 2005-12-22 Method of differentiating umbilical cord tissue into a chondrogenic phenotype

Family Applications After (18)

Application Number Title Priority Date Filing Date
US11/321,863 Abandoned US20060153817A1 (en) 2003-06-27 2005-12-29 Cartilage and bone repair and regeneration using postpartum-derived cells
US11/321,864 Abandoned US20060153818A1 (en) 2003-06-27 2005-12-29 Cartilage and bone repair and regeneration using postpartum-derived cells
US11/322,003 Abandoned US20060154367A1 (en) 2003-06-27 2005-12-29 Cartilage and bone repair and regeneration using postpartum-derived cells
US11/322,372 Abandoned US20060188983A1 (en) 2003-06-27 2005-12-30 Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US11/481,481 Abandoned US20070014771A1 (en) 2003-06-27 2006-07-06 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US11/481,456 Active 2025-12-28 US8318483B2 (en) 2003-06-27 2006-07-06 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US11/481,480 Abandoned US20070009494A1 (en) 2003-06-27 2006-07-06 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US12/389,305 Active 2025-11-18 US9498501B2 (en) 2003-06-27 2009-02-19 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US12/429,849 Active 2025-08-13 US8277796B2 (en) 2003-06-27 2009-04-24 Regeneration and repair of neural tissue using postpartum-derived cells
US13/605,716 Active US8658152B2 (en) 2003-06-27 2012-09-06 Regeneration and repair of neural tissue using postpartum-derived cells
US14/018,842 Active 2026-02-17 US9717763B2 (en) 2003-06-27 2013-09-05 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US15/362,453 Active 2024-08-28 US10039793B2 (en) 2003-06-27 2016-11-28 Soft tissue repair and regeneration using postpartum-derived cells and cell products
US15/443,602 Active US10220059B2 (en) 2003-06-27 2017-02-27 Postpartum cells derived from placental tissue, and methods of making and using the same
US15/443,733 Expired - Fee Related US10383898B2 (en) 2003-06-27 2017-02-27 Postpartum cells derived from placental tissue, and methods of making and using the same
US15/443,718 Active US10195233B2 (en) 2003-06-27 2017-02-27 Postpartum cells derived from placental tissue, and methods of making and using the same
US15/661,878 Expired - Fee Related US10500234B2 (en) 2003-06-27 2017-07-27 Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US16/056,146 Active 2024-09-27 US10758576B2 (en) 2003-06-27 2018-08-06 Soft tissue repair and regeneration using postpartum-derived cells and cell products
US16/291,784 Active 2024-06-27 US11000554B2 (en) 2003-06-27 2019-03-04 Postpartum cells derived from placental tissue, and methods of making and using the same

Country Status (8)

Country Link
US (28) US7413734B2 (en)
EP (14) EP1641913B1 (en)
JP (7) JP5148873B2 (en)
AU (7) AU2004252570C1 (en)
CA (7) CA2530255C (en)
ES (14) ES2600555T3 (en)
PL (14) PL2338980T3 (en)
WO (7) WO2005001076A2 (en)

Cited By (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020160510A1 (en) * 2001-02-14 2002-10-31 Hariri Robert J. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US20050019908A1 (en) * 2000-12-06 2005-01-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20060069008A1 (en) * 2004-09-28 2006-03-30 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US20060222634A1 (en) * 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US20060223177A1 (en) * 2003-06-27 2006-10-05 Ethicon Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20060233765A1 (en) * 2003-06-27 2006-10-19 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
WO2008083223A1 (en) * 2006-12-28 2008-07-10 Isolagen Technologies, Inc. Methods for culturing dermal cells for treatment of skin injuries such as burns
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers
US20080286324A1 (en) * 2007-05-14 2008-11-20 Cardiac Pacemakers, Inc. Media and devices for cold storage of therapeutic cells
US20080286267A1 (en) * 2007-04-13 2008-11-20 Sing George L Methods for treating nervous system inquiry and disease
US20090010899A1 (en) * 2005-03-31 2009-01-08 Palladino Linda O Methods related to surgery
WO2009052459A1 (en) * 2007-10-19 2009-04-23 University Of Medicine And Dentistry Of New Jersey A method of using an extracellular matrix to enhance cell transplant survival and differentiation
US20090220995A1 (en) * 2008-02-28 2009-09-03 Sachs David H Multiple administrations of umbilicus derived cells
US7682803B2 (en) 2005-10-13 2010-03-23 Anthrogenesis Corporation Immunomodulation using placental stem cells
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US20100196910A1 (en) * 2001-07-20 2010-08-05 Technion Research & Development Foundation Ltd. Methods of generating human cardiac cells and tissues and uses thereof
US7875273B2 (en) 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US7976836B2 (en) 2000-12-06 2011-07-12 Anthrogenesis Corporation Treatment of stroke using placental stem cells
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US8034329B2 (en) 2007-10-05 2011-10-11 Advanced Technologies And Regenerative Medicine, Llc Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US8057789B2 (en) 2002-02-13 2011-11-15 Anthrogenesis Corporation Placental stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US8221741B2 (en) 2007-01-17 2012-07-17 Marshall Vivienne S Methods for modulating inflammatory and/or immune responses
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US8273526B2 (en) 2007-06-18 2012-09-25 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US8562972B2 (en) 2006-10-23 2013-10-22 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
US8722034B2 (en) 2009-03-26 2014-05-13 Depuy Synthes Products Llc hUTC as therapy for Alzheimer's disease
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8741638B2 (en) 2005-12-19 2014-06-03 DePuy Synthes Products, LLC In vitro expansion of postpartum-derived cells in roller bottles
US8771677B2 (en) 2008-12-29 2014-07-08 Vladimir B Serikov Colony-forming unit cell of human chorion and method to obtain and use thereof
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US8808687B2 (en) 2010-07-12 2014-08-19 Mark Humayun Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
WO2016068616A1 (en) * 2014-10-29 2016-05-06 차의과학대학교 산학협력단 Placenta-derived cells excreting c3 or c1r complement and composition containing same
US9358320B2 (en) 2008-04-25 2016-06-07 Allosource Multi-layer tissue patches
US9446077B2 (en) 2013-03-13 2016-09-20 Allosource Fascia fibrous compositions and methods for their use and manufacture
US9480549B2 (en) 2008-04-25 2016-11-01 Allosource Multi-layer tissue patches
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US9598672B2 (en) 2007-10-19 2017-03-21 Rutgers, The State University Of New Jersey Production of extracellular matrix, conditioned media and uses thereof
US9611513B2 (en) 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
US9744266B2 (en) 2011-12-19 2017-08-29 Allosource Flowable matrix compositions and methods
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US9795707B2 (en) 2013-12-06 2017-10-24 Allosource Methods of drying sheets of donor-provided human birth tissue
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US10016600B2 (en) 2013-05-30 2018-07-10 Neurostim Solutions, Llc Topical neurological stimulation
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
US10413571B2 (en) 2006-10-12 2019-09-17 DePuy Synthes Products, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US10478206B2 (en) 2011-04-29 2019-11-19 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US10557116B2 (en) 2008-12-19 2020-02-11 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
US10568990B2 (en) 2013-03-15 2020-02-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
US10772986B2 (en) 2017-01-26 2020-09-15 Allosource Fascia fibrous compositions and methods for their use and manufacture
US10953225B2 (en) 2017-11-07 2021-03-23 Neurostim Oab, Inc. Non-invasive nerve activator with adaptive circuit
US11077301B2 (en) 2015-02-21 2021-08-03 NeurostimOAB, Inc. Topical nerve stimulator and sensor for bladder control
US11229789B2 (en) 2013-05-30 2022-01-25 Neurostim Oab, Inc. Neuro activator with controller
US11458311B2 (en) 2019-06-26 2022-10-04 Neurostim Technologies Llc Non-invasive nerve activator patch with adaptive circuit
US11730958B2 (en) 2019-12-16 2023-08-22 Neurostim Solutions, Llc Non-invasive nerve activator with boosted charge delivery

Families Citing this family (460)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080077251A1 (en) * 1999-06-07 2008-03-27 Chen Silvia S Cleaning and devitalization of cartilage
US8563232B2 (en) 2000-09-12 2013-10-22 Lifenet Health Process for devitalizing soft-tissue engineered medical implants, and devitalized soft-tissue medical implants produced
US20020095157A1 (en) * 1999-07-23 2002-07-18 Bowman Steven M. Graft fixation device combination
US6179840B1 (en) 1999-07-23 2001-01-30 Ethicon, Inc. Graft fixation device and method
US8147824B2 (en) 1999-08-05 2012-04-03 Athersys, Inc. Immunomodulatory properties of multipotent adult progenitor cells and uses thereof
US20080152629A1 (en) * 2000-12-06 2008-06-26 James Edinger Placental stem cell populations
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
US7677565B2 (en) 2001-09-28 2010-03-16 Shuffle Master, Inc Card shuffler with card rank and value reading capability
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20080299090A1 (en) * 2002-02-25 2008-12-04 Kansas State University Research Foundation Use Of Umbilical Cord Matrix Cells
US20080003258A1 (en) * 2002-10-16 2008-01-03 Marcum Frank D Composition and Method for Treating Rheumatoid Arthritis
US7824701B2 (en) 2002-10-18 2010-11-02 Ethicon, Inc. Biocompatible scaffold for ligament or tendon repair
US20040078090A1 (en) 2002-10-18 2004-04-22 Francois Binette Biocompatible scaffolds with tissue fragments
WO2004047770A2 (en) 2002-11-26 2004-06-10 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
US7807458B2 (en) 2003-01-30 2010-10-05 The United States Of America As Represented By The Secretary Of The Department Of Veterans Affairs Multilineage-inducible cells and uses thereof
CA2515469C (en) 2003-02-11 2013-12-17 John E. Davies Progenitor cells from wharton's jelly of human umbilical cord
KR20050105467A (en) * 2003-02-13 2005-11-04 안트로제네시스 코포레이션 Use of umbilical cord blood to treat individuals having a disease, disorder or condition
US8197837B2 (en) 2003-03-07 2012-06-12 Depuy Mitek, Inc. Method of preparation of bioabsorbable porous reinforced tissue implants and implants thereof
CN100377165C (en) * 2003-04-24 2008-03-26 皇家飞利浦电子股份有限公司 Non-invasive left ventricular volume determination
US8226715B2 (en) 2003-06-30 2012-07-24 Depuy Mitek, Inc. Scaffold for connective tissue repair
US10583220B2 (en) 2003-08-11 2020-03-10 DePuy Synthes Products, Inc. Method and apparatus for resurfacing an articular surface
AU2004262451B2 (en) * 2003-08-12 2009-11-26 Tigenix N.V. Use of CXCL6 chemokine in the prevention or repair of cartilage defects
US8043614B2 (en) * 2004-03-09 2011-10-25 Ahlfors Jan-Eric W Autogenic living scaffolds and living tissue matrices: methods and uses thereof
GB0321337D0 (en) * 2003-09-11 2003-10-15 Massone Mobile Advertising Sys Method and system for distributing advertisements
US7682828B2 (en) 2003-11-26 2010-03-23 Whitehead Institute For Biomedical Research Methods for reprogramming somatic cells
US7316822B2 (en) 2003-11-26 2008-01-08 Ethicon, Inc. Conformable tissue repair implant capable of injection delivery
WO2005055929A2 (en) * 2003-12-02 2005-06-23 Celgene Corporation Methods and compositions for the treatment and management of hemoglobinopathy and anemia
US7901461B2 (en) * 2003-12-05 2011-03-08 Ethicon, Inc. Viable tissue repair implants and methods of use
JP2005168360A (en) * 2003-12-09 2005-06-30 Olympus Corp Method for examining biological tissue-supplying material, device, cell culture container and method for examining culturing state
US20050176139A1 (en) * 2004-01-12 2005-08-11 Yao-Chang Chen Placental stem cell and methods thereof
WO2005071066A1 (en) * 2004-01-23 2005-08-04 Board Of Regents, The University Of Texas System Methods and compositions for preparing pancreatic insulin secreting cells
US11395865B2 (en) 2004-02-09 2022-07-26 DePuy Synthes Products, Inc. Scaffolds with viable tissue
EP2824174B1 (en) 2004-03-22 2018-11-28 Mesoblast International Sàrl Mesenchymal stem cells and uses therefor
US8137686B2 (en) 2004-04-20 2012-03-20 Depuy Mitek, Inc. Nonwoven tissue scaffold
US8221780B2 (en) * 2004-04-20 2012-07-17 Depuy Mitek, Inc. Nonwoven tissue scaffold
CN101080486B (en) * 2004-04-23 2012-05-16 佰欧益股份有限公司 Multi-lineage progenitor cells
US7622108B2 (en) 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
US8765119B2 (en) * 2004-05-06 2014-07-01 University Of South Florida Treating amyotrophic lateral sclerosis (ALS)with isolated aldehyde dehydrogenase-positive umbilical cord blood cells
US8802651B2 (en) * 2004-06-30 2014-08-12 Abbott Medical Optics Inc. Hyaluronic acid in the enhancement of lens regeneration
US7794697B2 (en) 2004-06-30 2010-09-14 Abbott Medical Optics Inc. Enhancement of lens regeneration using materials comprising polysiloxane polymers
US20060083732A1 (en) * 2004-06-30 2006-04-20 Arlene Gwon Hyaluronic acid in the enhancement of lens regeneration
MX2007001937A (en) 2004-08-16 2007-08-07 Cellres Corp Pte Ltd Isolation of stem/progenitor cells from amniotic membrane of umbilical cord.
US20060045872A1 (en) 2004-08-25 2006-03-02 Universidad Autonoma De Madrid Ciudad Universitaria de Cantoblanco Use of adipose tissue-derived stromal stem cells in treating fistula
US8039258B2 (en) * 2004-09-28 2011-10-18 Ethicon, Inc. Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20080038316A1 (en) * 2004-10-01 2008-02-14 Wong Vernon G Conveniently implantable sustained release drug compositions
US7473678B2 (en) * 2004-10-14 2009-01-06 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and methods of use thereof
US7842304B2 (en) * 2004-10-29 2010-11-30 Nexeon Medsystems, Inc. Methods and apparatus for treating an injured nerve pathway
US11660317B2 (en) 2004-11-08 2023-05-30 The Johns Hopkins University Compositions comprising cardiosphere-derived cells for use in cell therapy
US8017395B2 (en) 2004-12-17 2011-09-13 Lifescan, Inc. Seeding cells on porous supports
WO2006101548A2 (en) * 2004-12-21 2006-09-28 Ethicon, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
EP1838842A2 (en) * 2004-12-23 2007-10-03 Ethicon, Incorporated Treatment of osteochondral diseases using postpartum-derived cells and products thereof
EP1831355A2 (en) * 2004-12-23 2007-09-12 Ethicon, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
WO2006088867A2 (en) * 2005-02-15 2006-08-24 Medistem Laboratories, Incorporated Method for expansion of stem cells
JP4783909B2 (en) * 2005-03-04 2011-09-28 国立大学法人京都大学 Pluripotent stem cells derived from heart tissue
JP5017253B2 (en) * 2005-03-31 2012-09-05 ステムニオン,インコーポレイテッド Amnion-derived cell composition, preparation method and use thereof
US8999706B2 (en) * 2005-04-12 2015-04-07 The Trustees Of The University Of Pennsylvania Methods for preparation of human hair-follicle derived multipotent adult stem cells
WO2006113731A2 (en) * 2005-04-20 2006-10-26 University Of Florida Research Foundation, Inc. Bone marrow-derived neurogenic cells and uses thereof
US20100087546A1 (en) * 2005-04-20 2010-04-08 Biogenic Innovations, Llc Use of dimethyl sulfone (msm) to reduce homocysteine levels
KR20080007380A (en) * 2005-04-25 2008-01-18 메사추세츠 인스티튜트 오브 테크놀로지 Compositions and methods for promoting hemostasis and other physiological activities
AU2006202209B2 (en) * 2005-05-27 2011-04-14 Lifescan, Inc. Amniotic fluid derived cells
AU2006255183B2 (en) * 2005-06-08 2012-02-02 Centocor, Inc. A cellular therapy for ocular degeneration
GB0511723D0 (en) * 2005-06-09 2005-07-13 Smith & Nephew Placental stem cells
WO2006135899A2 (en) * 2005-06-13 2006-12-21 The Johns Hopkins University Survival, differentiation and structural intergration of human neural stem cells grafted into the adult spinal cord
US7838503B2 (en) * 2005-06-15 2010-11-23 Children's Medical Center Corporation Methods for extending the replicative lifespan of cells
US20140336514A1 (en) * 2005-08-05 2014-11-13 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
US20150119794A1 (en) * 2005-08-05 2015-04-30 Gholam A. Peyman Methods to regulate polarization and enhance function of cells
AU2006291134C1 (en) 2005-09-12 2013-08-15 Abela Pharmaceuticals, Inc. Systems for removing dimethyl sulfoxide (DMSO) or related compounds, or odors associated with same
US8435224B2 (en) 2005-09-12 2013-05-07 Abela Pharmaceuticals, Inc. Materials for facilitating administration of dimethyl sulfoxide (DMSO) and related compounds
US9427419B2 (en) 2005-09-12 2016-08-30 Abela Pharmaceuticals, Inc. Compositions comprising dimethyl sulfoxide (DMSO)
US8480797B2 (en) 2005-09-12 2013-07-09 Abela Pharmaceuticals, Inc. Activated carbon systems for facilitating use of dimethyl sulfoxide (DMSO) by removal of same, related compounds, or associated odors
JP2007106760A (en) * 2005-09-16 2007-04-26 Kenji Yoshida Hematopoietic stem cell proliferator
US8182840B2 (en) 2005-09-27 2012-05-22 Tissue Tech, Inc. Amniotic membrane preparations and purified compositions and therapy for scar reversal and inhibition
NZ567334A (en) * 2005-10-13 2012-08-31 Anthrogenesis Corp Production of oligodendrocytes from placenta-derived stem cells
JP4921767B2 (en) * 2005-10-14 2012-04-25 株式会社カネカ Cell differentiation induction method
US11000546B2 (en) 2005-11-09 2021-05-11 Athersys, Inc. Immunomodulatory properties of MAPCs and uses thereof
US10117900B2 (en) * 2005-11-09 2018-11-06 Athersys, Inc. MAPC treatment of brain injuries and diseases
US20070178067A1 (en) * 2005-11-09 2007-08-02 John Maier System and method for cytological analysis by raman spectroscopic imaging
US20090170059A1 (en) * 2005-11-14 2009-07-02 Hans Klingemann Methods for Preparing Cord Matrix Stem Cells (CMSC) for Long Term Storage and for Preparing a Segment of umbilical cord for cryopreservation
NZ571113A (en) * 2005-11-17 2012-02-24 Biomimetic Therapeutics Inc Maxillofacial bone augmentation using rhpdgf-bb and a biocompatible matrix
CN103173401A (en) 2005-12-22 2013-06-26 简·恩尼斯 Viable cells from frozen umbilical cord tissue
WO2007076522A2 (en) * 2005-12-28 2007-07-05 Ethicon, Incorporated Treatment of peripheral vascular disease using postpartum-derived cells
CN105106239A (en) * 2005-12-28 2015-12-02 伊西康公司 Treatment of peripheral vascular disease using postpartum-derived cells
US9598669B2 (en) * 2005-12-29 2017-03-21 Anthrogenesis Corporation Composition for collecting placental stem cells and methods of using the composition
EP1981970A4 (en) 2006-01-11 2009-10-21 Technion Res & Dev Foundation Human embryonic stem cell-derived connective tissue progenitors for tissue engineering
US20080286249A1 (en) * 2006-01-12 2008-11-20 Varney Timothy R Use of mesenchymal stem cells for treating genetic diseases and disorders
US20070253931A1 (en) * 2006-01-12 2007-11-01 Osiris Therapeutics, Inc. Use of mesenchymal stem cells for treating genetic diseases and disorders
US9944900B2 (en) * 2006-01-18 2018-04-17 Hemacell Perfusion Pulsatile perfusion extraction method for non-embryonic pluripotent stem cells
GB0600972D0 (en) * 2006-01-18 2006-03-01 Univ Leeds Enrichment of cells
US8871198B2 (en) * 2006-03-29 2014-10-28 Stemnion, Inc. Methods related to wound healing
US7875451B2 (en) * 2006-01-19 2011-01-25 The University Of Washington Formulation to improve survival of transplanted cells
NZ744465A (en) 2006-01-23 2023-04-28 Abt Holding Co Mapc therapeutics without adjunctive immunosuppressive treatment
CA2640185A1 (en) * 2006-01-24 2007-08-02 Christopher J. Centeno Mesenchymal stem cell isolation and transplantation method and system to be used in a clinical setting
FR2896511B1 (en) * 2006-01-26 2012-10-26 Centre Nat Rech Scient PROCESS FOR CULTIVATION OF CELLS FROM ADIPOSE TISSUE AND THEIR APPLICATIONS
EP2311505B1 (en) 2006-02-09 2013-11-06 BioMimetic Therapeutics, LLC Compositions and methods for treating bone
EP1996698A1 (en) * 2006-03-01 2008-12-03 Cartela R&D AB Expansion and differentiation of mesenchymal stem cells
WO2007099534A2 (en) * 2006-03-01 2007-09-07 The Regenerative Medicine Institute Compostions and populations of cells obtained from the umbilical cord and methods of producing the same
US20110171182A1 (en) * 2006-03-23 2011-07-14 Pluristem Ltd. Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
PT2366775E (en) * 2006-03-23 2015-07-20 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
US7727763B2 (en) * 2006-04-17 2010-06-01 Bioe, Llc Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
WO2007124023A2 (en) * 2006-04-21 2007-11-01 Wake Forest University Health Sciences Ink-jet printing of tissues
US9456979B2 (en) * 2006-04-27 2016-10-04 Sri International Adminstration of intact mammalian cells to the brain by the intranasal route
US8741643B2 (en) * 2006-04-28 2014-06-03 Lifescan, Inc. Differentiation of pluripotent stem cells to definitive endoderm lineage
PL2029149T3 (en) 2006-05-05 2017-12-29 John E. Davies Immune privileged and modulatory progenitor cells
EP2023717B1 (en) 2006-05-11 2016-04-13 Anthrogenesis Corporation Methods for collecting placenta cord blood stem cells
US20080050814A1 (en) * 2006-06-05 2008-02-28 Cryo-Cell International, Inc. Procurement, isolation and cryopreservation of fetal placental cells
WO2007146106A2 (en) * 2006-06-05 2007-12-21 Cryo- Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
US20080044848A1 (en) * 2006-06-09 2008-02-21 Heidaran Mohammad A Placental niche and use thereof to culture stem cells
WO2007145889A1 (en) * 2006-06-14 2007-12-21 Stemnion, Inc. Methods of treating spinal cord injury and minimizing scarring
US8475788B2 (en) 2006-06-14 2013-07-02 Stemnion, Inc. Methods of treating spinal cord injury and minimizing scarring
US20070292401A1 (en) * 2006-06-20 2007-12-20 Harmon Alexander M Soft tissue repair and regeneration using stem cell products
BRPI0713965A2 (en) * 2006-06-28 2012-11-27 Univ Kansas differentiation of umbilical cord matrix trunk in hepatocyte lineage cells
US9642891B2 (en) * 2006-06-30 2017-05-09 Biomimetic Therapeutics, Llc Compositions and methods for treating rotator cuff injuries
US9161967B2 (en) 2006-06-30 2015-10-20 Biomimetic Therapeutics, Llc Compositions and methods for treating the vertebral column
EP2049043B1 (en) 2006-07-24 2018-10-10 International Stem Cell Corporation Synthetic cornea from retinal stem cells
US8822030B2 (en) 2006-08-11 2014-09-02 Aqua Resources Corporation Nanoplatelet metal hydroxides and methods of preparing same
WO2008021256A2 (en) * 2006-08-11 2008-02-21 Aqua Resources Corporation Nanoplatelet metal hydroxides and methods of preparing same
US7671014B2 (en) * 2006-08-14 2010-03-02 Warsaw Orthopedic, Inc. Flowable carrier matrix and methods for delivering to a patient
US8372437B2 (en) 2006-08-17 2013-02-12 Mimedx Group, Inc. Placental tissue grafts
JP5656183B2 (en) * 2006-08-22 2015-01-21 国立大学法人 東京医科歯科大学 Application of synovial mesenchymal stem cells (MSCs) to cartilage and meniscal regeneration
US20080082170A1 (en) * 2006-09-29 2008-04-03 Peterman Marc M Apparatus and methods for surgical repair
US20080078412A1 (en) * 2006-10-03 2008-04-03 Restore Medical, Inc. Tongue implant
US20080078411A1 (en) * 2006-10-03 2008-04-03 Restore Medical, Inc. Tongue implant for sleep apnea
EP1913869A3 (en) * 2006-10-19 2008-12-10 Esaote S.p.A. Diagnostic imaging method and apparatus for the anatomical region of the pelvic floor
EP3181157B1 (en) * 2006-11-03 2019-08-14 BioMimetic Therapeutics, LLC Compositions and methods for arthrodetic procedures
US20080132803A1 (en) * 2006-11-30 2008-06-05 Hyman Friedlander Method and system for doing business by mining the placental-chord complex
US20080152630A1 (en) * 2006-12-07 2008-06-26 Irene Ginis Method of generation and expansion of tissue-progenitor cells and mature tissue cells from intact bone marrow or intact umbilical cord tissue
US20100143289A1 (en) * 2006-12-19 2010-06-10 Michael Cohen Umbilical cord stem cell secreted product derived topical compositions and methods of use thereof
US7980000B2 (en) 2006-12-29 2011-07-19 Applied Materials, Inc. Vapor dryer having hydrophilic end effector
KR20090109127A (en) 2007-02-12 2009-10-19 안트로제네시스 코포레이션 Hepatocytes and chondrocytes from adherent placental stem cells? and cd34?, cd45? placental stem cell-enriched cell populations
EP2125000A2 (en) * 2007-02-20 2009-12-02 Biomimetic Therapeutics, Inc. Prevention and treatment for osteonecrosis and osteoradionecrosis of the jaw using pdgf and a bone matrix
TW200902718A (en) * 2007-03-01 2009-01-16 Cryo Cell Int Procurement, isolation, and cryopreservation of endometrial/menstrual cells
WO2008109816A1 (en) * 2007-03-08 2008-09-12 Hemacell Perfusion, Inc. Method for isolation of afterbirth derived cells
US20100172830A1 (en) * 2007-03-29 2010-07-08 Cellx Inc. Extraembryonic Tissue cells and method of use thereof
CA3071055A1 (en) * 2007-04-07 2008-10-16 Whitehead Institute For Biomedical Research Reprogramming of somatic cells
WO2008132722A1 (en) 2007-04-26 2008-11-06 Ramot At Tel-Aviv University Ltd. Pluripotent autologous stem cells from oral mucosa and methods of use
US8574567B2 (en) 2007-05-03 2013-11-05 The Brigham And Women's Hospital, Inc. Multipotent stem cells and uses thereof
EP2155860B1 (en) * 2007-05-03 2014-08-27 The Brigham and Women's Hospital, Inc. Multipotent stem cells and uses thereof
US20100111914A1 (en) 2007-05-21 2010-05-06 Yuanyuan Zhang Stem cells from urine and methods for using the same
US10398804B2 (en) 2007-05-21 2019-09-03 Wake Forest University Health Sciences Progenitor cells from urine and methods for using the same
TWM322542U (en) * 2007-05-23 2007-11-21 Universal Scient Ind Co Ltd Testing machine
US20090053182A1 (en) * 2007-05-25 2009-02-26 Medistem Laboratories, Inc. Endometrial stem cells and methods of making and using same
EP2201910B1 (en) * 2007-06-01 2013-04-03 Allergan, Inc. Biological tissue growth support trough induced tensile stress
US9693486B1 (en) * 2007-06-14 2017-06-27 Switch, Ltd. Air handling unit with a canopy thereover for use with a data center and method of using the same
US20090004271A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Morselized foam for wound treatment
US20090004253A1 (en) * 2007-06-29 2009-01-01 Brown Laura J Composite device for the repair or regeneration of tissue
US9080145B2 (en) * 2007-07-01 2015-07-14 Lifescan Corporation Single pluripotent stem cell culture
US9095562B2 (en) * 2007-07-05 2015-08-04 Regenerative Sciences, Inc. Methods and compositions for optimized expansion and implantation of mesenchymal stem cells
DE102007034679A1 (en) * 2007-07-25 2009-01-29 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Material compositions containing adult stem cells derived from exocrine glandular tissue, especially for use in regenerative medicine, e.g. to restore injured or damaged myocardial tissue
US20090024224A1 (en) 2007-07-16 2009-01-22 Chen Silvia S Implantation of cartilage
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
EP2182971B1 (en) 2007-07-27 2013-12-18 Humacyte, Inc. Compositions comprising human collagen and human elastin and uses thereof
EP2185693B1 (en) 2007-07-31 2019-07-03 Lifescan, Inc. Differentiation of human embryonic stem cells
EP2175898A2 (en) * 2007-08-08 2010-04-21 Pervasis Therapeutics, Inc. Materials and methods for treating skeletal system damage and promoting skeletal system repair and regeneration
US20090062907A1 (en) * 2007-08-31 2009-03-05 Quijano Rodolfo C Self-expanding valve for the venous system
US20090068153A1 (en) * 2007-09-06 2009-03-12 Vitelli Francesca P Cell composition for tissue regeneration
CA2736663C (en) 2007-09-07 2018-01-02 Surgical Biologics, Llc. Placental tissue grafts and improved methods of preparing and using the same
PL2200622T5 (en) 2007-09-19 2016-08-31 Pluristem Ltd Adherent cells from adipose or placenta tissues and use thereof in therapy
WO2009042768A1 (en) * 2007-09-25 2009-04-02 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Triggerably dissolvable hollow fibers for controlled delivery
CN101978045A (en) * 2007-09-26 2011-02-16 细胞基因细胞疗法公司 Angiogenic cells from human placental perfusate
WO2009052209A2 (en) * 2007-10-16 2009-04-23 University Of Kansas Isolation of stem cells and effective control of contamination
KR20210056449A (en) * 2007-11-07 2021-05-18 안트로제네시스 코포레이션 Use of umbilical cord blood in the treatment of premature birth complications
US20110086080A1 (en) * 2007-11-14 2011-04-14 Osteosphere, Llc Ex-vivo production of human demineralized bone matrix
MX2010005805A (en) 2007-11-27 2010-06-09 Lifescan Inc Differentiation of human embryonic stem cells.
US20090163990A1 (en) * 2007-12-19 2009-06-25 Chunlin Yang Decellularized omentum matrix and uses thereof
CN101945994A (en) * 2007-12-19 2011-01-12 再生科学有限责任公司 Compositions and methods to promote implantation and engrafment of stem cells
US20120156134A1 (en) 2007-12-20 2012-06-21 Shayne Squires Compositions and methods for detecting or eliminating senescent cells to diagnose or treat disease
WO2009085860A1 (en) * 2007-12-27 2009-07-09 Ethicon, Incorporated Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
US20100279413A1 (en) * 2008-01-14 2010-11-04 Zymes, Llc Applications of ubiquinones and ubiquinols
US8088163B1 (en) 2008-02-06 2012-01-03 Kleiner Jeffrey B Tools and methods for spinal fusion
US7943573B2 (en) 2008-02-07 2011-05-17 Biomimetic Therapeutics, Inc. Methods for treatment of distraction osteogenesis using PDGF
EP3095470A1 (en) * 2008-02-07 2016-11-23 Shahar Cohen Compartmental extract compositions for tissue engineering
US20090209456A1 (en) * 2008-02-19 2009-08-20 Iliana Sweis Compositions and methods for improving facial and body aesthetics
CN105886459A (en) 2008-02-21 2016-08-24 詹森生物科技公司 Methods, surface modified plates and compositions for cell attachment, cultivation and detachment
US8318485B2 (en) * 2008-02-25 2012-11-27 Natalie Gavrilova Stem cell therapy for the treatment of diabetic retinopathy and diabetic optic neuropathy
EP2257176B1 (en) * 2008-03-14 2013-09-18 Regenerative Sciences, LLC Compositions and methods for cartilage repair
US20090232782A1 (en) * 2008-03-14 2009-09-17 Yu-Show Fu Method for treating brain ischemic injury through transplantation of human umbilical mesenchymal stem cells
CA2721870C (en) 2008-04-21 2020-12-22 Tissue Regeneration Therapeutics, Inc. Genetically modified human umbilical cord perivascular cells for prophylaxis against or treatment of biological or chemical agents
US8623648B2 (en) 2008-04-24 2014-01-07 Janssen Biotech, Inc. Treatment of pluripotent cells
EP2268326B1 (en) 2008-04-30 2016-11-23 Ethicon, Inc Tissue engineered blood vessel
US20110143429A1 (en) * 2008-04-30 2011-06-16 Iksoo Chun Tissue engineered blood vessels
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
WO2010011407A2 (en) * 2008-05-23 2010-01-28 President And Fellows Of Harvard College Methods of generating patterned soft substrates and uses thereof
US20090297485A1 (en) * 2008-05-28 2009-12-03 Allan Mishra Compositions and methods for treating psychiatric and neurodegenerative disorders
US9682173B2 (en) * 2008-06-11 2017-06-20 The Children's Mercy Hospital Solutions for tissue engineering and methods of use
JP2011524174A (en) 2008-06-13 2011-09-01 ホワイトヘッド・インスティテュート・フォー・バイオメディカル・リサーチ Cell programming and reprogramming
WO2009158480A2 (en) * 2008-06-26 2009-12-30 Kci Licensing, Inc. Stimulation of cartilage formation using reduced pressure treatment
WO2009157559A1 (en) * 2008-06-27 2009-12-30 独立行政法人産業技術総合研究所 Pancreatic cell regeneration/transplantation kit for pancreatic diseases or diabetes
CA2729734A1 (en) * 2008-06-30 2010-01-07 Centocor Ortho Biotech Inc. Differentiation of pluripotent stem cells
ES2697798T3 (en) 2008-06-30 2019-01-28 Janssen Biotech Inc Differentiation of pluripotent stem cells
US20100028307A1 (en) * 2008-07-31 2010-02-04 O'neil John J Pluripotent stem cell differentiation
SG190659A1 (en) 2008-09-02 2013-06-28 Pluristem Ltd Adherent cells from placenta tissue and use thereof in therapy
BR122020000059B8 (en) 2008-09-09 2021-06-22 Biomimetic Therapeutics Inc composition comprising a biocompatible matrix and a platelet-derived growth factor and kit
US9446227B2 (en) 2008-09-12 2016-09-20 Sonescence, Inc. Ultrasonic dispersion of compositions in tissue
US20100069827A1 (en) * 2008-09-12 2010-03-18 Barry Neil Silberg Pre-Surgical Prophylactic Administration of Antibiotics and Therapeutic Agents
CN102170919A (en) 2008-10-06 2011-08-31 三维肽胶株式会社 Tissue plug
WO2010048418A1 (en) * 2008-10-22 2010-04-29 The Trustees Of Columbia University In The City Of New York Cartilage regeneration without cell transplantation
CA2742268C (en) 2008-10-31 2020-02-18 Centocor Ortho Biotech Inc. Differentiation of human embryonic stem cells to the pancreatic endocrine lineage
EP2350265B1 (en) * 2008-10-31 2019-04-17 Janssen Biotech, Inc. Differentiation of human embryonic stem cells to the pancreatic endocrine lineage
JP5701765B2 (en) * 2008-10-31 2015-04-15 シンセス ゲーエムベーハー Methods and devices for activating stem cells
BRPI0921494A2 (en) 2008-11-03 2018-10-30 Prad Reasearch And Development Ltd method of planning a underground forming sampling operation, method of controlling a underground forming sampling operation, method of controlling a drilling operation for an underground formation, and method of sampling during the drilling operation.
EP2346452A2 (en) * 2008-11-20 2011-07-27 Lifecell Corporation Method for treatment and prevention of parastomal hernias
ES2584053T3 (en) * 2008-11-20 2016-09-23 Janssen Biotech, Inc. Methods and compositions for cell binding and culture in flat substrates
JP2012509085A (en) 2008-11-20 2012-04-19 ヤンセン バイオテツク,インコーポレーテツド Culture of pluripotent stem cells on microcarriers
WO2010060031A1 (en) * 2008-11-21 2010-05-27 Anthrogenesis Corporation Treatment of diseases, disorders or conditions of the lung using placental cells
WO2010064702A1 (en) * 2008-12-05 2010-06-10 国立大学法人 東京大学 Biomarker for predicting prognosis of cancer
US20110245804A1 (en) 2008-12-05 2011-10-06 Regenerative Sciences, Llc Methods and Compositions to Facilitate Repair of Avascular Tissue
US8366748B2 (en) 2008-12-05 2013-02-05 Kleiner Jeffrey Apparatus and method of spinal implant and fusion
US20100168022A1 (en) * 2008-12-11 2010-07-01 Centeno Christopher J Use of In-Vitro Culture to Design or Test Personalized Treatment Regimens
US20130302283A1 (en) 2012-05-14 2013-11-14 Advanced Technologies And Regenerative Medicine, Llc hUTC MODULATION OF PRO-INFLAMMATORY MEDIATORS OF LUNG AND PULMONARY DISEASES AND DISORDERS
ES2520393T3 (en) * 2008-12-19 2014-11-11 DePuy Synthes Products, LLC Cells derived from umbilical cord tissue to treat neuropathic pain and spasticity
JP6095893B2 (en) * 2008-12-19 2017-03-15 デピュイ・シンセス・プロダクツ・インコーポレイテッド Regeneration and repair of nerve tissue after injury
KR20110107357A (en) * 2008-12-30 2011-09-30 케이씨아이 라이센싱 인코포레이티드 Reduced pressure augmentation of microfracture procedures for cartilage repair
WO2010084943A1 (en) * 2009-01-23 2010-07-29 国立大学法人東京大学 Method for evaluating cultured cells
US9247943B1 (en) 2009-02-06 2016-02-02 Kleiner Intellectual Property, Llc Devices and methods for preparing an intervertebral workspace
WO2010093468A1 (en) * 2009-02-12 2010-08-19 University Of Southern California Bioadhesive patch for sutureless closure of soft tissue
KR101422690B1 (en) * 2009-02-27 2014-07-23 (주)차바이오앤디오스텍 Composition for skin regeneration by using medium or secretion of embryonic stem cell derived endothelial progenitor cells and use thereof
US20120114618A1 (en) * 2009-03-26 2012-05-10 The Regents Of The University Of California Mesenchymal Stem Cells Producing Inhibitory RNA for Disease Modification
US20120039857A1 (en) * 2009-04-06 2012-02-16 Capricor, Inc. Systems and methods for cardiac tissue repair
MX337823B (en) * 2009-05-13 2016-03-22 Medipost Co Ltd Tsp-1, tsp-2, il-17br and hb-egf associated with stem cell activities and applications thereof.
WO2010135527A2 (en) * 2009-05-20 2010-11-25 Humacyte, Inc. Elastin for soft tissue augmentation
EP2434896A4 (en) * 2009-05-28 2014-01-22 Univ Central Florida Res Found In vitro production of oligodendrocytes from human umbilical cord stem cells
US20150335400A1 (en) * 2009-06-17 2015-11-26 The Trustees Of Columbia University In The City Of New York Tooth scaffolds
RU2540016C2 (en) * 2009-07-20 2015-01-27 Янссен Байотек, Инк. Differentiating human embryonic stem cells
CA2768644A1 (en) 2009-07-20 2011-01-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
MX348077B (en) 2009-07-20 2017-05-25 Janssen Biotech Inc Differentiation of human embryonic stem cells.
ES2360434B1 (en) * 2009-07-21 2012-04-12 Universitat Internacional De Catalunya PLURIPOTENTIAL MOTHER CELLS OBTAINED FROM THE DENTAL PULP.
DK2456853T3 (en) * 2009-07-21 2021-02-01 Abt Holding Co USE OF STEM CELLS TO REDUCE LEUCOCYT EXTRAVASATION
WO2011022071A2 (en) * 2009-08-20 2011-02-24 The Regents Of The University Of California Cardiac compositions
US20110054929A1 (en) * 2009-09-01 2011-03-03 Cell Solutions Colorado Llc Stem Cell Marketplace
US8207651B2 (en) 2009-09-16 2012-06-26 Tyco Healthcare Group Lp Low energy or minimum disturbance method for measuring frequency response functions of ultrasonic surgical devices in determining optimum operating point
US9060877B2 (en) 2009-09-18 2015-06-23 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US9629729B2 (en) 2009-09-18 2017-04-25 Spinal Surgical Strategies, Llc Biological delivery system with adaptable fusion cage interface
US10973656B2 (en) 2009-09-18 2021-04-13 Spinal Surgical Strategies, Inc. Bone graft delivery system and method for using same
US8906028B2 (en) 2009-09-18 2014-12-09 Spinal Surgical Strategies, Llc Bone graft delivery device and method of using the same
USD723682S1 (en) 2013-05-03 2015-03-03 Spinal Surgical Strategies, Llc Bone graft delivery tool
USD750249S1 (en) 2014-10-20 2016-02-23 Spinal Surgical Strategies, Llc Expandable fusion cage
US9173694B2 (en) 2009-09-18 2015-11-03 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
US20170238984A1 (en) 2009-09-18 2017-08-24 Spinal Surgical Strategies, Llc Bone graft delivery device with positioning handle
US10245159B1 (en) 2009-09-18 2019-04-02 Spinal Surgical Strategies, Llc Bone graft delivery system and method for using same
US8685031B2 (en) 2009-09-18 2014-04-01 Spinal Surgical Strategies, Llc Bone graft delivery system
US9186193B2 (en) 2009-09-18 2015-11-17 Spinal Surgical Strategies, Llc Fusion cage with combined biological delivery system
EP2483392B1 (en) * 2009-09-23 2015-11-11 Davinci Biosciences LLC Umbilical cord lining stem cells and methods and material for isolating and culturing same
CA2777663A1 (en) 2009-10-16 2011-04-21 University Of Medicine And Dentistry Of New Jersey Method for treating chronic nerve tissue injury using a cell therapy strategy
MX350966B (en) * 2009-10-29 2017-09-26 Janssen Biotech Inc Pluripotent stem cells.
US9855212B2 (en) 2009-10-30 2018-01-02 Abela Pharmaceuticals, Inc. Dimethyl sulfoxide (DMSO) or DMSO and methylsulfonylmethane (MSM) formulations to treat infectious diseases
US9113950B2 (en) 2009-11-04 2015-08-25 Regenerative Sciences, Llc Therapeutic delivery device
EP2498798A4 (en) * 2009-11-10 2014-01-01 Univ Columbia Compositions and methods for wound treatment
WO2011062584A1 (en) * 2009-11-19 2011-05-26 Regents Of The University Of Minnesota Reducing inflammation using cell therapy
KR101867369B1 (en) 2009-12-23 2018-06-14 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
AU2010333840B2 (en) * 2009-12-23 2016-01-07 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
CA2787894A1 (en) 2010-01-26 2011-08-04 Stem Cells Spin S.A. Cell homogenate from stem cells derived from growing deer antlers, a method of obtaining it and its use
US20110212065A1 (en) 2010-02-18 2011-09-01 Timothy Jansen Methods of manufacture of therapeutic products comprising vitalized placental dispersions
EP3769695A1 (en) * 2010-02-19 2021-01-27 LifeCell Corporation Abdominal wall treatment devices
JP6144049B2 (en) 2010-02-22 2017-06-07 バイオミメティック セラピューティクス,リミテッド ライアビリティ カンパニー Platelet-derived growth factor compositions and methods for treating tendon disorders
KR20130025375A (en) 2010-03-01 2013-03-11 얀센 바이오테크 인코포레이티드 Methods for purifying cells derived from pluripotent stem cells
US9845457B2 (en) 2010-04-30 2017-12-19 Cedars-Sinai Medical Center Maintenance of genomic stability in cultured stem cells
US8529883B2 (en) 2010-05-07 2013-09-10 Fibrocell Technologies, Inc. Dosage unit formulations of autologous dermal fibroblasts
CA2800610C (en) 2010-05-12 2019-09-24 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
WO2011153236A1 (en) * 2010-06-03 2011-12-08 The Board Of Trustees Of The Leland Stanford Junior University Purified compositions of cardiovascular progenitor cells
ES2879246T3 (en) * 2010-07-02 2021-11-22 Univ North Carolina Chapel Hill Biomatrix scaffolds
US8825388B2 (en) 2010-07-13 2014-09-02 Qualcomm Incorporated Indoor likelihood heatmap
SG187667A1 (en) * 2010-08-03 2013-03-28 Agency Science Tech & Res Microfabricated scaffold structures
JP2012031127A (en) * 2010-08-03 2012-02-16 Nagoya Univ Composition including umbilical cord matrix stromal cell
IL207586A0 (en) 2010-08-12 2010-12-30 Omrix Biopharmaceuticals Ltd A fibrin based therapeutic preparation and use thereof
WO2012021885A1 (en) * 2010-08-13 2012-02-16 The Trustees Of Columbia University In The City Of New York Three-dimensional tissue engineering devices and uses thereof
CN103154237B (en) 2010-08-31 2016-03-16 詹森生物科技公司 The differentiation of multipotential stem cell
EP2853589B1 (en) 2010-08-31 2017-12-27 Janssen Biotech, Inc. Differentiation of human embryonic stem cells
KR101851956B1 (en) 2010-08-31 2018-04-25 얀센 바이오테크 인코포레이티드 Differentiation of human embryonic stem cells
JP2013536860A (en) * 2010-08-31 2013-09-26 クック・ジェネラル・バイオテクノロジー・エルエルシー Systemic, allogeneic stem cell therapy for the treatment of animal diseases
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
US8546338B2 (en) 2010-12-08 2013-10-01 Johnson & Johnson Consumer Companies, Inc. Self-assembling hydrogels based on dicephalic peptide amphiphiles
JP5388233B2 (en) * 2011-01-19 2014-01-15 富士ソフト株式会社 Method for evaluating the cartilage characteristics of regenerated cartilage
CN102614546B (en) * 2011-01-26 2014-10-15 香港中文大学 Cell sheet for tissue repair and bio-artificial tissue engineering, method of producing the same and method of using the same
ES2613547T3 (en) * 2011-03-18 2017-05-24 Microvascular Tissues, Inc. Allogeneic microvascular tissue for soft tissue treatments
KR101903339B1 (en) 2011-03-22 2018-10-01 플루리스템 리미티드 Methods for treating radiation or chemical injury
CA2834496A1 (en) 2011-04-28 2012-11-01 Aquero Company, Llc Lysine-based polymer coagulants for use in clarification of process waters
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
EP2714111B1 (en) * 2011-05-31 2021-03-17 LifeCell Corporation Adipose tissue matrices
CA2837878A1 (en) 2011-06-10 2012-12-13 Tissuetech, Inc. Methods of processing fetal support tissues, fetal support tissue powder products, and uses thereof
WO2012177927A1 (en) 2011-06-21 2012-12-27 Mayo Foundation For Medical Education And Research Transgenic animals capable of being induced to delete senescent cells
CN103930542A (en) 2011-06-29 2014-07-16 生物修复疗法有限公司 Brown fat cell compositions and methods
US20130005829A1 (en) 2011-06-30 2013-01-03 Advanced Technologies And Regenerative Medicine, Llc. Segmented, epsilon-Caprolactone-Rich, Poly(epsilon-Caprolactone-co-p-Dioxanone) Copolymers for Medical Applications and Devices Therefrom
US10865383B2 (en) 2011-07-12 2020-12-15 Lineage Cell Therapeutics, Inc. Methods and formulations for orthopedic cell therapy
WO2013022447A1 (en) * 2011-08-10 2013-02-14 Advanced Technologies And Regenerative Medicine, Llc Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
JP2015502366A (en) 2011-12-13 2015-01-22 バック インスティテュート フォー リサーチ オン エイジング Methods for improving drug therapy
EP3549615B1 (en) 2011-12-20 2020-12-16 LifeCell Corporation Sheet tissue products
US20130157365A1 (en) * 2011-12-20 2013-06-20 Advanced Technologies And Regenerative Medicine, Llc Induced pluripotent stem cells from human umbilical cord tissue-derived cells
DK2793965T3 (en) 2011-12-20 2019-05-20 Lifecell Corp TISSUE PRODUCTS WITH FLYING DUTIES
BR112014015417A8 (en) 2011-12-22 2017-07-04 Janssen Biotech Inc differentiation of human embryonic stem cells into single hormone insulin positive cells
DK3321355T3 (en) 2011-12-30 2021-11-08 Amit Patel PROCEDURES AND COMPOSITIONS FOR CLINICAL DETERMINATION OF AN ALLOGENIC CELL AND THERAPEUTIC APPLICATIONS
WO2013112350A1 (en) 2012-01-24 2013-08-01 Lifecell Corporation Elongated tissue matrices
EP3338548B1 (en) * 2012-02-23 2020-04-08 Celularity, Inc. Identification of antitumor compounds using placenta
US8940294B2 (en) 2012-03-02 2015-01-27 Tissuetech, Inc. Methods of isolating and culturing stem cells
KR20140131999A (en) 2012-03-07 2014-11-14 얀센 바이오테크 인코포레이티드 Defined Media for Expansion and Maintenance of Pluripotent Stem Cells
US20150064137A1 (en) 2012-04-17 2015-03-05 Kythera Biopharmaceuticals, Inc. Use of engineered viruses to specifically kill senescent cells
EP2841115B1 (en) 2012-04-24 2019-07-10 LifeCell Corporation Functionalized tissue matrices
JP2015521054A (en) 2012-06-05 2015-07-27 カプリコール,インコーポレイテッド Optimized methods for generating cardiac stem cells from heart tissue and their use in cardiac therapy
ES2897649T3 (en) 2012-06-08 2022-03-02 Janssen Biotech Inc Differentiation of human embryonic stem cells into pancreatic endocrine cells
EP3466964A1 (en) * 2012-07-06 2019-04-10 3-D Matrix Ltd. Fill-finish process for peptide solutions
JP6282270B2 (en) * 2012-07-09 2018-02-21 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Neutral protease derived from recombinant Paenibacillus polymixer
US11090338B2 (en) 2012-07-13 2021-08-17 Lifecell Corporation Methods for improved treatment of adipose tissue
US8960674B2 (en) 2012-07-27 2015-02-24 Bally Gaming, Inc. Batch card shuffling apparatuses including multi-card storage compartments, and related methods
CA2881394A1 (en) 2012-08-13 2014-02-20 Cedars-Sinai Medical Center Exosomes and micro-ribonucleic acids for tissue regeneration
US9901081B2 (en) 2012-08-23 2018-02-27 Buck Institute For Research On Aging Transgenic mouse for determining the role of senescent cells in cancer
US9901080B2 (en) 2012-08-23 2018-02-27 Buck Institute For Research On Aging Transgenic mouse having a transgene that converts a prodrug into a cytotoxic compound in senescent cells
US10596202B2 (en) 2012-09-19 2020-03-24 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
WO2014047067A1 (en) 2012-09-19 2014-03-27 Tornier, Inc. Compositions and methods for treating and preventing tissue injury and disease
US11819522B2 (en) 2012-09-19 2023-11-21 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
US9872937B2 (en) 2012-09-19 2018-01-23 Microvascular Tissues, Inc. Compositions and methods for treating and preventing tissue injury and disease
WO2014052376A1 (en) 2012-09-26 2014-04-03 Lifecell Corporation Processed adipose tissue
KR101728808B1 (en) * 2012-09-28 2017-04-20 한국생명공학연구원 Pharmaceutical composition for preventing or treating muscle weakness diseases comprising Acecainide or derivatives thereof
ITTO20120859A1 (en) * 2012-10-02 2014-04-03 Univ Degli Studi Torino NEW THERAPEUTIC APPLICATION OF A MEANS CONDITIONED BY PLACENTARY STEM CELLS
US20150284689A1 (en) * 2012-10-26 2015-10-08 The Regents Of The University Of California Strategy for engineering various 3d tissues, organoids and vasculature
WO2014089124A1 (en) 2012-12-03 2014-06-12 Cenexys, Inc. Immunogenic compositions for inducing an immune response for elimination of senescent cells
US20140170748A1 (en) 2012-12-14 2014-06-19 DePuy Synthes Products, LLC Nutrient Enriched Media for hUTC Growth
SG10201709338RA (en) 2012-12-31 2017-12-28 Janssen Biotech Inc Culturing of human embryonic stem cells at the air-liquid interface for differentiation into pancreatic endocrine cells
CA2896658C (en) 2012-12-31 2021-06-22 Janssen Biotech, Inc. Differentiation of human embryonic stem cells into pancreatic endocrine cells using hb9 regulators
MX2015008619A (en) 2012-12-31 2016-01-12 Janssen Biotech Inc Suspension and clustering of human pluripotent cells for differentiation into pancreatic endocrine cells.
US10370644B2 (en) 2012-12-31 2019-08-06 Janssen Biotech, Inc. Method for making human pluripotent suspension cultures and cells derived therefrom
EP2756754B1 (en) 2013-01-17 2017-01-04 Vita 34 Ag Method for the treatment of umbilical cord tissue, in particular associated with the preservation of the tissue
US20140212390A1 (en) * 2013-01-30 2014-07-31 NuTech Medical, Inc. Placental Membrane Preparation and Methods of Making and Using Same
US9867939B2 (en) 2013-03-12 2018-01-16 Allergan, Inc. Adipose tissue combinations, devices, and uses thereof
US10329529B2 (en) 2013-03-14 2019-06-25 Celularity, Inc. Enhanced placental stem cells and uses thereof
RU2539750C2 (en) * 2013-04-09 2015-01-27 Федеральное государственное бюджетное образовательное учреждение высшего профессионального образования "Московский государственный университет имени М.В. Ломоносова" (МГУ) Method of evaluating immunosuppressive properties of human mesenchymal stromal cells
US20140350516A1 (en) 2013-05-23 2014-11-27 Allergan, Inc. Mechanical syringe accessory
US10058572B2 (en) * 2013-08-15 2018-08-28 The Regents Of The University Of California Placenta-derived multipotent stem cells
US9248384B2 (en) 2013-10-02 2016-02-02 Allergan, Inc. Fat processing system
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
GB2536174B (en) * 2013-12-17 2020-12-16 Dtherapeutics Llc Devices, systems and methods for tissue engineering of luminal grafts
US20170216286A1 (en) 2014-01-28 2017-08-03 Mayo Foundation For Medical Education And Research Killing senescent cells and treating senescence-associated conditions using a src inhibitor and a flavonoid
US10328058B2 (en) 2014-01-28 2019-06-25 Mayo Foundation For Medical Education And Research Treating atherosclerosis by removing senescent foam cell macrophages from atherosclerotic plaques
WO2015116740A1 (en) 2014-01-28 2015-08-06 Buck Institute For Research On Aging Methods and compositions for killing senescent cells and for treating senescence-associated diseases and disorders
EP3797742A1 (en) 2014-02-12 2021-03-31 Gyroscope Therapeutics Limited Apparatus for suprachoroidal administration of therapeutic agent
US10369237B2 (en) 2014-03-10 2019-08-06 3-D Matrix, Ltd. Sterilization and filtration of peptide compositions
WO2015138514A1 (en) 2014-03-10 2015-09-17 3-D Matrix, Ltd. Self-assembling peptide compositions
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
CA2948126A1 (en) * 2014-05-07 2015-11-12 Osiris Therapeutics, Inc. Therapeutic placental compositions, methods of making and methods of use
WO2015170347A2 (en) 2014-05-09 2015-11-12 Reelabs Private Limited Foetal polymix of mesenchymal stem cells under hypoxic conditions for the treatment of clinical disorders
US10029048B2 (en) 2014-05-13 2018-07-24 Allergan, Inc. High force injection devices
EP3954759A1 (en) 2014-05-16 2022-02-16 Janssen Biotech, Inc. Use of small molecules to enhance mafa expression in pancreatic endocrine cells
TW201603818A (en) 2014-06-03 2016-02-01 組織科技股份有限公司 Compositions and methods
US9949874B2 (en) 2014-06-06 2018-04-24 Janssen Biotech, Inc. Therapeutic agent delivery device with convergent lumen
US9925088B2 (en) 2014-06-06 2018-03-27 Janssen Biotech, Inc. Sub-retinal tangential needle catheter guide and introducer
US20160000550A1 (en) * 2014-07-05 2016-01-07 Deborah Nagle Methods for treating diseases of the colon
CN106661552B (en) * 2014-07-11 2020-07-07 玛土撤拉有限公司 Cardiac cell culture material
US10219936B2 (en) 2014-09-11 2019-03-05 Orbit Biomedical Limited Therapeutic agent delivery device with advanceable cannula and needle
US10064752B2 (en) 2014-09-11 2018-09-04 Orbit Biomedical Limited Motorized suprachoroidal injection of therapeutic agent
US10322028B2 (en) 2014-09-11 2019-06-18 Orbit Biomedical Limited Method and apparatus for sensing position between layers of an eye
US10258502B2 (en) 2014-09-18 2019-04-16 Orbit Biomedical Limited Therapeutic agent delivery device
US9504905B2 (en) 2014-09-19 2016-11-29 Bally Gaming, Inc. Card shuffling device and calibration method
KR101613478B1 (en) * 2014-09-22 2016-04-19 (주)안트로젠 Composition comprising mesenchymal stem cell-hydrogel and preparation method thereof
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
CA2962444C (en) 2014-10-03 2023-09-05 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy
EP3206670A4 (en) 2014-10-14 2018-06-20 Samuel Lynch Compositions for treating wounds
US10709811B2 (en) * 2014-10-31 2020-07-14 The Administrators Of The Tulane Educational Fund Surgical grafts for replacing the nipple and areola or damaged epidermis
CN107106613B (en) 2014-11-07 2021-07-06 胞外体干细胞株式会社 Composition comprising stem cell-derived exosomes for adipogenic differentiation induction, adipose tissue regeneration, skin whitening or wrinkle improvement
EP3227682B1 (en) * 2014-12-05 2019-05-01 Meridigen Biotech Co., Ltd. Method of distinguishing mesenchymal stem cells
WO2016090215A2 (en) * 2014-12-05 2016-06-09 Janssen Biotech, Inc. Treatment of ocular conditions using progenitor cells
KR20170094368A (en) * 2014-12-16 2017-08-17 얀센 바이오테크 인코포레이티드 Treatment of retinal degeneration using progenitor cells
US20170080033A1 (en) * 2014-12-16 2017-03-23 Janssen Biotech, Inc. Treatment of retinal degeneration using progenitor cells
JP6800854B2 (en) 2014-12-19 2020-12-16 ヤンセン バイオテツク,インコーポレーテツド Suspension culture of pluripotent stem cells
US10342830B2 (en) 2015-01-05 2019-07-09 Gary M. Petrucci Methods and materials for treating lung disorders
WO2016126122A2 (en) * 2015-02-04 2016-08-11 한양대학교 에리카산학협력단 Composition for chondrocyte differentiation induction or cartilage tissue regeneration, containing exosomes extracted from stem cells differentiating into chondrocytes
US20160243288A1 (en) 2015-02-23 2016-08-25 Tissuetech, Inc. Apparatuses and methods for treating ophthalmic diseases and disorders
SG11201706680SA (en) 2015-03-10 2017-09-28 Allergan Pharmaceuticals Holdings (Ireland) Unlimited Company Multiple needle injector
CA2979293C (en) 2015-03-11 2022-01-04 Timothy J. Kieffer Pancreatic endocrine progenitor cell therapies for the treatment of obesity and type 2 diabetes (t2d)
US11027047B2 (en) 2015-03-31 2021-06-08 The University Of North Carolina At Chapel Hill Delivery vehicles for stem cells and uses thereof
US10286009B2 (en) * 2015-05-16 2019-05-14 Asterias Biotherapeutics, Inc. Pluripotent stem cell-derived oligodendrocyte progenitor cells for the treatment of spinal cord injury
JP2018516869A (en) 2015-05-20 2018-06-28 ティッシュテック,インク. Compositions and methods for preventing epithelial cell proliferation and epithelial-mesenchymal transition
EP3297694A1 (en) 2015-05-21 2018-03-28 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
US10335435B2 (en) 2015-05-22 2019-07-02 Marco Merida Method for endoscopically delivering stem cells to the brain using an intranasal, injectable approach
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US10384207B2 (en) 2015-07-21 2019-08-20 Neuro Probe Incorporated Assay apparatus and methods
WO2017019822A1 (en) * 2015-07-29 2017-02-02 Medivation Technologies, Inc. Pellet composition containing repair cells
WO2017019832A1 (en) 2015-07-29 2017-02-02 Medivation Technologies, Inc. Methods and compositions using repair cells and cationic dyes
CN108350427B (en) 2015-08-28 2022-03-11 日本乐敦制药株式会社 ROR1 positive mesenchymal stem cells and pharmaceutical composition comprising same
USD797290S1 (en) 2015-10-19 2017-09-12 Spinal Surgical Strategies, Llc Bone graft delivery tool
US11384328B2 (en) 2015-11-18 2022-07-12 President And Fellows Of Harvard College Cartridge-based system for long term culture of cell clusters
CN108291199A (en) * 2015-12-04 2018-07-17 詹森生物科技公司 Retinosis is treated using progenitor cells
KR20170076484A (en) * 2015-12-24 2017-07-04 삼성전자주식회사 Method of isolating senescent cell with high expression of Protocadherin gene
WO2017120092A1 (en) 2016-01-06 2017-07-13 3-D Matrix, Ltd. Combination compositions
EP3402543B1 (en) 2016-01-11 2021-09-08 Cedars-Sinai Medical Center Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction
CA3010792A1 (en) 2016-01-14 2017-07-20 DePuy Synthes Products, Inc. Composition and methods for cryopreservation of hutc
TW201733600A (en) 2016-01-29 2017-10-01 帝聖工業公司 Fetal support tissue products and methods of use
WO2017136557A1 (en) * 2016-02-05 2017-08-10 Petrucci Gary M Methods and materials for treating nerve injuries and neurological disorders
US11458224B2 (en) * 2016-03-04 2022-10-04 University of Pittsburgh—of the Commonwealth System of Higher Education Ovarian-derived hydrogels for biomedical and biotechnology applications
US10478553B2 (en) 2016-03-09 2019-11-19 Orbit Biomedical Limited Apparatus for subretinal administration of therapeutic agent via a curved needle
GB201604304D0 (en) 2016-03-14 2016-04-27 Tigenix S A U Adipose tissue-derived stromal stem cells for use in treating refractory complex perianal fistulas in crohn's disease
EP3223181B1 (en) * 2016-03-24 2019-12-18 Sofradim Production System and method of generating a model and simulating an effect on a surgical repair site
US11920155B2 (en) 2016-03-30 2024-03-05 Asterias Biotherapeutics, Inc. Oligodendrocyte progenitor cell compositions
KR102232054B1 (en) 2016-04-08 2021-03-26 알레간 인코포레이티드 Suction and injection device
MA45479A (en) 2016-04-14 2019-02-20 Janssen Biotech Inc DIFFERENTIATION OF PLURIPOTENT STEM CELLS IN ENDODERMAL CELLS OF MIDDLE INTESTINE
WO2017188403A1 (en) 2016-04-27 2017-11-02 ロート製薬株式会社 Mesenchymal stem cell expressing at least one cell surface marker selected from the group consisting of cd201, cd46, cd56, cd147, and cd165 and method for preparing same, and pharmaceutical composition containing the mesenchymal stem cell and method for preparing same
WO2017210652A1 (en) 2016-06-03 2017-12-07 Cedars-Sinai Medical Center Cdc-derived exosomes for treatment of ventricular tachyarrythmias
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US10806629B2 (en) 2016-06-17 2020-10-20 Gyroscope Therapeutics Limited Injection device for subretinal delivery of therapeutic agent
US10646374B2 (en) 2016-06-17 2020-05-12 Orbit Biomedical Limited Apparatus and method to form entry bleb for subretinal delivery of therapeutic agent
US11000410B2 (en) 2016-06-17 2021-05-11 Gyroscope Therapeutics Limited Guide apparatus for tangential entry into suprachoroidal space
MA45502A (en) 2016-06-21 2019-04-24 Janssen Biotech Inc GENERATION OF FUNCTIONAL BETA CELLS DERIVED FROM HUMAN PLURIPOTENT STEM CELLS WITH GLUCOSE-DEPENDENT MITOCHONDRIAL RESPIRATION AND TWO-PHASE INSULIN SECRETION RESPONSE
EP3777907A1 (en) 2016-07-05 2021-02-17 LifeCell Corporation Tissue matrices incorporating multiple tissue types
SG11201811100RA (en) * 2016-07-05 2019-01-30 Janssen Biotech Inc Treatment of retinal vascular disease using progenitor cells
US20190184060A1 (en) * 2016-08-19 2019-06-20 Regentys Corporation Extracellular matrix for tissue reconstruction of mucosal tissue
EP3405204A4 (en) * 2016-08-26 2020-03-18 Restem Llc Composition and methods of using umbilical cord lining stem cells
EP3515459A4 (en) 2016-09-20 2020-08-05 Cedars-Sinai Medical Center Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders
EP3534981A4 (en) * 2016-11-02 2020-06-24 Axogen Corporation Amnion tissue grafts and methods of preparing and using same
RU2644306C1 (en) * 2016-11-22 2018-02-08 Общество с ограниченной ответственностью "ДЖИ-Групп" Method of recovery of defects of covering tissues
WO2018125851A1 (en) * 2016-12-26 2018-07-05 Michael Moeller Systems and methods to isolate and expand stem cells from urine
US11273072B2 (en) 2017-01-13 2022-03-15 Gyroscope Therapeutics Limited Suprachoroidal injection device
JPWO2018164228A1 (en) 2017-03-08 2020-01-09 ロート製薬株式会社 Pharmaceutical composition for preventing or treating a disease associated with fibrosis, comprising a ROR1-positive mesenchymal stem cell, a method for preparing the same, and prevention or treatment of a disease associated with fibrosis using ROR1-positive mesenchymal stem cell Treatment method
US11076984B2 (en) 2017-03-13 2021-08-03 Gyroscope Therapeutics Limited Method of performing subretinal drainage and agent delivery
EP3595680A1 (en) * 2017-03-17 2020-01-22 Rutgers, The State University of New Jersey Compositions and methods for wound healing
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
KR20190141695A (en) 2017-04-13 2019-12-24 센티 바이오사이언시스, 인코포레이티드 Combination Cancer Immunotherapy
EP3612191A4 (en) 2017-04-19 2020-12-30 Cedars-Sinai Medical Center Methods and compositions for treating skeletal muscular dystrophy
US10478531B2 (en) 2017-06-22 2019-11-19 Gary M. Petrucci Methods and materials for treating blood vessels
JP7195033B2 (en) 2017-07-18 2022-12-23 ザ リサーチ ファウンデイション フォー ザ ステイト ユニバーシティー オブ ニューヨーク Biomarkers for intracranial aneurysms
US10251917B1 (en) 2017-09-19 2019-04-09 Gary M. Petrucci Methods and materials for treating tumors
US11123375B2 (en) 2017-10-18 2021-09-21 Lifecell Corporation Methods of treating tissue voids following removal of implantable infusion ports using adipose tissue products
AU2018351051A1 (en) 2017-10-18 2020-03-19 Lifecell Corporation Adipose tissue products and methods of production
CA3075106A1 (en) 2017-10-19 2019-04-25 Lifecell Corporation Flowable acellular tissue matrix products and methods of production
US11246994B2 (en) 2017-10-19 2022-02-15 Lifecell Corporation Methods for introduction of flowable acellular tissue matrix products into a hand
WO2019079681A1 (en) 2017-10-20 2019-04-25 President And Fellows Of Harvard College Methods for producing mature adipocytes and methods of use thereof
WO2019083995A1 (en) * 2017-10-23 2019-05-02 Cell Medicine, Inc. Mesenchymal stem cell therapy of leigh syndrome
WO2019087130A2 (en) * 2017-11-03 2019-05-09 Janssen Biotech, Inc. Method of inhibiting angiogenesis
US11660355B2 (en) 2017-12-20 2023-05-30 Cedars-Sinai Medical Center Engineered extracellular vesicles for enhanced tissue delivery
US11285177B2 (en) 2018-01-03 2022-03-29 Globus Medical, Inc. Allografts containing viable cells and methods thereof
CN111565732A (en) 2018-01-12 2020-08-21 国立大学法人大阪大学 Agent for promoting normal differentiation-maturation of stratified squamous epithelial cell, agent for treating epithelial disease, and method for promoting normal differentiation-maturation of stratified squamous epithelial cell
US20190218521A1 (en) * 2018-01-18 2019-07-18 Lorenzo Bracco Culture medium of viruses for human vaccines have to consist of human cells from placenta and/or from umbilical cord of a fetus of the blood type 0 Rh- and of the mother of the blood type 0 Rh- (both, fetus and mother, must be of the blood type 0 Rh-)
WO2019152483A1 (en) * 2018-01-30 2019-08-08 University Of Georgia Research Foundation, Inc. Methods for vascular construction and products therefrom
CN108384752A (en) * 2018-02-13 2018-08-10 中国人民解放军第四五五医院 Application at Cartilage culture base and its in micro- splitting technique in knee cartilage differentiation
JP2021526556A (en) * 2018-05-30 2021-10-07 ダイレクト バイオロジクス エルエルシー Auxiliary agents containing mesenchymal stem cell (MSC) preparations, which are frozen or powdered growth factors and extracellular vesicles, and how to use them.
CN108961233A (en) * 2018-06-28 2018-12-07 华侨大学 A kind of composite polycrystal-diamond surface defect classifying identification method
US11419898B2 (en) 2018-10-17 2022-08-23 Senti Biosciences, Inc. Combinatorial cancer immunotherapy
WO2020081869A1 (en) 2018-10-17 2020-04-23 Senti Biosciences, Inc. Combinatorial cancer immunotherapy
JP2022520722A (en) 2019-01-23 2022-04-01 アステリアス バイオセラピューティクス インコーポレイテッド Dorsal-derived oligodendrocyte progenitor cells from human pluripotent stem cells
US11759355B1 (en) 2019-02-26 2023-09-19 Gyroscope Therapeutics Limited Method of delivering leading blebs and agent to subretinal space
KR102169924B1 (en) * 2019-03-26 2020-10-26 연세대학교 산학협력단 Composition for inducing chondrogenesis and use thereof
KR102167257B1 (en) * 2019-05-28 2020-10-19 부산대학교 산학협력단 Method of promoting differentiation of stem cells into mature cardiomyocytes using tomatidine
MX2021014654A (en) 2019-05-30 2022-03-11 Lifecell Corp Biologic breast implant.
CN114286860A (en) * 2019-08-29 2022-04-05 味之素株式会社 Method for producing mesenchymal stem cells from biological cell sample containing mesenchymal stem cells
WO2021050907A1 (en) * 2019-09-13 2021-03-18 The University Of North Carolina At Chapel Hill Method of making human mouse xenografts
WO2021071875A1 (en) * 2019-10-07 2021-04-15 University Of Utah Research Foundation Chondrogenic human mesenchymal stem cell (msc) sheets
IL292024A (en) 2019-10-11 2022-06-01 Gyroscope Therapeutics Ltd Dose clip assembly for syringe
CN111718410B (en) * 2020-06-05 2022-10-11 江南大学 Method for preparing yolk immunoglobulin
JP6967308B1 (en) 2020-06-30 2021-11-17 国立大学法人高知大学 Cranial nerve disorder therapeutic agent containing tissue cell culture supernatant derived from fetal appendages
WO2022046954A1 (en) * 2020-08-25 2022-03-03 Celularity Inc. Appl cells improved placental-derived adherent cells and methods of their use
US20230390111A1 (en) 2020-12-22 2023-12-07 Gyroscope Therapeutics Limited Ocular cannula guide
EP4279919A1 (en) * 2021-01-12 2023-11-22 Cellatoz Therapeutics, Inc. Method for screening neuronal regeneration promoting cells having neuronal regeneration activity
CN115068438B (en) * 2022-04-28 2023-09-22 浙江大学医学院附属邵逸夫医院 Method for preparing cell membrane nano vesicle with osteoclast precursor homology targeting and application thereof
CN115044542B (en) * 2022-06-30 2023-09-26 上海市东方医院(同济大学附属东方医院) Application of SJ000291942 in inducing mesenchymal stem cells to osteogenic differentiation
WO2024024708A1 (en) * 2022-07-26 2024-02-01 国立大学法人大阪大学 Composition for cartilage repair and method for manufacturing same
WO2024052733A1 (en) 2022-09-06 2024-03-14 Gyroscope Therapeutics Limited Apparatus for subretinal administration of therapeutic agent via dual-curved needle

Citations (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3665061A (en) * 1969-07-16 1972-05-23 United States Banknote Corp Process for producing collagen sponges
US3930954A (en) * 1973-05-31 1976-01-06 Godo Shusei Kabushiki Kaisha Neutral protease useful for animal tissue and cell culture
US4925667A (en) * 1986-05-27 1990-05-15 Qmax Technology Group, Inc. Substrate with particulate cosmetic
US5004681A (en) * 1987-11-12 1991-04-02 Biocyte Corporation Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US5286632A (en) * 1991-01-09 1994-02-15 Jones Douglas H Method for in vivo recombination and mutagenesis
US5320962A (en) * 1992-07-22 1994-06-14 Duke University DNA encoding the human A1 adenosine receptor
US5336616A (en) * 1990-09-12 1994-08-09 Lifecell Corporation Method for processing and preserving collagen-based tissues for transplantation
US5342761A (en) * 1990-10-01 1994-08-30 Research Development Foundation Oncofetal gene, gene product and uses therefor
US5437994A (en) * 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5443950A (en) * 1986-04-18 1995-08-22 Advanced Tissue Sciences, Inc. Three-dimensional cell and tissue culture system
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5707643A (en) * 1993-02-26 1998-01-13 Santen Pharmaceutical Co., Ltd. Biodegradable scleral plug
US5718922A (en) * 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
US5736516A (en) * 1995-11-29 1998-04-07 Amgen Inc. Methods for treating photoreceptors using glial cell line-derived neurotrophic factor (GDNF) protein protein
US5869079A (en) * 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
US5902598A (en) * 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
US5902741A (en) * 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US5906934A (en) * 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5919702A (en) * 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
US5935849A (en) * 1994-07-20 1999-08-10 Cytotherapeutics, Inc. Methods and compositions of growth control for cells encapsulated within bioartificial organs
US6022743A (en) * 1986-04-18 2000-02-08 Advanced Tissue Sciences, Inc. Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
US6059968A (en) * 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
US6179872B1 (en) * 1998-03-17 2001-01-30 Tissue Engineering Biopolymer matt for use in tissue repair and reconstruction
US6200606B1 (en) * 1996-01-16 2001-03-13 Depuy Orthopaedics, Inc. Isolation of precursor cells from hematopoietic and nonhematopoietic tissues and their use in vivo bone and cartilage regeneration
US6200806B1 (en) * 1995-01-20 2001-03-13 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US6221904B1 (en) * 1997-05-21 2001-04-24 Sloan-Kettering Institute For Cancer Research Method for increasing the concentration of ascorbic acid in brain tissues of a subject
US6251090B1 (en) * 1994-12-12 2001-06-26 Robert Logan Avery Intravitreal medicine delivery
US6335239B1 (en) * 1998-12-04 2002-01-01 International Business Machines Corporation Manufacturing a DRAM cell having an annular signal transfer region
US20020022676A1 (en) * 1999-04-16 2002-02-21 Shulin He Poly(Propylene Fumarate) cross linked with Poly(Ethylene Glycol)
US20020028510A1 (en) * 2000-03-09 2002-03-07 Paul Sanberg Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US6355699B1 (en) * 1999-06-30 2002-03-12 Ethicon, Inc. Process for manufacturing biomedical foams
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US6372494B1 (en) * 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
US6375972B1 (en) * 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
US6387367B1 (en) * 1998-05-29 2002-05-14 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US6391297B1 (en) * 1997-12-02 2002-05-21 Artecel Sciences, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US20020062151A1 (en) * 1999-05-14 2002-05-23 Gregory Altman Bioengineered anterior cruciate ligament
US20020081725A1 (en) * 2000-06-30 2002-06-27 Wen-Ghih Tsang Culturing pancreatic stem cells having a specified, intermediate stage of development
US20020098584A1 (en) * 2000-11-06 2002-07-25 Palmer Theo D. Postmortem stem cells
US20030003574A1 (en) * 2000-01-24 2003-01-02 Jean Toma Multipotent stem cells from peripheral tissues and uses thereof
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US20030022369A1 (en) * 2001-05-18 2003-01-30 Helen Fillmore Differentiation of specialized dermal and epidermal cells into neuronal cells
US20030032179A1 (en) * 2000-12-06 2003-02-13 Hariri Robert J. Post-partum mammalian placenta, its use and placental stem cells therefrom
US20030031657A1 (en) * 1999-12-06 2003-02-13 Massachusetts General Hospital Stem cells and their use in transplantation
US20030032183A1 (en) * 2001-05-25 2003-02-13 Sheridan Steven D. Stem cell differentiation
US20030032178A1 (en) * 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
US6528245B2 (en) * 1998-05-07 2003-03-04 University Of South Florida Bone marrow cells as a source of neurons for brain and spinal cord repair
US20030049837A1 (en) * 1991-07-08 2003-03-13 Samuel Weiss In vitro and in vivo proliferation and use of multipotent neural stem cells and their progeny
US6534084B1 (en) * 1999-06-30 2003-03-18 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US20030059939A1 (en) * 2001-08-27 2003-03-27 Raymond Page Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US6555374B1 (en) * 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US20030082160A1 (en) * 2001-10-25 2003-05-01 Yu John S. Differentiation of whole bone marrow
US20030082155A1 (en) * 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20030096409A1 (en) * 2001-11-22 2003-05-22 Shigeru Yasumoto Cultured skin and method of manufacturing the same
US20030104997A1 (en) * 2001-09-05 2003-06-05 Black Ira B. Multi-lineage directed induction of bone marrow stromal cell differentiation
US20030109036A1 (en) * 2001-12-06 2003-06-12 The Regents Of The University Of California Method for differentiating islet precursor cells into beta cells
US20030113910A1 (en) * 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
US20030118566A1 (en) * 2001-08-08 2003-06-26 Toomas Neuman Compositions and methods for isolation, propagation, and differentiation of human stem cells and uses thereof
US20030124721A1 (en) * 2001-11-09 2003-07-03 Bentley Cheatham Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
US20030138948A1 (en) * 2001-12-07 2003-07-24 Fisk Gregory J. Islet cells from human embryonic stem cells
US20030138951A1 (en) * 2001-10-18 2003-07-24 Li Yin Conversion of liver stem and progenitor cells to pancreatic functional cells
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
US6673606B1 (en) * 2000-04-12 2004-01-06 The Children's Hospital Of Philadelphia Therapeutic uses for mesenchymal stromal cells
US20040005704A1 (en) * 1998-11-18 2004-01-08 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US20040009593A1 (en) * 2002-07-11 2004-01-15 Keirstead Hans S. Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US6680198B1 (en) * 1998-08-14 2004-01-20 The Children's Medical Center Corporation Engraftable human neural stem cells
US20040014206A1 (en) * 1999-10-28 2004-01-22 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20040014211A1 (en) * 2002-06-11 2004-01-22 Ogle Roy C. Meningeal-derived stem cells
US20040014662A1 (en) * 2002-05-08 2004-01-22 Per Lindquist Modulation of neural stem cells and neural progenitor cells
US20040014210A1 (en) * 2002-07-16 2004-01-22 Jessell Thomas M. Methods for inducing differentiation of embryonic stem cells and uses thereof
US6686198B1 (en) * 1993-10-14 2004-02-03 President And Fellows Of Harvard College Method of inducing and maintaining neuronal cells
US20040028660A1 (en) * 2002-05-30 2004-02-12 Anthrogenesis Corporation Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US20040029269A1 (en) * 2002-05-07 2004-02-12 Goldman Steven A Promoter-based isolation, purification, expansion, and transplantation of neuronal progenitor cells, oligodendrocyte progenitor cells, or neural stem cells from a population of embryonic stem cells
US20040033597A1 (en) * 1999-08-29 2004-02-19 Jean Toma Multipotent neural stemcells from peripheral tissues and uses thereof
US20040037818A1 (en) * 1998-07-30 2004-02-26 Brand Stephen J. Treatment for diabetes
US6703017B1 (en) * 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US20040058412A1 (en) * 2002-09-20 2004-03-25 Neuronyx, Inc. Cell populations which co-express CD49c and CD90
US20040063202A1 (en) * 2002-08-28 2004-04-01 Petersen Bryon E. Neurogenesis from hepatic stem cells
US20040072344A1 (en) * 2002-01-25 2004-04-15 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
US20050037491A1 (en) * 2003-06-27 2005-02-17 Sanjay Mistry Repair and regeneration of ocular tissue using postpartum-derived cells
US20050074435A1 (en) * 2001-12-21 2005-04-07 Robert Casper Cellular compositions and methods of making and using them
US20060128014A1 (en) * 2002-08-26 2006-06-15 Johan Haggblad Compositions and methods for culturing stem cells
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
US20070141700A1 (en) * 2005-12-19 2007-06-21 Ethicon, Incorporated In vitro expansion of postpartum-derived cells in roller bottles
US20070160588A1 (en) * 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
US20080112939A1 (en) * 2006-10-12 2008-05-15 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US20080145934A1 (en) * 2004-09-28 2008-06-19 Ian Ross Harris Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers
US7875273B2 (en) * 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US7875272B2 (en) * 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells

Family Cites Families (234)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US229971A (en) 1880-07-13 Ttornets
US2324800A (en) 1941-08-14 1943-07-20 Pfizer Charles & Co Purification of riboflavin
US2654735A (en) * 1949-07-29 1953-10-06 Us Vitamin Corp Process for the production of derivatives of 9-polyhydroxyalkylisoalloxazines and products obtained
US2864848A (en) * 1954-07-19 1958-12-16 Ca Nat Research Council Method of producing l-alpha-glycerylphosphorylcholine
US2912332A (en) 1958-02-27 1959-11-10 Swift & Co Stabilized thiamine composition and method of enriching food products
CH635748A5 (en) 1977-08-16 1983-04-29 Cellorgan Laboratoires Sa PROCESS FOR OBTAINING PLACENTAL CELLS OF SHEEP.
US4216144A (en) * 1977-10-20 1980-08-05 Ashmead H H Soluble iron proteinates
JPS6040439B2 (en) 1978-03-29 1985-09-11 大正製薬株式会社 hydrocortisone derivatives
US4352883A (en) 1979-03-28 1982-10-05 Damon Corporation Encapsulation of biological material
US4393240A (en) 1981-07-06 1983-07-12 Stille John K Optically active phosphines
US4544516A (en) 1982-07-28 1985-10-01 Battelle Development Corporation Collagen orientation
US4465776A (en) 1982-09-27 1984-08-14 Research Corporation Monoclonal antibodies to vitamin B6 and immunoassay method
US4657866A (en) * 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
US4487865A (en) 1983-12-15 1984-12-11 Biomatrix, Inc. Polymeric articles modified with hyaluronate
US4963489A (en) 1987-04-14 1990-10-16 Marrow-Tech, Inc. Three-dimensional cell and tissue culture system
JPS642643A (en) 1987-06-26 1989-01-06 Bio Material Yunibaasu:Kk Artificial skin
NZ226750A (en) 1987-10-29 1990-09-26 Amrad Corp Ltd Immortalisation of neural precursor cells by introducing a retrovirus vector containing a myc-oncogene
US5162405A (en) * 1987-12-24 1992-11-10 Elf Atochem North America, Inc. Single-functional and mixtures of multi-functional oligomeric performance additive compositions and their uses
GB8803697D0 (en) 1988-02-17 1988-03-16 Deltanine Research Ltd Clinical developments using amniotic membrane cells
US4963439A (en) 1988-04-19 1990-10-16 Ube Industries, Ltd. Continuous fiber-reinforced Al-Co alloy matrix composite
US5618670A (en) * 1988-08-26 1997-04-08 The United States Of America As Represented By The Department Of Health & Human Services Detection method for c-raf-1 genes
US4925677A (en) * 1988-08-31 1990-05-15 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
US5284766A (en) * 1989-02-10 1994-02-08 Kao Corporation Bed material for cell culture
JPH06104061B2 (en) 1989-02-10 1994-12-21 花王株式会社 Cell culture support material
DE69009237T2 (en) * 1989-02-27 1994-12-01 Takasago Perfumery Co Ltd Process for the preparation of optically active esters of 6-t-butoxy-3,5-dihydroxyhexanoic acid.
FR2646438B1 (en) 1989-03-20 2007-11-02 Pasteur Institut A METHOD FOR SPECIFIC REPLACEMENT OF A COPY OF A GENE PRESENT IN THE RECEIVER GENOME BY INTEGRATION OF A GENE DIFFERENT FROM THAT OR INTEGRATION
US5574205A (en) 1989-07-25 1996-11-12 Cell Genesys Homologous recombination for universal donor cells and chimeric mammalian hosts
US5840580A (en) 1990-05-01 1998-11-24 Becton Dickinson And Company Phenotypic characterization of the hematopoietic stem cell
US5145770A (en) * 1990-06-04 1992-09-08 Biosurface Technology, Inc. Cryopreservation of cultured epithelial sheets
WO1992003917A1 (en) 1990-08-29 1992-03-19 Genpharm International Homologous recombination in mammalian cells
US5506134A (en) * 1990-10-22 1996-04-09 Corvas International, Inc. Hypridoma and monoclonal antibody which inhibits blood coagulation tissue factor/factor VIIa complex
US5811094A (en) 1990-11-16 1998-09-22 Osiris Therapeutics, Inc. Connective tissue regeneration using human mesenchymal stem cell preparations
US5140100A (en) * 1990-12-28 1992-08-18 Cedars-Sinai Medical Center Protein that inhibits production of human choriogonadotropin
NL9100038A (en) 1991-01-11 1992-08-03 Stamicarbon ENZYME-CATALYZED PREPARATION OF OPTICALLY ACTIVE CARBONIC ACIDS.
CZ232593A3 (en) 1991-05-02 1994-07-13 Yeda Res & Dev Pharmaceutical preparation for preventing and/or therapy of pathological states
WO1993003139A1 (en) 1991-08-08 1993-02-18 Kao Corporation Cell culture support, production thereof, and production of cell cluster using same
EP0529751A1 (en) 1991-08-09 1993-03-03 W.R. Grace & Co.-Conn. Cell culture substrate, test material for cell culture and preparations thereof
AU2515992A (en) 1991-08-20 1993-03-16 Genpharm International, Inc. Gene targeting in animal cells using isogenic dna constructs
WO1993006726A1 (en) * 1991-09-30 1993-04-15 Mackenzie Walser Methods for treatment of free-radical-mediated tissue injury
US5308763A (en) * 1991-10-01 1994-05-03 The Johns Hopkins University Method of making primary culture of olfactory neurons
US5914265A (en) * 1992-04-30 1999-06-22 Baylor College Of Medicine Keratin K1 expression vectors and methods of use
AU4543193A (en) 1992-06-22 1994-01-24 Henry E. Young Scar inhibitory factor and use thereof
US5589376A (en) 1992-07-27 1996-12-31 California Institute Of Technology Mammalian neural crest stem cells
US5356807A (en) * 1992-09-08 1994-10-18 Cornell Research Foundation Cultured cell line of adult diploid cells from human brain and meningeal tissue
US20040224409A1 (en) 1992-09-25 2004-11-11 Laurent Pradier Recombinant adenoviruses coding for brain-derived neurotrophic factor (BDNF)
WO1994010331A1 (en) 1992-10-29 1994-05-11 The Australian National University Angiogenesis inhibitory antibodies
US5670483A (en) 1992-12-28 1997-09-23 Massachusetts Insititute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5955343A (en) 1992-12-28 1999-09-21 Massachusetts Institute Of Technology Stable macroscopic membranes formed by self-assembly of amphiphilic peptides and uses therefor
US5494899A (en) * 1993-04-07 1996-02-27 Oklahoma Medical Research Foundation Selective regulation of B lymphocyte precursors by hormones
WO1994025584A1 (en) 1993-04-28 1994-11-10 Johns Hopkins University School Of Medicine Chronic endothelial cell culture under flow
IL110589A0 (en) 1993-08-10 1994-11-11 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF- beta family
JP3680114B2 (en) 1993-09-17 2005-08-10 敏一 中村 Cranial nerve disorder treatment
US6432711B1 (en) 1993-11-03 2002-08-13 Diacrin, Inc. Embryonic stem cells capable of differentiating into desired cell lines
US5456835A (en) * 1993-11-08 1995-10-10 Hemasure, Inc. Device and process for removing free hemoglobin from blood
DE4406073A1 (en) * 1994-02-24 1995-08-31 Univ Ludwigs Albert Process for the production of human, clonogenic fibroblasts, process for gene transfection of fibroblasts and fibroblasts obtained in this way
US5698518A (en) 1994-03-30 1997-12-16 Oklahoma Medical Research Foundation Method for regulating inflammation and tumor growth with calmodulin, calmodulin analogues or calmodulin antagonists
US5466233A (en) 1994-04-25 1995-11-14 Escalon Ophthalmics, Inc. Tack for intraocular drug delivery and method for inserting and removing same
US5834308A (en) 1994-04-28 1998-11-10 University Of Florida Research Foundation, Inc. In vitro growth of functional islets of Langerhans
US6001647A (en) 1994-04-28 1999-12-14 Ixion Biotechnology, Inc. In vitro growth of functional islets of Langerhans and in vivo uses thereof
DE69531638T2 (en) 1994-06-06 2004-06-17 Osiris Therapeutics, Inc. BIOMATRIX FOR TISSUE REGENATION
IL114397A0 (en) 1994-07-01 1995-10-31 Bioph Biotech Entw Pharm Gmbh Growth/differentiation factor of the TGF-beta-family
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
US5660982A (en) * 1994-10-04 1997-08-26 Tryggvason; Karl Laminin chains: diagnostic uses
US5789147A (en) 1994-12-05 1998-08-04 New York Blood Center, Inc. Method for concentrating white cells from whole blood by adding a red cell sedimentation reagent to whole anticoagulated blood
US5684032A (en) 1994-12-13 1997-11-04 Smithkline Beecham Corporation Compounds
US5736396A (en) 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
US5693332C1 (en) 1995-08-11 2001-01-09 Univ California Human keratinocytes supported on a hydrophilic membrane and methods of using same to effect wound closure
US5842477A (en) 1996-02-21 1998-12-01 Advanced Tissue Sciences, Inc. Method for repairing cartilage
JP4307552B2 (en) 1996-03-15 2009-08-05 ミューニン コーポレイション Extracellular matrix signaling molecules
US6223188B1 (en) * 1996-04-10 2001-04-24 Sun Microsystems, Inc. Presentation of link information as an aid to hypermedia navigation
WO1997041208A1 (en) 1996-04-26 1997-11-06 Case Western Reserve University Skin regeneration using mesenchymal stem cells
WO1998031316A1 (en) 1997-01-17 1998-07-23 Celadon Science, Llc Methods for promoting healing of corneal resurfacing wounds
AU6144698A (en) 1997-02-05 1998-08-25 Case Western Reserve University Stimulatory effects of bfgf and bmp-2 on osteogenic differentiation of mesenchymal stem cells
EP2110431A1 (en) 1997-05-13 2009-10-21 Osiris Therapeutics, Inc. Cartilage regeneration using human mesenchymal stem cells
AU738334B2 (en) * 1997-05-30 2001-09-13 Osteobiologics, Inc. Fiber-reinforced, porous, biodegradable implant device
ATE307195T1 (en) 1997-07-14 2005-11-15 Osiris Therapeutics Inc CARDIAC MUSCLE REGENERATION USING MESENCHYMAL STEM CELLS
US6291240B1 (en) * 1998-01-29 2001-09-18 Advanced Tissue Sciences, Inc. Cells or tissues with increased protein factors and methods of making and using same
EP1062321B1 (en) * 1998-03-13 2004-12-29 Osiris Therapeutics, Inc. Uses for humane non-autologous mesenchymal stem cells
EP1076563B1 (en) 1998-03-16 2005-05-11 Cytovia, Inc. Dipeptide caspase inhibitors and the use thereof
US6171610B1 (en) 1998-04-24 2001-01-09 University Of Massachusetts Guided development and support of hydrogel-cell compositions
US6323188B1 (en) 1998-07-01 2001-11-27 Donald L. Weissman Treatment and prevention of cardiovascular diseases, heart attack, and stroke, primary and subsequent, with help of aspirin and certain vitamins
DE19833340A1 (en) 1998-07-24 2000-02-10 Karlsruhe Forschzent Worm-shaped working mechanism
AU776514B2 (en) 1998-08-10 2004-09-09 General Hospital Corporation, The Differentiation of non-insulin producing cells into insulin producing cells by GLP-1 or exendin-4 and uses thereof
US6284245B1 (en) 1998-08-25 2001-09-04 Diacrin, Inc. Neural retinal cells and retinal pigment epithelium cells and their use in treatment of retinal disorders
US6444205B2 (en) 1998-09-30 2002-09-03 Diacrin, Inc. Transplantation of neural cells for the treatment of chronic pain or spasticity
US6241369B1 (en) * 1998-11-20 2001-06-05 Cooper Technologies Company Quick mount fixture
DE19856428C1 (en) * 1998-12-08 2000-05-04 Heraeus Noblelight Gmbh Discharge lamp, having spiral, inner electrode arranged at inner wall of interior tube
WO2000038762A1 (en) 1998-12-24 2000-07-06 Biosafe S.A. Blood separation system particularly for concentrating hematopoietic stem cells
JP4932069B2 (en) 1999-01-25 2012-05-16 株式会社セルシード Acrylamide derivatives and polymers containing the derivatives
CA2359821A1 (en) 1999-02-04 2000-08-10 Mcgill University Platform for the differentiation of cells
AU780794B2 (en) 1999-02-10 2005-04-14 Es Cell International Pte Ltd Pancreatic progenitor cells, methods and uses related thereto
US6592623B1 (en) 1999-08-31 2003-07-15 Virginia Commonwealth University Intellectual Property Foundation Engineered muscle
KR100979664B1 (en) 1999-03-10 2010-09-02 유니버시티 오브 피츠버그 오브 더 커먼웰쓰 시스템 오브 하이어 에듀케이션 Adipose-derived stem cells and lattices
US6261600B1 (en) * 1999-04-30 2001-07-17 Drugtech Corporation Folic acid supplement
AU4860900A (en) 1999-06-02 2000-12-18 Lifebank Services, L.L.C. Methods of isolation, cryopreservation, and therapeutic use of human amniotic epithelial cells
US6329904B1 (en) * 1999-06-11 2001-12-11 Safety Through Cellular, Inc. Apparatus and method for providing weather and other alerts
US6261841B1 (en) * 1999-06-25 2001-07-17 The Board Of Trustees Of Northwestern University Compositions, kits, and methods for modulating survival and differentiation of multi-potential hematopoietic progenitor cells
JP5398941B2 (en) 1999-08-05 2014-01-29 エイビーティー ホールディング カンパニー Pluripotent adult stem cell and method for isolating the same
US6429013B1 (en) 1999-08-19 2002-08-06 Artecel Science, Inc. Use of adipose tissue-derived stromal cells for chondrocyte differentiation and cartilage repair
AU7378600A (en) 1999-09-14 2001-04-17 Children's Medical Center Corporation Methods for treating muscular dystrophy
US6331313B1 (en) 1999-10-22 2001-12-18 Oculex Pharmaceticals, Inc. Controlled-release biocompatible ocular drug delivery implant devices and methods
EP1099754A1 (en) 1999-11-10 2001-05-16 Universiteit Leiden Mesenchymal stem cells and/or progenitor cells, their isolation and use
US20020164307A1 (en) 1999-12-06 2002-11-07 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
WO2001053503A1 (en) 2000-01-18 2001-07-26 Cornell Research Foundation, Inc. Neural progenitor cells from hippocampal tissue and a method for isolating and purifying them
US6610535B1 (en) 2000-02-10 2003-08-26 Es Cell International Pte Ltd. Progenitor cells and methods and uses related thereto
JP2003521935A (en) 2000-02-11 2003-07-22 フイラデルフイア・ヘルス・アンド・エデユケーシヨン・コーポレーシヨン Differentiation of bone marrow cells into neuronal cells and uses therefor
ATE473751T1 (en) 2000-02-11 2010-07-15 Schepens Eye Res Inst ISOLATION AND TRANSPLANTATION OF RETINAL STEM CELLS
AU2001245550A1 (en) * 2000-03-09 2001-09-17 Lee Walters Applications of immune system tolerance to treatment of various diseases
EP2298858A1 (en) 2000-03-16 2011-03-23 Cellseed Inc. Bed material for cell culture, method for co-culture of cell and co-cultured cell sheet obtainable therefrom
US6436704B1 (en) * 2000-04-10 2002-08-20 Raven Biotechnologies, Inc. Human pancreatic epithelial progenitor cells and methods of isolation and use thereof
US20030212024A1 (en) 2000-05-12 2003-11-13 Keating Mark T Compositions and methods for cell dedifferentiation and tissue regeneration
US8273570B2 (en) 2000-05-16 2012-09-25 Riken Process of inducing differentiation of embryonic cell to cell expressing neural surface marker using OP9 or PA6 cells
US7049072B2 (en) 2000-06-05 2006-05-23 University Of South Florida Gene expression analysis of pluri-differentiated mesenchymal progenitor cells and methods for diagnosing a leukemic disease state
EP1312669B1 (en) 2000-07-21 2011-09-14 CellSeed Inc. Cultured epidermal cell sheet, laminated cultured skin sheet and process for producing the same
JP4679795B2 (en) * 2000-07-21 2011-04-27 株式会社セルシード Myocardial cell sheet, three-dimensional structure, myocardial tissue, and production method thereof
US6984522B2 (en) * 2000-08-03 2006-01-10 Regents Of The University Of Michigan Isolation and use of solid tumor stem cells
DE10038814A1 (en) * 2000-08-09 2002-02-21 Abb Research Ltd High voltage direct current transformer
WO2002026941A2 (en) 2000-09-29 2002-04-04 Kooy Derek V D Primitive neural stem cells and method for differentiation of stem cells to neural cells
US6639470B1 (en) 2000-10-06 2003-10-28 Skyworks Solutions, Inc. Constant current biasing circuit for linear power amplifiers
US7560280B2 (en) 2000-11-03 2009-07-14 Kourion Therapeutics Gmbh Human cord blood derived unrestricted somatic stem cells (USSC)
US20020168763A1 (en) 2000-11-30 2002-11-14 Yan Wen Liang Isolated homozygous stem cells, differentiated cells derived therefrom, and materials and methods for making and using same
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
CA2365376C (en) 2000-12-21 2006-03-28 Ethicon, Inc. Use of reinforced foam implants with enhanced integrity for soft tissue repair and regeneration
WO2002053193A2 (en) 2001-01-02 2002-07-11 The Charles Stark Draper Laboratory, Inc. Tissue engineering of three-dimensional vascularized using microfabricated polymer assembly technology
EP1366148A2 (en) 2001-01-24 2003-12-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE DEPARTMENT OF HEALTH &amp; HUMAN SERVICES Differentiation of stem cells to pancreatic endocrine cells
WO2002061053A1 (en) 2001-01-31 2002-08-08 The General Hospital Corporation Renal stem cells and uses thereof
EP1367961B1 (en) 2001-02-06 2008-12-31 Massachusetts Institute Of Technology Peptide scaffold encapsulation of tissue cells and uses thereof
US7449180B2 (en) 2001-02-06 2008-11-11 John Kisiday Macroscopic scaffold containing amphiphilic peptides encapsulating cells
KR101132545B1 (en) * 2001-02-14 2012-04-02 안트로제네시스 코포레이션 Post-partum mammalian placenta, its use and placental stem cells therefrom
EP1367899A4 (en) 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US20030008820A1 (en) * 2001-03-27 2003-01-09 Massachusetts Institute Of Technology Methods and products related to FGF dimerization
US7202080B2 (en) 2001-03-29 2007-04-10 Ixion Biotechnology, Inc. Method for transdifferentiation of non-pancreatic stem cells to the pancreatic differentiation pathway
US7838292B1 (en) 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
EP1379626A2 (en) 2001-04-19 2004-01-14 DeveloGen Aktiengesellschaft für entwicklungsbiologische Forschung A method for differentiating stem cells into insulin-producing cells
KR100762261B1 (en) 2001-04-27 2007-10-04 (주)바이오니아 Process for preparation of full-length cDNA and anchor and primer used for the same
US20030211605A1 (en) 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
WO2002098365A2 (en) 2001-06-07 2002-12-12 Skinmedica, Inc. Conditioned cell culture media and uses thereof
RU2183966C1 (en) 2001-07-06 2002-06-27 Общество с ограниченной ответственностью "Сибларекс" Composition of flavonoid complex "siblareks" for biologically active supplements of medicinal and chemical-pharmaceutical articles and method of preparing bioflavonoid complex "siblareks" for biologically active supplements of medicinal and chemical-pharmaceutical articles
US6402263B1 (en) 2001-07-24 2002-06-11 Robert Bosch Corporation Dual actuation master cylinder
US20030211603A1 (en) 2001-08-14 2003-11-13 Earp David J. Reprogramming cells for enhanced differentiation capacity using pluripotent stem cells
CN1195055C (en) * 2001-09-06 2005-03-30 周胜利 Method for establishing hematopoietic stem cells bank by extracting hematopoietic cells from placenta tissues
US20050064587A1 (en) 2001-09-07 2005-03-24 Lawrence Rosenberg Pancreatic small cells and uses thereof
US9969980B2 (en) 2001-09-21 2018-05-15 Garnet Biotherapeutics Cell populations which co-express CD49c and CD90
US7072332B2 (en) * 2001-09-27 2006-07-04 Samsung Electronics Co., Ltd. Soft switch using distributed firewalls for load sharing voice-over-IP traffic in an IP network
EP1298201A1 (en) 2001-09-27 2003-04-02 Cardion AG Process for the production of cells exhibiting an islet-beta-cell-like state
EP1446477A4 (en) 2001-09-28 2006-06-07 Es Cell Int Pte Ltd Methods of derivation and propagation of undifferentiated human embryonic stem (hes) cells on feeder-free matrices and human feeder layers
AU2002365162A1 (en) 2001-11-14 2003-07-09 Northwestern University Self-assembly and mineralization of peptide-amphiphile nanofibers
JP4330995B2 (en) 2001-11-15 2009-09-16 チルドレンズ メディカル センター コーポレーション Methods for isolating, proliferating, and differentiating fetal stem cells from chorionic villi, amniotic fluid, and placenta, and methods for their therapeutic use
CA2466478A1 (en) 2001-11-28 2003-06-05 Anges Mg, Inc. Gene therapy agents for neurodegenerative diseases
US6712850B2 (en) 2001-11-30 2004-03-30 Ethicon, Inc. Porous tissue scaffolds for the repair and regeneration of dermal tissue
JP2005511046A (en) 2001-12-04 2005-04-28 オーガノジェネシス インコーポレーテッド Cultured cells derived from islets
TW200301132A (en) * 2001-12-06 2003-07-01 Sankyo Co Pharmaceutical compositions containing cells derived from human caul
AU2002364558A1 (en) * 2001-12-11 2003-06-23 Cytograft Tissue Engineering, Inc. Tissue engineered cellular sheets, methods of making and use thereof
JP3934539B2 (en) 2001-12-12 2007-06-20 独立行政法人科学技術振興機構 Adult or postnatal tissue progenitor cells derived from placenta
US7101546B2 (en) 2001-12-21 2006-09-05 Amcyte, Inc. In situ maturation of cultured pancreatic stem cells having a specified, intermediate stage of development
CN1671835A (en) 2001-12-28 2005-09-21 塞拉提斯股份公司 A method for the establishment of a pluripotent human blastocyst-derived stem cell line
CA2473503C (en) 2002-01-14 2010-01-05 The Board Of Trustees Of The University Of Illinois Use of modified pyrimidine compounds to promote stem cell migration and proliferation
US20030158089A1 (en) 2002-01-24 2003-08-21 Xenoport, Inc. Administrative agents via the SMVT transporter
WO2003066832A2 (en) 2002-02-07 2003-08-14 The Research Foundation Of The State University Of New York Generation of new insulin cells from progenitor cells present in adult pancreatic islets
KR101176146B1 (en) * 2002-02-13 2012-08-22 안트로제네시스 코포레이션 Embryonic-like stem cells derived from post-partum mammalian placenta and uses and methods of treatment using said cells
WO2003070749A2 (en) 2002-02-15 2003-08-28 Northwestern University Self-assembly of peptide-amphiphile nanofibers under physiological conditions
US7576065B2 (en) 2002-02-15 2009-08-18 Cornell Research Foundation, Inc. Enhancing neurotrophin-induced neurogenesis by endogenous neural progenitor cells by concurrent overexpression of brain derived neurotrophic factor and an inhibitor of a pro-gliogenic bone morphogenetic protein
US20050118714A1 (en) 2002-02-19 2005-06-02 Chul-Won Ha Isolation and culture-expansion methods of mesenchymal stem/progenitor cells from umbilical cord blood and differentation method of umbilical cord blood-derived mesenchymal stem/progenitor cells into various mesenchymal tissues
WO2003072728A2 (en) 2002-02-22 2003-09-04 University Of Florida Cellular trans-differentiation
US7736892B2 (en) * 2002-02-25 2010-06-15 Kansas State University Research Foundation Cultures, products and methods using umbilical cord matrix cells
US20030161818A1 (en) * 2002-02-25 2003-08-28 Kansas State University Research Foundation Cultures, products and methods using stem cells
US7150990B2 (en) 2002-03-06 2006-12-19 Reprocell, Inc. Self-renewing pluripotent hepatic stem cells
JP2003259862A (en) 2002-03-12 2003-09-16 Fuji Photo Film Co Ltd Cell-culturing carrier
AU2003221173A1 (en) 2002-03-27 2003-10-08 Asahi Kasei Kabushiki Kaisha Placenta-origin mesenchymal cells and medicinal use thereof
US7498171B2 (en) 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
EP1538913A2 (en) 2002-04-12 2005-06-15 Celgene Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
JP4136434B2 (en) 2002-04-17 2008-08-20 進 清野 Induction of insulin-producing cells
US20030235563A1 (en) 2002-04-19 2003-12-25 Strom Stephen C. Placental derived stem cells and uses thereof
US20040161419A1 (en) 2002-04-19 2004-08-19 Strom Stephen C. Placental stem cells and uses thereof
JP2005526838A (en) 2002-04-25 2005-09-08 ウイスコンシン アラムナイ リサーチ フオンデーシヨン Use of GDNF-secreting human neural stem cells to treat Parkinson's disease and other neurodegenerative diseases
BR0311360A (en) 2002-05-28 2006-06-06 Becton Dickinson Co methods for in vitro expansion and transdifferentiation of human pancreatic acinar cells into insulin producing cells
WO2003104442A1 (en) 2002-06-07 2003-12-18 Es Cell International Pte Ltd Methods of regulating differentiation in stem cells
WO2004003561A1 (en) 2002-06-27 2004-01-08 Northwestern University Peptide rod amphiphiles and self-assembly of same
GB0216286D0 (en) 2002-07-15 2002-08-21 Univ Leeds Network
US20050249731A1 (en) 2002-07-16 2005-11-10 Hadi Aslan Methods of implating mesenchymal stem cells for tissue repair and formation
CA2493363A1 (en) 2002-07-29 2004-02-05 Asahi Kasei Kabushiki Kaisha Stem cells for treating pancreatic damage
CA2494040A1 (en) 2002-07-29 2004-02-05 Es Cell International Pte Ltd. Multi-step method for the differentiation of insulin positive, glucose
AU2003262628A1 (en) 2002-08-14 2004-03-03 University Of Florida Bone marrow cell differentiation
US7371576B2 (en) 2002-09-06 2008-05-13 Reneuron, Inc. CD56 positive human adult pancreatic endocrine progenitor cells
US6766863B2 (en) * 2002-09-20 2004-07-27 Hypro Corporation Fire fighting foam injection system with auto-start feature
US20040062753A1 (en) 2002-09-27 2004-04-01 Alireza Rezania Composite scaffolds seeded with mammalian cells
JP4262465B2 (en) * 2002-10-24 2009-05-13 富士フイルム株式会社 Cell culture method
AU2003287444A1 (en) 2002-10-31 2004-05-25 The General Hospital Corporation Repairing or replacing tissues or organs
AU2003296338A1 (en) 2002-12-05 2004-06-30 Case Western Reserve University Cell-based therapies for ischemia
JP4571387B2 (en) 2003-02-07 2010-10-27 宣男 櫻川 Human amnion-derived side population cells and uses thereof
AU2004210853A1 (en) 2003-02-11 2004-08-26 Northwestern University Methods and materials for nanocrystalline surface coatings and attachment of peptide amphiphile nanofibers thereon
CA2515469C (en) 2003-02-11 2013-12-17 John E. Davies Progenitor cells from wharton's jelly of human umbilical cord
US7521481B2 (en) 2003-02-27 2009-04-21 Mclaurin Joanne Methods of preventing, treating and diagnosing disorders of protein aggregation
US20040224401A1 (en) 2003-03-28 2004-11-11 Ludwig Tenneille E. Physiochemical culture conditions for embryonic stem cells
EP1636351A4 (en) 2003-05-21 2007-04-04 Gen Hospital Corp Microfabricated compositions and processes for engineering tissues containing multiple cell types
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US9572840B2 (en) * 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
CA2536909A1 (en) 2003-08-29 2005-03-10 Regents Of The University Of Minnesota Kidney derived stem cells and methods for their isolation, differentiation and use
US20050089513A1 (en) * 2003-10-28 2005-04-28 Norio Sakuragawa Side population cells originated from human amnion and their uses
CA2548305C (en) 2003-12-23 2014-07-15 Fmc Biopolymer As Use of alginate matrices to control cell growth
TWI276685B (en) * 2003-12-30 2007-03-21 Ind Tech Res Inst Conditional medium for culturing Schwann cells
US7534606B2 (en) 2004-01-12 2009-05-19 National Health Research Institutes Placental stem cell and methods thereof
DE102004043256B4 (en) 2004-09-07 2013-09-19 Rheinische Friedrich-Wilhelms-Universität Bonn Scalable process for culturing undifferentiated stem cells in suspension
JP2008518597A (en) 2004-10-29 2008-06-05 セントカー・インコーポレーテツド Chemically defined medium composition
US7655678B2 (en) * 2004-11-30 2010-02-02 Council of Scientfic & Industrial Research Pharmaceutical composition for the management of tumors
WO2006101548A2 (en) 2004-12-21 2006-09-28 Ethicon, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making, culturing, and using the same
EP1831355A2 (en) 2004-12-23 2007-09-12 Ethicon, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
EP1838842A2 (en) 2004-12-23 2007-10-03 Ethicon, Incorporated Treatment of osteochondral diseases using postpartum-derived cells and products thereof
PL1831356T3 (en) * 2004-12-23 2017-07-31 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US7741311B2 (en) 2005-01-03 2010-06-22 Shaker Mousa Composition and method for treating occlusive vascular diseases, nerve regeneration, and wound healing
AU2006208944A1 (en) 2005-01-28 2006-08-03 Imperial College Innovations Limited Methods for embryonic stem cell culture
JP5017253B2 (en) 2005-03-31 2012-09-05 ステムニオン,インコーポレイテッド Amnion-derived cell composition, preparation method and use thereof
US7923007B2 (en) 2005-08-08 2011-04-12 Academia Sinica Brain tissue damage therapies
WO2007070870A1 (en) 2005-12-16 2007-06-21 Ethicon, Inc. Compositions and methods for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
WO2007081841A2 (en) 2006-01-06 2007-07-19 Stratagene California Reaction buffer composition for nucleic acid replication with packed dna polymerases
PT2366775E (en) 2006-03-23 2015-07-20 Pluristem Ltd Methods for cell expansion and uses of cells and conditioned media produced thereby for therapy
WO2007146106A2 (en) * 2006-06-05 2007-12-21 Cryo- Cell International, Inc. Procurement, isolation and cryopreservation of maternal placental cells
NZ597779A (en) 2007-02-12 2013-07-26 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
US20080305148A1 (en) 2007-03-19 2008-12-11 National Yang Ming University Treatment of spinal injuries using human umbilical mesenchymal stem cells
UA99152C2 (en) 2007-10-05 2012-07-25 Этикон, Инкорпорейтед Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
US20090123620A1 (en) * 2007-11-14 2009-05-14 Hiti Thomas R Automated Application of an Antimycotic Composition to Sliced Foodstuffs and an Antimycotic Application Apparatus
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
US20090186358A1 (en) 2007-12-21 2009-07-23 Wyeth Pathway Analysis of Cell Culture Phenotypes and Uses Thereof
WO2009085860A1 (en) 2007-12-27 2009-07-09 Ethicon, Incorporated Treatment of intervertebral disc degeneration using human umbilical cord tissue-derived cells
TWI363852B (en) * 2008-03-28 2012-05-11 Ind Tech Res Inst Stacked light emitting device
ES2520393T3 (en) 2008-12-19 2014-11-11 DePuy Synthes Products, LLC Cells derived from umbilical cord tissue to treat neuropathic pain and spasticity
CA2747794C (en) 2008-12-19 2018-10-30 Advanced Technologies And Regenerative Medicine, Llc Treatment of lung and pulmonary diseases and disorders
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
JP6095893B2 (en) 2008-12-19 2017-03-15 デピュイ・シンセス・プロダクツ・インコーポレイテッド Regeneration and repair of nerve tissue after injury
BRPI1013409A2 (en) 2009-03-26 2018-01-16 Advanced Tech And Regenerative Medicine Llc human umbilical cord tissue cells as therapy for alzheimer's disease
CN103329308B (en) * 2011-01-19 2016-09-14 纳幕尔杜邦公司 There is the lithium battery separator of closing function

Patent Citations (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3665061A (en) * 1969-07-16 1972-05-23 United States Banknote Corp Process for producing collagen sponges
US3930954A (en) * 1973-05-31 1976-01-06 Godo Shusei Kabushiki Kaisha Neutral protease useful for animal tissue and cell culture
US6022743A (en) * 1986-04-18 2000-02-08 Advanced Tissue Sciences, Inc. Three-dimensional culture of pancreatic parenchymal cells cultured living stromal tissue prepared in vitro
US5902741A (en) * 1986-04-18 1999-05-11 Advanced Tissue Sciences, Inc. Three-dimensional cartilage cultures
US5443950A (en) * 1986-04-18 1995-08-22 Advanced Tissue Sciences, Inc. Three-dimensional cell and tissue culture system
US4925667A (en) * 1986-05-27 1990-05-15 Qmax Technology Group, Inc. Substrate with particulate cosmetic
US5004681A (en) * 1987-11-12 1991-04-02 Biocyte Corporation Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5004681B1 (en) * 1987-11-12 2000-04-11 Biocyte Corp Preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US5192553A (en) * 1987-11-12 1993-03-09 Biocyte Corporation Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood and methods of therapeutic use
US20030032178A1 (en) * 1988-08-04 2003-02-13 Williams Robert Lindsay In vitro propagation of embryonic stem cells
US5437994A (en) * 1989-06-15 1995-08-01 Regents Of The University Of Michigan Method for the ex vivo replication of stem cells, for the optimization of hematopoietic progenitor cell cultures, and for increasing the metabolism, GM-CSF secretion and/or IL-6 secretion of human stromal cells
US5336616A (en) * 1990-09-12 1994-08-09 Lifecell Corporation Method for processing and preserving collagen-based tissues for transplantation
US5342761A (en) * 1990-10-01 1994-08-30 Research Development Foundation Oncofetal gene, gene product and uses therefor
US5486359A (en) * 1990-11-16 1996-01-23 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US5286632A (en) * 1991-01-09 1994-02-15 Jones Douglas H Method for in vivo recombination and mutagenesis
US20030049837A1 (en) * 1991-07-08 2003-03-13 Samuel Weiss In vitro and in vivo proliferation and use of multipotent neural stem cells and their progeny
US5320962A (en) * 1992-07-22 1994-06-14 Duke University DNA encoding the human A1 adenosine receptor
US5707643A (en) * 1993-02-26 1998-01-13 Santen Pharmaceutical Co., Ltd. Biodegradable scleral plug
US6686198B1 (en) * 1993-10-14 2004-02-03 President And Fellows Of Harvard College Method of inducing and maintaining neuronal cells
US6703017B1 (en) * 1994-04-28 2004-03-09 Ixion Biotechnology, Inc. Reversal of insulin-dependent diabetes by islet-producing stem cells, islet progenitor cells and islet-like structures
US5935849A (en) * 1994-07-20 1999-08-10 Cytotherapeutics, Inc. Methods and compositions of growth control for cells encapsulated within bioartificial organs
US6251090B1 (en) * 1994-12-12 2001-06-26 Robert Logan Avery Intravitreal medicine delivery
US6200806B1 (en) * 1995-01-20 2001-03-13 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5906934A (en) * 1995-03-14 1999-05-25 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US6214369B1 (en) * 1995-03-14 2001-04-10 Morphogen Pharmaceuticals, Inc. Mesenchymal stem cells for cartilage repair
US5718922A (en) * 1995-05-31 1998-02-17 Schepens Eye Research Institute, Inc. Intravitreal microsphere drug delivery and method of preparation
US5869079A (en) * 1995-06-02 1999-02-09 Oculex Pharmaceuticals, Inc. Formulation for controlled release of drugs by combining hydrophilic and hydrophobic agents
US5736516A (en) * 1995-11-29 1998-04-07 Amgen Inc. Methods for treating photoreceptors using glial cell line-derived neurotrophic factor (GDNF) protein protein
US6200606B1 (en) * 1996-01-16 2001-03-13 Depuy Orthopaedics, Inc. Isolation of precursor cells from hematopoietic and nonhematopoietic tissues and their use in vivo bone and cartilage regeneration
US6358737B1 (en) * 1996-07-31 2002-03-19 Board Of Regents, The University Of Texas System Osteocyte cell lines
US5919702A (en) * 1996-10-23 1999-07-06 Advanced Tissue Science, Inc. Production of cartilage tissue using cells isolated from Wharton's jelly
US6221904B1 (en) * 1997-05-21 2001-04-24 Sloan-Kettering Institute For Cancer Research Method for increasing the concentration of ascorbic acid in brain tissues of a subject
US5902598A (en) * 1997-08-28 1999-05-11 Control Delivery Systems, Inc. Sustained release drug delivery devices
US6391297B1 (en) * 1997-12-02 2002-05-21 Artecel Sciences, Inc. Differentiation of adipose stromal cells into osteoblasts and uses thereof
US6059968A (en) * 1998-01-20 2000-05-09 Baxter International Inc. Systems for processing and storing placenta/umbilical cord blood
US6179872B1 (en) * 1998-03-17 2001-01-30 Tissue Engineering Biopolymer matt for use in tissue repair and reconstruction
US6528245B2 (en) * 1998-05-07 2003-03-04 University Of South Florida Bone marrow cells as a source of neurons for brain and spinal cord repair
US6387367B1 (en) * 1998-05-29 2002-05-14 Osiris Therapeutics, Inc. Human mesenchymal stem cells
US20040037818A1 (en) * 1998-07-30 2004-02-26 Brand Stephen J. Treatment for diabetes
US6680198B1 (en) * 1998-08-14 2004-01-20 The Children's Medical Center Corporation Engraftable human neural stem cells
US20040005704A1 (en) * 1998-11-18 2004-01-08 California Institute Of Technology Low oxygen culturing of central nervous system progenitor cells
US6335239B1 (en) * 1998-12-04 2002-01-01 International Business Machines Corporation Manufacturing a DRAM cell having an annular signal transfer region
US20030007954A1 (en) * 1999-04-12 2003-01-09 Gail K. Naughton Methods for using a three-dimensional stromal tissue to promote angiogenesis
US20020022676A1 (en) * 1999-04-16 2002-02-21 Shulin He Poly(Propylene Fumarate) cross linked with Poly(Ethylene Glycol)
US20020062151A1 (en) * 1999-05-14 2002-05-23 Gregory Altman Bioengineered anterior cruciate ligament
US6372494B1 (en) * 1999-05-14 2002-04-16 Advanced Tissue Sciences, Inc. Methods of making conditioned cell culture medium compositions
US6355699B1 (en) * 1999-06-30 2002-03-12 Ethicon, Inc. Process for manufacturing biomedical foams
US6534084B1 (en) * 1999-06-30 2003-03-18 Ethicon, Inc. Porous tissue scaffoldings for the repair or regeneration of tissue
US6555374B1 (en) * 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
US20040033597A1 (en) * 1999-08-29 2004-02-19 Jean Toma Multipotent neural stemcells from peripheral tissues and uses thereof
US20040014206A1 (en) * 1999-10-28 2004-01-22 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US20030031657A1 (en) * 1999-12-06 2003-02-13 Massachusetts General Hospital Stem cells and their use in transplantation
US20030082155A1 (en) * 1999-12-06 2003-05-01 Habener Joel F. Stem cells of the islets of langerhans and their use in treating diabetes mellitus
US20030003574A1 (en) * 2000-01-24 2003-01-02 Jean Toma Multipotent stem cells from peripheral tissues and uses thereof
US20020028510A1 (en) * 2000-03-09 2002-03-07 Paul Sanberg Human cord blood as a source of neural tissue for repair of the brain and spinal cord
US6673606B1 (en) * 2000-04-12 2004-01-06 The Children's Hospital Of Philadelphia Therapeutic uses for mesenchymal stromal cells
US6375972B1 (en) * 2000-04-26 2002-04-23 Control Delivery Systems, Inc. Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
US20020081725A1 (en) * 2000-06-30 2002-06-27 Wen-Ghih Tsang Culturing pancreatic stem cells having a specified, intermediate stage of development
US20020098584A1 (en) * 2000-11-06 2002-07-25 Palmer Theo D. Postmortem stem cells
US20030032179A1 (en) * 2000-12-06 2003-02-13 Hariri Robert J. Post-partum mammalian placenta, its use and placental stem cells therefrom
US20040048372A1 (en) * 2000-12-06 2004-03-11 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US6599323B2 (en) * 2000-12-21 2003-07-29 Ethicon, Inc. Reinforced tissue implants and methods of manufacture and use
US20030022369A1 (en) * 2001-05-18 2003-01-30 Helen Fillmore Differentiation of specialized dermal and epidermal cells into neuronal cells
US20030032183A1 (en) * 2001-05-25 2003-02-13 Sheridan Steven D. Stem cell differentiation
US20030118566A1 (en) * 2001-08-08 2003-06-26 Toomas Neuman Compositions and methods for isolation, propagation, and differentiation of human stem cells and uses thereof
US20030059939A1 (en) * 2001-08-27 2003-03-27 Raymond Page Trans-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20030104997A1 (en) * 2001-09-05 2003-06-05 Black Ira B. Multi-lineage directed induction of bone marrow stromal cell differentiation
US20030138951A1 (en) * 2001-10-18 2003-07-24 Li Yin Conversion of liver stem and progenitor cells to pancreatic functional cells
US20030082160A1 (en) * 2001-10-25 2003-05-01 Yu John S. Differentiation of whole bone marrow
US20030124721A1 (en) * 2001-11-09 2003-07-03 Bentley Cheatham Endocrine pancreas differentiation of adipose tissue-derived stromal cells and uses thereof
US20030096409A1 (en) * 2001-11-22 2003-05-22 Shigeru Yasumoto Cultured skin and method of manufacturing the same
US20030109036A1 (en) * 2001-12-06 2003-06-12 The Regents Of The University Of California Method for differentiating islet precursor cells into beta cells
US20030138948A1 (en) * 2001-12-07 2003-07-24 Fisk Gregory J. Islet cells from human embryonic stem cells
US20030113910A1 (en) * 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
US20050074435A1 (en) * 2001-12-21 2005-04-07 Robert Casper Cellular compositions and methods of making and using them
US20040072344A1 (en) * 2002-01-25 2004-04-15 Kazutomo Inoue Method for inducing differentiation of embryonic stem cells into functioning cells
US20040029269A1 (en) * 2002-05-07 2004-02-12 Goldman Steven A Promoter-based isolation, purification, expansion, and transplantation of neuronal progenitor cells, oligodendrocyte progenitor cells, or neural stem cells from a population of embryonic stem cells
US20040014662A1 (en) * 2002-05-08 2004-01-22 Per Lindquist Modulation of neural stem cells and neural progenitor cells
US20040028660A1 (en) * 2002-05-30 2004-02-12 Anthrogenesis Corporation Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US20040014211A1 (en) * 2002-06-11 2004-01-22 Ogle Roy C. Meningeal-derived stem cells
US20040009593A1 (en) * 2002-07-11 2004-01-15 Keirstead Hans S. Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US20040014210A1 (en) * 2002-07-16 2004-01-22 Jessell Thomas M. Methods for inducing differentiation of embryonic stem cells and uses thereof
US20060128014A1 (en) * 2002-08-26 2006-06-15 Johan Haggblad Compositions and methods for culturing stem cells
US20040063202A1 (en) * 2002-08-28 2004-04-01 Petersen Bryon E. Neurogenesis from hepatic stem cells
US20040058412A1 (en) * 2002-09-20 2004-03-25 Neuronyx, Inc. Cell populations which co-express CD49c and CD90
US7524489B2 (en) * 2003-06-27 2009-04-28 Ethicon Incorporated Regeneration and repair of neural tissue using postpartum-derived cells
US20130022585A1 (en) * 2003-06-27 2013-01-24 Advanced Technologies And Regenerative Medicine, Llc Regeneration and repair of neural tissue using postpartum-derived cells
US8658152B2 (en) * 2003-06-27 2014-02-25 DePuy Synthes Products, LLC Regeneration and repair of neural tissue using postpartum-derived cells
US20140045263A1 (en) * 2003-06-27 2014-02-13 DePuy Synthes Products, LLC. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US7510873B2 (en) * 2003-06-27 2009-03-31 Ethicon, Incorporated Postpartum cells isolated from umbilical cord tissue, and methods of making and using the same
US20050037491A1 (en) * 2003-06-27 2005-02-17 Sanjay Mistry Repair and regeneration of ocular tissue using postpartum-derived cells
US7560276B2 (en) * 2003-06-27 2009-07-14 Ethicon, Incorporated Soft tissue repair and regeneration using postpartum-derived cells
US7875272B2 (en) * 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US20080145934A1 (en) * 2004-09-28 2008-06-19 Ian Ross Harris Tissue-engineering scaffolds containing self-assembled-peptide hydrogels
US20060166361A1 (en) * 2004-12-21 2006-07-27 Agnieszka Seyda Postpartum cells derived from placental tissue, and methods of making, culturing, and using the same
US20060153815A1 (en) * 2004-12-21 2006-07-13 Agnieszka Seyda Tissue engineering devices for the repair and regeneration of tissue
US7875273B2 (en) * 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US20070141700A1 (en) * 2005-12-19 2007-06-21 Ethicon, Incorporated In vitro expansion of postpartum-derived cells in roller bottles
US20070160588A1 (en) * 2005-12-28 2007-07-12 Ethicon, Incorporated Treatment Of Peripheral Vascular Disease Using Postpartum-Derived Cells
US20080112939A1 (en) * 2006-10-12 2008-05-15 Ethicon, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Solomon An in vitro examination of an extracellular matrix scaffold for use in wound healing. Int. J. Exp. Path (2002) 83, 209-216 *

Cited By (159)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8057788B2 (en) 2000-12-06 2011-11-15 Anthrogenesis Corporation Placental stem cell populations
US9149569B2 (en) 2000-12-06 2015-10-06 Anthrogenesis Corporation Treatment of diseases or disorders using placental stem cells
US20050019908A1 (en) * 2000-12-06 2005-01-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US7976836B2 (en) 2000-12-06 2011-07-12 Anthrogenesis Corporation Treatment of stroke using placental stem cells
US8580563B2 (en) 2000-12-06 2013-11-12 Anthrogenesis Corporation Placental stem cells
US8293223B2 (en) 2000-12-06 2012-10-23 Anthrogenesis Corporation Treatment of organ injuries and burns using placental stem cells
US8545833B2 (en) 2000-12-06 2013-10-01 Anthrogenesis Corporation Treatment of radiation injury using placental stem cells
US20020160510A1 (en) * 2001-02-14 2002-10-31 Hariri Robert J. Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US20080131966A1 (en) * 2001-02-14 2008-06-05 Hariri Robert J Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US9139813B2 (en) 2001-02-14 2015-09-22 Anthrogenesis Corporation Renovation and repopulation of decellularized tissues and cadaveric organs by stem cells
US8435788B2 (en) 2001-02-14 2013-05-07 Anthrogenesis Corporation Tissue matrices comprising placental stem cells
US7914779B2 (en) 2001-02-14 2011-03-29 Anthrogenesis Corporation Tissue matrices comprising placental stem cells, and methods of making the same
US20100196910A1 (en) * 2001-07-20 2010-08-05 Technion Research & Development Foundation Ltd. Methods of generating human cardiac cells and tissues and uses thereof
US8753883B2 (en) 2002-02-13 2014-06-17 Anthrogenesis Corporation Treatment of psoriasis using placental stem cells
US8057789B2 (en) 2002-02-13 2011-11-15 Anthrogenesis Corporation Placental stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
US7700090B2 (en) 2002-02-13 2010-04-20 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US9234172B2 (en) 2003-06-27 2016-01-12 DePuy Synthes Products, Inc. Repair and regeneration of ocular tissue using postpartum-derived cells
US10039793B2 (en) 2003-06-27 2018-08-07 DePuy Synthes Products, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US8815587B2 (en) 2003-06-27 2014-08-26 DePuy Synthes Products, LLC Postpartum cells derived from umbilical tissue and methods of making and using the same
US11179422B2 (en) 2003-06-27 2021-11-23 DePuy Synthes Products, Inc. Method of differentiating umbilical cord tissue into a chondrogenic phenotype
US8518390B2 (en) 2003-06-27 2013-08-27 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders via intranasal administration of umbilical cord-derived cells
US10758576B2 (en) 2003-06-27 2020-09-01 DePuy Synthes Products, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US9592258B2 (en) 2003-06-27 2017-03-14 DePuy Synthes Products, Inc. Treatment of neurological injury by administration of human umbilical cord tissue-derived cells
US7875272B2 (en) 2003-06-27 2011-01-25 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8491883B2 (en) 2003-06-27 2013-07-23 Advanced Technologies And Regenerative Medicine, Llc Treatment of amyotrophic lateral sclerosis using umbilical derived cells
US11191789B2 (en) 2003-06-27 2021-12-07 DePuy Synthes Products, Inc. Cartilage and bone repair and regeneration using postpartum-derived cells
US20050019865A1 (en) * 2003-06-27 2005-01-27 Kihm Anthony J. Cartilage and bone repair and regeneration using postpartum-derived cells
US8790637B2 (en) 2003-06-27 2014-07-29 DePuy Synthes Products, LLC Repair and regeneration of ocular tissue using postpartum-derived cells
US10744164B2 (en) 2003-06-27 2020-08-18 DePuy Synthes Products, Inc. Repair and regeneration of ocular tissue using postpartum-derived cells
US9579351B2 (en) 2003-06-27 2017-02-28 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US9572840B2 (en) 2003-06-27 2017-02-21 DePuy Synthes Products, Inc. Regeneration and repair of neural tissue using postpartum-derived cells
US9717763B2 (en) 2003-06-27 2017-08-01 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US9504719B2 (en) 2003-06-27 2016-11-29 DePuy Synthes Products, Inc. Soft tissue repair and regeneration using postpartum-derived cells and cell products
US11000554B2 (en) 2003-06-27 2021-05-11 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US8703121B2 (en) 2003-06-27 2014-04-22 DePuy Synthes Products, LLC Postpartum-derived cells for use in treatment of disease of the heart and circulatory system
US8658152B2 (en) 2003-06-27 2014-02-25 DePuy Synthes Products, LLC Regeneration and repair of neural tissue using postpartum-derived cells
US10195233B2 (en) 2003-06-27 2019-02-05 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US10500234B2 (en) 2003-06-27 2019-12-10 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US9498501B2 (en) 2003-06-27 2016-11-22 DePuy Synthes Products, Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US20060233765A1 (en) * 2003-06-27 2006-10-19 Ethicon, Incorporated Treatment of stroke and other acute neuraldegenerative disorders using postpartum derived cells
US8277796B2 (en) 2003-06-27 2012-10-02 Advanced Technologies And Regenerative Medicine, Llc Regeneration and repair of neural tissue using postpartum-derived cells
US10220059B2 (en) 2003-06-27 2019-03-05 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US20060223177A1 (en) * 2003-06-27 2006-10-05 Ethicon Inc. Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8318483B2 (en) 2003-06-27 2012-11-27 Advanced Technologies And Regenerative Medicine, Llc Postpartum cells derived from umbilical cord tissue, and methods of making and using the same
US8361459B2 (en) 2003-06-27 2013-01-29 Advanced Technologies And Regenerative Medicine, Llc Treatment of stroke and other acute neural degenerative disorders using postpartum-derived cells
US10383898B2 (en) 2003-06-27 2019-08-20 DePuy Synthes Products, Inc. Postpartum cells derived from placental tissue, and methods of making and using the same
US20060069008A1 (en) * 2004-09-28 2006-03-30 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US8846394B2 (en) * 2004-09-28 2014-09-30 Ethicon, Inc. Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US20090149371A1 (en) * 2004-09-28 2009-06-11 Sanjay Mistry Treatment of neurological deficits in the striatum or substanta nigra pars compacta
US7875273B2 (en) 2004-12-23 2011-01-25 Ethicon, Incorporated Treatment of Parkinson's disease and related disorders using postpartum derived cells
US20090010899A1 (en) * 2005-03-31 2009-01-08 Palladino Linda O Methods related to surgery
US8685390B2 (en) 2005-03-31 2014-04-01 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US8153430B2 (en) 2005-03-31 2012-04-10 Stemnion, Inc. Methods related to surgery
US20060222634A1 (en) * 2005-03-31 2006-10-05 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US8877181B2 (en) 2005-03-31 2014-11-04 Stemnion, Inc. Amnion-derived cells, methods of making and uses thereof
US20090075381A1 (en) * 2005-03-31 2009-03-19 Clarke Diana L Amnion-derived cell compositions, methods of making and uses thereof
US8278095B2 (en) 2005-03-31 2012-10-02 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US8709402B2 (en) 2005-03-31 2014-04-29 Stemnion, Inc. Amnion-derived cells, methods of making and uses thereof
US8741646B2 (en) 2005-03-31 2014-06-03 Stemnion, Inc. Amnion-derived cell compositions, methods of making and uses thereof
US8895256B2 (en) 2005-10-13 2014-11-25 Anthrogenesis Corporation Immunomodulation using placental stem cells
US9539288B2 (en) 2005-10-13 2017-01-10 Anthrogenesis Corporation Immunomodulation using placental stem cells
US7682803B2 (en) 2005-10-13 2010-03-23 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8216566B2 (en) 2005-10-13 2012-07-10 Anthrogenesis Corporation Treatment of multiple sclerosis using placental stem cells
US9175261B2 (en) 2005-12-16 2015-11-03 DePuy Synthes Products, Inc. Human umbilical cord tissue cells for inhibiting adverse immune response in histocompatibility-mismatched transplantation
US8741638B2 (en) 2005-12-19 2014-06-03 DePuy Synthes Products, LLC In vitro expansion of postpartum-derived cells in roller bottles
US9125906B2 (en) 2005-12-28 2015-09-08 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US9585918B2 (en) 2005-12-28 2017-03-07 DePuy Synthes Products, Inc. Treatment of peripheral vascular disease using umbilical cord tissue-derived cells
US8691217B2 (en) 2005-12-29 2014-04-08 Anthrogenesis Corporation Placental stem cell populations
US8591883B2 (en) 2005-12-29 2013-11-26 Anthrogenesis Corporation Placental stem cell populations
US8202703B2 (en) 2005-12-29 2012-06-19 Anthrogenesis Corporation Placental stem cell populations
US9078898B2 (en) 2005-12-29 2015-07-14 Anthrogenesis Corporation Placental stem cell populations
US8455250B2 (en) 2005-12-29 2013-06-04 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
US10383897B2 (en) 2005-12-29 2019-08-20 Celularity, Inc. Placental stem cell populations
US7993918B2 (en) 2006-08-04 2011-08-09 Anthrogenesis Corporation Tumor suppression using placental stem cells
US10413571B2 (en) 2006-10-12 2019-09-17 DePuy Synthes Products, Inc. Kidney-derived cells and methods of use in tissue repair and regeneration
US10105399B2 (en) 2006-10-23 2018-10-23 Celularity, Inc. Methods and compositions for treatment of bone defects with placental cell populations
US8562972B2 (en) 2006-10-23 2013-10-22 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US9339520B2 (en) 2006-10-23 2016-05-17 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US20080166328A1 (en) * 2006-11-13 2008-07-10 Ethicon, Inc. In vitro expansion of postpartum-derived cells using microcarriers
US9102915B2 (en) 2006-11-13 2015-08-11 DePuy Synthes Products, Inc. In vitro expansion of postpartum-derived cells using microcarriers
WO2008083223A1 (en) * 2006-12-28 2008-07-10 Isolagen Technologies, Inc. Methods for culturing dermal cells for treatment of skin injuries such as burns
US20100303770A1 (en) * 2006-12-28 2010-12-02 John Maslowski Methods for culturing dermal cells for treatment of skin injuries such as burns
US9173908B2 (en) 2007-01-17 2015-11-03 Stemnion, Inc. Methods for modulating immune responses
US8221741B2 (en) 2007-01-17 2012-07-17 Marshall Vivienne S Methods for modulating inflammatory and/or immune responses
US8506949B2 (en) 2007-01-17 2013-08-13 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
US9138443B2 (en) 2007-01-17 2015-09-22 Stemnion, Inc. Methods for modulating immune responses
US8916146B2 (en) 2007-02-12 2014-12-23 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US8460650B2 (en) 2007-02-12 2013-06-11 Anthrogenesis Corporation Treatment of inflammatory diseases using placental stem cells
US20080286267A1 (en) * 2007-04-13 2008-11-20 Sing George L Methods for treating nervous system inquiry and disease
US8197804B2 (en) 2007-04-13 2012-06-12 Stemnion, Inc. Methods for treating nervous system injury and disease
US10286015B2 (en) 2007-04-13 2019-05-14 Noveome Biotherapeutics, Inc. Methods for treating traumatic brain injury with amnion-derived cellular cytokine solution (ACCS) or amnion-derived multipotent progenitor (AMP) cells
US20080286324A1 (en) * 2007-05-14 2008-11-20 Cardiac Pacemakers, Inc. Media and devices for cold storage of therapeutic cells
US8114668B2 (en) 2007-05-14 2012-02-14 Cardiac Pacemakers, Inc. Composition for cold storage of stem cells
US8273526B2 (en) 2007-06-18 2012-09-25 Children's Hospital & Research Center At Oakland Method of isolating stem and progenitor cells from placenta
US9216200B2 (en) 2007-09-28 2015-12-22 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US8263065B2 (en) 2007-09-28 2012-09-11 Anthrogenesis Corporation Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US8034329B2 (en) 2007-10-05 2011-10-11 Advanced Technologies And Regenerative Medicine, Llc Repair and regeneration of renal tissue using human umbilical cord tissue-derived cells
WO2009052459A1 (en) * 2007-10-19 2009-04-23 University Of Medicine And Dentistry Of New Jersey A method of using an extracellular matrix to enhance cell transplant survival and differentiation
US9598672B2 (en) 2007-10-19 2017-03-21 Rutgers, The State University Of New Jersey Production of extracellular matrix, conditioned media and uses thereof
US20100297234A1 (en) * 2007-10-19 2010-11-25 Ilene Sugino Method of using an extracellular matrix to enhance cell transplant survival and differentiation
US8574897B2 (en) 2007-12-20 2013-11-05 DePuy Synthes Products, LLC Methods for sterilizing materials containing biologically active agents
US8236538B2 (en) 2007-12-20 2012-08-07 Advanced Technologies And Regenerative Medicine, Llc Methods for sterilizing materials containing biologically active agents
EP2103935A1 (en) 2008-02-28 2009-09-23 Centocor Ortho Biotech, Inc. Multiple administrations of umbilicus derived cells
US20090220995A1 (en) * 2008-02-28 2009-09-03 Sachs David H Multiple administrations of umbilicus derived cells
US9480549B2 (en) 2008-04-25 2016-11-01 Allosource Multi-layer tissue patches
US9358320B2 (en) 2008-04-25 2016-06-07 Allosource Multi-layer tissue patches
US9616152B2 (en) 2008-04-25 2017-04-11 Allosource Multi-layer tissue systems and methods
US10104880B2 (en) 2008-08-20 2018-10-23 Celularity, Inc. Cell composition and methods of making the same
US8828376B2 (en) 2008-08-20 2014-09-09 Anthrogenesis Corporation Treatment of stroke using isolated placental cells
US8728805B2 (en) 2008-08-22 2014-05-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
US8367409B2 (en) 2008-11-19 2013-02-05 Anthrogenesis Corporation Amnion derived adherent cells
US9198938B2 (en) 2008-11-19 2015-12-01 Antrhogenesis Corporation Amnion derived adherent cells
US10179900B2 (en) 2008-12-19 2019-01-15 DePuy Synthes Products, Inc. Conditioned media and methods of making a conditioned media
US10557116B2 (en) 2008-12-19 2020-02-11 DePuy Synthes Products, Inc. Treatment of lung and pulmonary diseases and disorders
US8771677B2 (en) 2008-12-29 2014-07-08 Vladimir B Serikov Colony-forming unit cell of human chorion and method to obtain and use thereof
US9943552B2 (en) 2009-03-26 2018-04-17 DePuy Synthes Products, Inc. hUTC as therapy for Alzheimer's disease
US8722034B2 (en) 2009-03-26 2014-05-13 Depuy Synthes Products Llc hUTC as therapy for Alzheimer's disease
US8647617B2 (en) 2009-07-13 2014-02-11 Stemnion, Inc. Methods for modulating inflammatory and/or immune responses
US9121007B2 (en) 2010-01-26 2015-09-01 Anthrogenesis Corporatin Treatment of bone-related cancers using placental stem cells
US9254302B2 (en) 2010-04-07 2016-02-09 Anthrogenesis Corporation Angiogenesis using placental stem cells
US8562973B2 (en) 2010-04-08 2013-10-22 Anthrogenesis Corporation Treatment of sarcoidosis using placental stem cells
US8808687B2 (en) 2010-07-12 2014-08-19 Mark Humayun Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
US11154639B2 (en) 2010-07-12 2021-10-26 University Of Southern California Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
US10188769B2 (en) 2010-07-12 2019-01-29 University Of Southern California Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
US9464274B2 (en) 2010-07-13 2016-10-11 Anthrogenesis Corporation Methods of generating natural killer cells
US8926964B2 (en) 2010-07-13 2015-01-06 Anthrogenesis Corporation Methods of generating natural killer cells
US8574899B2 (en) 2010-12-22 2013-11-05 Vladimir B Serikov Methods for augmentation collection of placental hematopoietic stem cells and uses thereof
US8969315B2 (en) 2010-12-31 2015-03-03 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory RNA molecules
US10478206B2 (en) 2011-04-29 2019-11-19 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US11090339B2 (en) 2011-06-01 2021-08-17 Celularity Inc. Treatment of pain using placental stem cells
US9040035B2 (en) 2011-06-01 2015-05-26 Anthrogenesis Corporation Treatment of pain using placental stem cells
US9925221B2 (en) 2011-09-09 2018-03-27 Celularity, Inc. Treatment of amyotrophic lateral sclerosis using placental stem cells
US11318225B2 (en) 2011-12-05 2022-05-03 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
US9642940B2 (en) 2011-12-05 2017-05-09 California Institute Of Technology 3-dimensional parylene scaffold cage
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
US9744266B2 (en) 2011-12-19 2017-08-29 Allosource Flowable matrix compositions and methods
US9801976B2 (en) 2011-12-19 2017-10-31 Allosource Flowable matrix compositions and methods
US9801975B2 (en) 2011-12-19 2017-10-31 Allosource Flowable matrix compositions and methods
US10724105B2 (en) 2011-12-23 2020-07-28 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue-derived cells
US9611513B2 (en) 2011-12-23 2017-04-04 DePuy Synthes Products, Inc. Detection of human umbilical cord tissue derived cells
US9763983B2 (en) 2013-02-05 2017-09-19 Anthrogenesis Corporation Natural killer cells from placenta
US9446077B2 (en) 2013-03-13 2016-09-20 Allosource Fascia fibrous compositions and methods for their use and manufacture
US10568990B2 (en) 2013-03-15 2020-02-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
US11229725B2 (en) 2013-03-15 2022-01-25 Allosource Cell repopulated collagen matrix for soft tissue repair and regeneration
US10918853B2 (en) 2013-05-30 2021-02-16 Neurostim Solutions, Llc Topical neurological stimulation
US11291828B2 (en) 2013-05-30 2022-04-05 Neurostim Solutions LLC Topical neurological stimulation
US10946185B2 (en) 2013-05-30 2021-03-16 Neurostim Solutions, Llc Topical neurological stimulation
US10307591B2 (en) 2013-05-30 2019-06-04 Neurostim Solutions, Llc Topical neurological stimulation
US10016600B2 (en) 2013-05-30 2018-07-10 Neurostim Solutions, Llc Topical neurological stimulation
US11229789B2 (en) 2013-05-30 2022-01-25 Neurostim Oab, Inc. Neuro activator with controller
US10842910B2 (en) 2013-12-06 2020-11-24 Allosource Systems for drying sheets of donor-provided human tissue
US9795707B2 (en) 2013-12-06 2017-10-24 Allosource Methods of drying sheets of donor-provided human birth tissue
WO2016068616A1 (en) * 2014-10-29 2016-05-06 차의과학대학교 산학협력단 Placenta-derived cells excreting c3 or c1r complement and composition containing same
US11077301B2 (en) 2015-02-21 2021-08-03 NeurostimOAB, Inc. Topical nerve stimulator and sensor for bladder control
US10772986B2 (en) 2017-01-26 2020-09-15 Allosource Fascia fibrous compositions and methods for their use and manufacture
US10953225B2 (en) 2017-11-07 2021-03-23 Neurostim Oab, Inc. Non-invasive nerve activator with adaptive circuit
US11458311B2 (en) 2019-06-26 2022-10-04 Neurostim Technologies Llc Non-invasive nerve activator patch with adaptive circuit
US11730958B2 (en) 2019-12-16 2023-08-22 Neurostim Solutions, Llc Non-invasive nerve activator with boosted charge delivery

Also Published As

Publication number Publication date
WO2005001077A2 (en) 2005-01-06
WO2005003334A3 (en) 2005-04-07
ES2554343T3 (en) 2015-12-18
EP1641914A2 (en) 2006-04-05
US20060154367A1 (en) 2006-07-13
US20170143770A1 (en) 2017-05-25
US20050058631A1 (en) 2005-03-17
EP2322599B1 (en) 2015-04-15
CA2530732A1 (en) 2005-01-06
US9579351B2 (en) 2017-02-28
US10383898B2 (en) 2019-08-20
PL1641917T3 (en) 2016-11-30
US9717763B2 (en) 2017-08-01
PL1641916T3 (en) 2016-08-31
WO2005001079A2 (en) 2005-01-06
EP1641918A2 (en) 2006-04-05
US20100210013A1 (en) 2010-08-19
US20070009494A1 (en) 2007-01-11
PL1641915T3 (en) 2017-01-31
US20170166864A1 (en) 2017-06-15
US8277796B2 (en) 2012-10-02
ES2542070T3 (en) 2015-07-30
AU2004252570C1 (en) 2012-03-01
JP2007528706A (en) 2007-10-18
ES2564044T3 (en) 2016-03-17
WO2005001076A2 (en) 2005-01-06
EP2341131A2 (en) 2011-07-06
JP2007521793A (en) 2007-08-09
EP2338980A2 (en) 2011-06-29
CA2530533C (en) 2015-02-10
AU2004254616A1 (en) 2005-01-13
CA2530421A1 (en) 2005-01-06
EP1641913B1 (en) 2016-01-06
EP1641916B1 (en) 2016-02-17
AU2004281371A1 (en) 2005-04-28
PL1641913T3 (en) 2016-06-30
US10500234B2 (en) 2019-12-10
PL2338980T3 (en) 2016-04-29
US7524489B2 (en) 2009-04-28
US10758576B2 (en) 2020-09-01
WO2005001078A3 (en) 2005-04-21
US20060154366A1 (en) 2006-07-13
US7510873B2 (en) 2009-03-31
PL2338981T3 (en) 2016-01-29
ES2552226T3 (en) 2015-11-26
JP4950659B2 (en) 2012-06-13
CA2530412A1 (en) 2005-04-28
EP2399990B1 (en) 2015-07-22
US20170166863A1 (en) 2017-06-15
PL1649013T3 (en) 2016-07-29
CA2530732C (en) 2015-03-31
ES2569780T3 (en) 2016-05-12
WO2005003334A2 (en) 2005-01-13
ES2565582T3 (en) 2016-04-05
EP2338980B1 (en) 2015-04-22
US20050032209A1 (en) 2005-02-10
EP2322599A2 (en) 2011-05-18
US20070014771A1 (en) 2007-01-18
JP4950660B2 (en) 2012-06-13
PL2336298T3 (en) 2016-08-31
WO2005001079A3 (en) 2005-04-28
AU2004254616B2 (en) 2011-09-01
EP2338982A2 (en) 2011-06-29
US20060153816A1 (en) 2006-07-13
US10220059B2 (en) 2019-03-05
ES2550267T3 (en) 2015-11-05
EP1641915A2 (en) 2006-04-05
EP1649013A2 (en) 2006-04-26
ES2542069T3 (en) 2015-07-30
PL2322599T3 (en) 2015-10-30
EP2338981B1 (en) 2015-04-22
WO2005001077A3 (en) 2005-03-03
US11179422B2 (en) 2021-11-23
AU2004252571C1 (en) 2012-03-01
ES2568463T3 (en) 2016-04-29
AU2004252568A1 (en) 2005-01-06
AU2004254616C1 (en) 2012-03-01
AU2004252566A1 (en) 2005-01-06
EP2322599A3 (en) 2011-07-27
US10195233B2 (en) 2019-02-05
US20170326183A1 (en) 2017-11-16
WO2005038012A3 (en) 2005-09-15
US20100260843A1 (en) 2010-10-14
EP1641918B1 (en) 2016-01-06
US8318483B2 (en) 2012-11-27
AU2004252566B2 (en) 2011-09-15
AU2004252567B2 (en) 2011-10-06
PL1641914T3 (en) 2017-01-31
JP2007528705A (en) 2007-10-18
JP2007528703A (en) 2007-10-18
CA2530422A1 (en) 2005-01-06
EP1641917B1 (en) 2016-04-27
PL1641918T3 (en) 2016-06-30
US20170239299A1 (en) 2017-08-24
WO2005001080A2 (en) 2005-01-06
US11191789B2 (en) 2021-12-07
US11000554B2 (en) 2021-05-11
JP4948165B2 (en) 2012-06-06
EP2341131A3 (en) 2011-08-03
EP1641913A2 (en) 2006-04-05
CA2530422C (en) 2016-01-05
EP2336298A3 (en) 2011-08-03
JP5148873B2 (en) 2013-02-20
PL2341131T3 (en) 2016-01-29
US20050054098A1 (en) 2005-03-10
US8658152B2 (en) 2014-02-25
JP2007521008A (en) 2007-08-02
AU2004252571B2 (en) 2011-09-15
EP2336298B1 (en) 2016-02-17
EP2338980A3 (en) 2011-08-10
ES2600555T3 (en) 2017-02-09
WO2005038012A2 (en) 2005-04-28
US20050058629A1 (en) 2005-03-17
US20060188983A1 (en) 2006-08-24
US9498501B2 (en) 2016-11-22
ES2597837T3 (en) 2017-01-23
EP1641914B1 (en) 2016-07-20
CA2530412C (en) 2016-02-09
AU2004252570A1 (en) 2005-01-06
CA2530421C (en) 2015-04-21
PL2338982T3 (en) 2016-01-29
US20140045263A1 (en) 2014-02-13
EP1649013B1 (en) 2016-01-20
ES2541604T3 (en) 2015-07-22
EP2338982B1 (en) 2015-08-26
EP2341131B1 (en) 2015-08-05
JP4950661B2 (en) 2012-06-13
AU2004281371C1 (en) 2012-10-11
US20190192577A1 (en) 2019-06-27
EP2399990A1 (en) 2011-12-28
EP2338981A3 (en) 2011-08-03
US20050037491A1 (en) 2005-02-17
US20070036767A1 (en) 2007-02-15
JP2007521009A (en) 2007-08-02
CA2530416C (en) 2015-08-25
AU2004252567A1 (en) 2005-01-06
ES2582342T3 (en) 2016-09-12
EP2336298A2 (en) 2011-06-22
PL2399990T3 (en) 2015-12-31
WO2005001078A2 (en) 2005-01-06
US8703121B2 (en) 2014-04-22
JP2007528702A (en) 2007-10-18
US20050058630A1 (en) 2005-03-17
CA2530255A1 (en) 2005-01-06
US20060153817A1 (en) 2006-07-13
US20060153818A1 (en) 2006-07-13
JP4948166B2 (en) 2012-06-06
JP4948164B2 (en) 2012-06-06
US7413734B2 (en) 2008-08-19
AU2004252571A1 (en) 2005-01-06
AU2004252570B2 (en) 2011-09-15
US10039793B2 (en) 2018-08-07
EP1641915B1 (en) 2016-07-27
CA2530255C (en) 2015-12-08
EP2338982A3 (en) 2011-08-03
AU2004252568B2 (en) 2011-06-30
US7560276B2 (en) 2009-07-14
WO2005001076A3 (en) 2005-03-31
US9504719B2 (en) 2016-11-29
CA2530416A1 (en) 2005-01-06
US20130022585A1 (en) 2013-01-24
EP1641916A2 (en) 2006-04-05
US20050019865A1 (en) 2005-01-27
AU2004281371B2 (en) 2012-05-10
WO2005001080A3 (en) 2005-03-24
US20180338999A1 (en) 2018-11-29
ES2564045T3 (en) 2016-03-17
CA2530533A1 (en) 2005-01-13
EP2338981A2 (en) 2011-06-29
EP1641917A2 (en) 2006-04-05

Similar Documents

Publication Publication Date Title
US7413734B2 (en) Treatment of retinitis pigmentosa with human umbilical cord cells
US10744164B2 (en) Repair and regeneration of ocular tissue using postpartum-derived cells
US9572840B2 (en) Regeneration and repair of neural tissue using postpartum-derived cells
US20160166619A1 (en) Treatment of Retinal Degeneration Using Progenitor Cells
US20160158293A1 (en) Treatment of Ocular Conditions Using Progenitor Cells
US20170157179A1 (en) Treatment of retinal degeneration using progenitor cells
WO2018102174A1 (en) Treatment of retinal degeneration using progenitor cells
US20180015129A1 (en) Treatment of retinal vascular disease using progenitor cells
AU2011226961B2 (en) Regeneration and repair of neural tissue using postpartum-derived cells

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO PAY ISSUE FEE