US20080107633A1 - Cultures of human CNS neural stem cells - Google Patents

Cultures of human CNS neural stem cells Download PDF

Info

Publication number
US20080107633A1
US20080107633A1 US11/903,408 US90340807A US2008107633A1 US 20080107633 A1 US20080107633 A1 US 20080107633A1 US 90340807 A US90340807 A US 90340807A US 2008107633 A1 US2008107633 A1 US 2008107633A1
Authority
US
United States
Prior art keywords
cells
neural stem
stem cells
human
cell
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
US11/903,408
Inventor
Melissa Carpenter
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.)
Individual
Original Assignee
Individual
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=25453045&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080107633(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US10/328,644 external-priority patent/US20030166276A1/en
Application filed by Individual filed Critical Individual
Priority to US11/903,408 priority Critical patent/US20080107633A1/en
Publication of US20080107633A1 publication Critical patent/US20080107633A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K35/30Nerves; Brain; Eyes; Corneal cells; Cerebrospinal fluid; Neuronal stem cells; Neuronal precursor cells; Glial cells; Oligodendrocytes; Schwann cells; Astroglia; Astrocytes; Choroid plexus; Spinal cord tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • 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
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal 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
    • A61K2035/126Immunoprotecting barriers, e.g. jackets, diffusion chambers
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • 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
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • C12N2500/24Iron; Fe chelators; Transferrin
    • C12N2500/25Insulin-transferrin; Insulin-transferrin-selenium
    • 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
    • C12N2500/90Serum-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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • 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
    • C12N2501/11Epidermal growth factor [EGF]
    • 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
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • 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]
    • C12N2501/235Leukemia inhibitory factor [LIF]
    • 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
    • C12N2501/30Hormones
    • C12N2501/38Hormones with nuclear receptors
    • C12N2501/39Steroid hormones
    • C12N2501/392Sexual steroids
    • 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
    • C12N2503/00Use of cells in diagnostics
    • C12N2503/02Drug screening

Definitions

  • This invention relates to isolation of human central nervous system stem cells, and methods and media for proliferating, differentiating and transplanting them.
  • neural stem cells During development of the central nervous system (“CNS”), multipotent precursor cells, also known as neural stem cells, proliferate, giving rise to transiently dividing progenitor cells that eventually differentiate into the cell types that compose the adult brain. Stem cells (from other tissues) have classically been defined as having the ability to self-renew (i.e., form more stem cells), to proliferate, and to differentiate into multiple different phenotypic lineages. In the case of neural stem cells this includes neurons, astrocytes and oligodendrocytes.
  • Potten and Loeffler (Development, 110:1001, 1990) define stem cells as “undifferentiated cells capable of a) proliferation, b) self-maintenance, c) the production of a large number of differentiated functional progeny, d) regenerating the tissue after injury, and e) a flexibility in the use of these options.”
  • neural stem cells have been isolated from several mammalian species, including mice, rats, pigs and humans. See, e.g., WO 93/01275, WO 94/09119, WO 94/10292, WO 94/16718 and Cattaneo et al., Mol. Brain. Res., 42, pp. 161-66 (1996), all herein incorporated by reference.
  • Human CNS neural stem cells like their rodent homologues, when maintained in a mitogen-containing (typically epidermal growth factor or epidermal growth factor plus basic fibroblast growth factor), serum-free culture medium, grow in suspension culture to form aggregates of cells known as “neurospheres”.
  • mitogen-containing typically epidermal growth factor or epidermal growth factor plus basic fibroblast growth factor
  • serum-free culture medium grow in suspension culture to form aggregates of cells known as “neurospheres”.
  • human neural stem cells have doubling rates of about 30 days. See, e.g., Cattaneo et al., Mol. Brain. Res., 42, pp. 161-66 (1996).
  • the stem cells Upon removal of the mitogen(s) and provision of a substrate, the stem cells differentiate into neurons, astrocytes and oligodendrocytes.
  • the majority of cells in the differentiated cell population have been identified as astrocytes, with very few neurons ( ⁇ 10%) being observed.
  • This invention provides novel human central nervous system stem cells, and methods and media for proliferating, differentiating and transplanting them.
  • this invention provides novel human stem cells with a doubling rate of between 5-10 days, as well as defined growth media for prolonged proliferation of human neural stem cells.
  • this invention provides a defined media for differentiation of human neural stem cells so as to enrich for neurons, oligodendrocytes, astrocytes, or a combination thereof.
  • the invention also provides differentiated cell populations of human neural stem cells that provide previously unobtainable large numbers of neurons, as well as astrocytes and oligodendrocytes.
  • This invention also provides novel methods for transplanting neural stem cells that improve the viability of the graft upon implantation in a host.
  • FIG. 1 shows a representation of spheres of proliferating 9FBr human neural stem cells (passage 6) derived from human forebrain tissue.
  • FIG. 2 Panel A, shows a growth curve for a human neural stem cell line designated 6.5Fbr cultured in (a) defined media containing EGF, FGF and leukemia inhibitory factor (“LIF”) (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds);
  • Panel B shows a growth curve for a human neural stem cell line designated 9Fbr cultured in (a) defined media containing EGF, FGF and LIF (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds);
  • Panel C shows a growth curve for a human neural stem cell line designated 9.5Fbr cultured in (a) defined media containing EGF, FGF and LIF (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds);
  • Panel D shows a growth curve for a human neural stem cell line designated 10.5Fbr cultured in (a) defined media containing EGF, FGF and leuk
  • FIG. 3 shows a growth curve for a human neural stem cell line designated 9Fbr cultured in (a) defined media containing EGF and basic fibroblast growth factor (“bFGF”) (shown as open diamonds), and (b) defined media with EGF but without bFGF (shown as closed diamonds).
  • bFGF basic fibroblast growth factor
  • FIG. 4 shows a graph of cell number versus days in culture for an Mx-1 conditionally immortalized human glioblast line derived from a human neural stem cell line.
  • the open squares denote growth in the presence of interferon, the closed diamonds denote growth in the absence of interferon.
  • This invention relates to isolation, characterization, proliferation, differentiation and transplantation of CNS neural stem cells.
  • the neural stem cells described and claimed in the applications may be proliferated in suspension culture or in adherent culture.
  • human nestin antibody may be used as a marker to identify undifferentiated cells.
  • the proliferating cells show little GFAP staining and little ⁇ -tubulin staining (although some staining might be present due to diversity of cells within the spheres).
  • Neurons may be identified using antibodies to neuron specific enolase (“NSE”), neurofilament, tau, beta-tubulin, or other known neuronal markers.
  • NSE neuron specific enolase
  • Astrocytes may be identified using antibodies to glial fibrillary acidic protein (“GFAP”), or other known astrocytic markers.
  • Oligodendrocytes may be identified using antibodies to galactocerebroside, O4, myelin basic protein (“MBP”) or other known oligodendrocytic markers.
  • Glial cells in general may be identified by staining with antibodies, such as the M2 antibody, or other known glial markers.
  • the invention provides novel human CNS stem cells isolated from the forebrain.
  • Neural stem cells can be induced to proliferate and differentiate either by culturing the cells in suspension or on an adherent substrate. See, e.g., U.S. Pat. No. 5,750,376 and U.S. Pat. No. 5,753,506 (both incorporated herein by reference in their entirety), and prior art medium described therein. Both allografts and autografts are contemplated for transplantation purposes.
  • This invention also provides a novel growth media for proliferation of neural stem cells.
  • a serum-free or serum-depleted culture medium for the short term and long term proliferation of neural stem cells.
  • neural stem cells Prior to development of the novel media described herein, neural stem cells have been cultured in serum-free media containing epidermal growth factor (“EGF”) or an analog of EGF, such as amphiregulin or transforming growth factor alpha (“TGF- ⁇ ”), as the mitogen for proliferation. See, e.g., WO 93/01275, WO 94/16718, both incorporated herein by reference. Further, basic fibroblast growth factor (“bFGF”) has been used, either alone, or in combination with EGF, to enhance long term neural stem cell survival.
  • EGF epidermal growth factor
  • TGF- ⁇ transforming growth factor alpha
  • the improved medium according to this invention which contains leukemia inhibitory factor (“LIF”), markedly and unexpectedly increases the rate of proliferation of neural stem cells, particularly human neural stem cells.
  • LIF leukemia inhibitory factor
  • Standard culture media typically contains a variety of essential components required for cell viability, including inorganic salts, carbohydrates, hormones, essential amino acids, vitamins, and the like.
  • DMEM or F-12 as the standard culture medium, most preferably a 50/50 mixture of DMEM and F-12. Both media are commercially available (DMEM-Gibco 12100-046; F-12-Gibco 21700-075).
  • a premixed formulation is also commercially available (N2-Gibco 17502-030). It is advantageous to provide additional glutamine, preferably at about 2 mM. It is also advantageous to provide heparin in the culture medium.
  • the conditions for culturing should be as close to physiological as possible.
  • the pH of the culture medium is typically between 6-8, preferably about 7, most preferably about 7.4.
  • Cells are typically cultured between 30-40° C., preferably between 32-38° C., most preferably between 35-37° C.
  • Cells are preferably grown in 5% CO 2 .
  • Cells are preferably grown in suspension culture.
  • the neural stem cell culture comprises the following components in the indicated concentrations: Component Final Concentration 50/50 mix of DMEM/F-12 0.5-2.0 ⁇ , preferably 1 ⁇ glucose 0.2-1.0%, preferably 0.6% w/v glutamine 0.1-10 mM, preferably 2 mM NaHCO 3 0.1-10 mM, preferably 3 mM HEPES 0.1-10 mM, preferably 5 mM apo-human transferrin (Sigma 1-1000 ⁇ g/ml, preferably 100 ⁇ g/ml T-2252) human insulin (Sigma I-2767) 1-100, preferably 25 ⁇ g/ml putrescine (Sigma P-7505) 1-500, preferably 60 ⁇ M selenium (Sigma S-9133) 1-100, preferably 30 nM progesterone (Sigma P-6149) 1-100, preferably 20 nM human EGF (Gibco 13247-010) 0.2-200, preferably 20 ng/ml human bFGF (Sigma 1-1000
  • the human neural stem cells described herein may be cryopreserved according to routine procedures. We prefer cryopreserving about one to ten million cells in “freeze” medium which consists of proliferation medium (absent the growth factor mitogens), 10% BSA (Sigma A3059) and 7.5% DMSO. Cells are centrifuged. Growth medium is aspirated and replaced with freeze medium. Cells are resuspended gently as spheres, not as dissociated cells. Cells are slowly frozen, by, e.g., placing in a container at ⁇ 80° C. Cells are thawed by swirling in a 37° C. bath, resuspended in fresh proliferation medium, and grown as usual.
  • “freeze” medium which consists of proliferation medium (absent the growth factor mitogens), 10% BSA (Sigma A3059) and 7.5% DMSO. Cells are centrifuged. Growth medium is aspirated and replaced with freeze medium. Cells are resuspended gently as spheres, not as dissociated
  • this invention provides a differentiated cell culture containing previously unobtainable large numbers of neurons, as well as astrocytes and oligodendrocytes.
  • typically the differentiated human diencephalon-derived neural stem cell cultures formed very few neurons (i.e., less than 5-10%).
  • neuron concentrations of between 20% and 35% (and much higher in other cases) in differentiated human forebrain-derived neural stem cell cultures. This is highly advantageous as it permits enrichment of the neuronal population prior to implantation in the host in disease indications where neuronal function has been impaired or lost.
  • GABA-ergic neuron enriched cell cultures are particularly advantageous in the potential therapy of excitotoxic neurodegenerative disorders, such as Huntington's disease or epilepsy.
  • various cell surface or intracellular markers may be used.
  • the neural stem cells of this invention are proliferating as neurospheres, we contemplate using human nestin antibody as a marker to identify undifferentiated cells.
  • the proliferating cells should show little GFAP staining and little ⁇ -tubulin staining (although some staining might be present due to diversity of cells within the spheres).
  • Neurons may be identified using antibodies to neuron specific enolase (“NSE”), neurofilament, tau, ⁇ -tubulin, or other known neuronal markers.
  • NSE neuron specific enolase
  • Astrocytes may be identified using antibodies to glial fibrillary acidic protein (“GFAP”), or other known astrocytic markers.
  • Oligodendrocytes may be identified using antibodies to galactocerebroside, O4, myelin basic protein (“MBP”) or other known oligodendrocytic markers.
  • cell phenotypes by identifying compounds characteristically produced by those phenotypes.
  • neurotransmitters such as acetylcholine, dopamine, epinephrine, norepinephrine, and the like.
  • GABA-ergic neurons may be identified by their production of glutamic acid decarboxylase (“GAD”) or GABA.
  • GABA glutamic acid decarboxylase
  • DDC dopa decarboxylase
  • TH dopamine or tyrosine hydroxylase
  • Cholinergic neurons may be identified by their production of choline acetyltransferase (“ChAT”).
  • Hippocampal neurons may be identified by staining with NeuN. It will be appreciated that any suitable known marker for identifying specific neuronal phenotypes may be used.
  • the human neural stem cells described herein can be genetically engineered or modified according to known methodology.
  • the term “genetic modification” refers to the stable or transient alteration of the genotype of a cell by intentional introduction of exogenous DNA.
  • DNA may be synthetic, or naturally derived, and may contain genes, portions of genes, or other useful DNA sequences.
  • the term “genetic modification” is not meant to include naturally occurring alterations such as that which occurs through natural viral activity, natural genetic recombination, or the like.
  • a gene of interest i.e., a gene that encodes a biologically active molecule
  • a gene of interest can be inserted into a cloning site of a suitable expression vector by using standard techniques. These techniques are well known to those skilled in the art. See, e.g., WO 94/16718, incorporated herein by reference.
  • the expression vector containing the gene of interest may then be used to transfect the desired cell line.
  • Standard transfection techniques such as calcium phosphate co-precipitation, DEAE-dextran transfection, electroporation, biolistics, or viral transfection may be utilized.
  • Commercially available mammalian transfection kits may be purchased from e.g., Stratagene. Human adenoviral transfection may be accomplished as described in Berg et al. Exp. Cell Res., 192, pp. (1991). Similarly, lipofectamine-based transfection may be accomplished as described in Cattaneo, Mol. Brain. Res., 42, pp. 161-66 (1996).
  • host/expression vector combinations may be used to express a gene encoding a biologically active molecule of interest. See, e.g., U.S. Pat. No. 5,545,723, herein incorporated by reference, for suitable cell-based production expression vectors.
  • Increased expression of the biologically active molecule can be achieved by increasing or amplifying the transgene copy number using amplification methods well known in the art.
  • amplification methods include, e.g., DHFR amplification (see, e.g., Kaufman et al., U.S. Pat. No. 4,470,461) or glutamine synthetase (“GS”) amplification (see, e.g., U.S. Pat. No. 5,122,464, and European published application EP 338,841), all herein incorporated by reference.
  • the genetically modified neural stem cells are derived from transgenic animals.
  • the substance will preferably be useful for the treatment of a CNS disorder.
  • genetically modified neural stem cells that are capable of secreting a therapeutically effective biologically active molecule in patients.
  • the genetically modified neural stem cells thus provide cell-based delivery of biological agents of therapeutic value.
  • the neural stem cells described herein, and their differentiated progeny may be immortalized or conditionally immortalized using known techniques.
  • conditional immortalization of stem cells and most preferably conditional immortalization of their differentiated progeny.
  • conditional immortalization techniques contemplated are Tet-conditional immortalization (see WO 96/31242, incorporated herein by reference), and Mx-1 conditional immortalization (see WO 96/02646, incorporated herein by reference).
  • This invention also provides methods for differentiating neural stem cells to yield cell cultures enriched with neurons to a degree previously unobtainable.
  • the proliferating neurospheres are induced to differentiate by removal of the growth factor mitogens and LIF, and provision of 1% serum, a substrate and a source of ionic charges (e.g., glass cover slip covered with poly-ornithine or extracellular matrix components).
  • the preferred base medium for this differentiation protocol excepting the growth factor mitogens and LIF, is otherwise the same as the proliferation medium.
  • This differentiation protocol produces a cell culture enriched in neurons. According to this protocol, we have routinely achieved neuron concentrations of between 20% and 35% in differentiated human forebrain-derived neural stem cell cultures.
  • the proliferating neurospheres are induced to differentiate by removal of the growth factor mitogens, and provision of 1% serum, a substrate and a source of ionic charges (e.g., glass cover slip covered with poly-ornithine or extracellular matrix components), as well as a mixture of growth factors including PDGF, CNTF, IGF-1, LIF, forskolin, T-3 and NT-3.
  • the cocktail of growth factors may be added at the same time as the neurospheres are removed from the proliferation medium, or may be added to the proliferation medium and the cells pre-incubated with the mixture prior to removal from the mitogens.
  • This protocol produces a cell culture highly enriched in neurons and enriched in oligodendrocytes. According to this protocol, we have routinely achieved neuron concentrations of higher than 35% in differentiated human forebrain-derived neural stem cell cultures.
  • bFGF is trophic for the oligodendrocyte precursor cell line. Oligodendrocytes are induced under differentiation conditions when passaged with EGF and LIF in proliferating media, without bFGF.
  • the human stem cells of this invention have numerous uses, including for drug screening, diagnostics, genomics and transplantation.
  • Stem cells can be induced to differentiate into the neural cell type of choice using the appropriate media described in this invention.
  • the drug to be tested can be added prior to differentiation to test for developmental inhibition, or added post-differentiation to monitor neural cell-type specific reactions.
  • the cells of this invention may be transplanted “naked” into patients according to conventional techniques, into the CNS, as described for example, in U.S. Pat. Nos. 5,082,670 and 5,618,531, each incorporated herein by reference, or into any other suitable site in the body.
  • the human stem cells are transplanted directly into the CNS. Parenchymal and intrathecal sites are contemplated. It will be appreciated that the exact location in the CNS will vary according to the disease state.
  • Implanted cells may be labeled with bromodeoxyuridine (BrdU) prior to transplantation.
  • BrdU bromodeoxyuridine
  • Transplantation of human forebrain derived neural stem cells to the hippocampus produced neurons that were predominantly NeuN staining but GABA negative.
  • the NeuN antibody is known to stain neurons of the hippocampus.
  • GABA-ergic neurons were formed when these same cell lines were transplanted into the striatum.
  • transplanted cells respond to environmental clues in both the adult and the neonatal brain.
  • graft viability improves if the transplanted neural stem cells are allowed to aggregate, or to form neurospheres prior to implantation, as compared to transplantation of dissociated single cell suspensions.
  • transplanting small sized neurospheres approximately 10-500 ⁇ m in diameter, preferably 40-50 ⁇ m in diameter.
  • spheres containing about 5-100, preferably 5-20 cells per sphere.
  • the cells may also be encapsulated and used to deliver biologically active molecules, according to known encapsulation technologies, including microencapsulation (see, e.g., U.S. Pat. Nos. 4,352,883; 4,353,888; and 5,084,350, herein incorporated by reference), (b) macroencapsulation (see, e.g., U.S. Pat. Nos. 5,284,761, 5,158,881, 4,976,859 and 4,968,733 and published PCT patent applications WO92/19195, WO 95/05452, each incorporated herein by reference).
  • microencapsulation see, e.g., U.S. Pat. Nos. 4,352,883; 4,353,888; and 5,084,350, herein incorporated by reference
  • macroencapsulation see, e.g., U.S. Pat. Nos. 5,284,761, 5,158,881, 4,976,859 and 4,968,733 and published PCT patent applications WO92/19195, WO 95/05
  • the human neural stem cells are encapsulated, we prefer macroencapsulation, as described in U.S. Pat. Nos. 5,284,761; 5,158,881; 4,976,859; 4,968,733; 5,800,828 and published PCT patent application WO 95/05452, each incorporated herein by reference.
  • Cell number in the devices can be varied; preferably each device contains between 10 3 -10 9 cells, most preferably 10 5 to 10 7 cells.
  • a large number of macroencapsulation devices may be implanted in the patient; we prefer between one to 10 devices.
  • a capsular device secretes a biologically active molecule that is therapeutically effective in the patient or that produces a biologically active molecule that has a growth or trophic effect on the transplanted neural stem cells, or that induces differentiation of the neural stem cells towards a particular phenotypic lineage.
  • the cells and methods of this invention may be useful in the treatment of various neurodegenerative diseases and other disorders. It is contemplated that the cells will replace diseased, damaged or lost tissue in the host. Alternatively, the transplanted tissue may augment the function of the endogenous affected host tissue.
  • the transplanted neural stem cells may also be genetically modified to provide a therapeutically effective biologically active molecule.
  • Neural stem cells may provide one means of preventing or replacing the cell loss and associated behavioral abnormalities of these disorders.
  • Neural stem cells may replace cerebellar neurons lost in cerebellar ataxia, with clinical outcomes readily measurable by methods known in the medical arts.
  • Huntington's disease is an autosomal dominant neurodegenerative disease characterized by a relentlessly progressive movement disorder with devastating psychiatric and cognitive deterioration. HD is associated with a consistent and severe atrophy of the neostriatum which is related to a marked loss of the GABAergic medium-sized spiny projection neurons, the major output neurons of the striatum.
  • Intrastriatal injections of excitotoxins such as quinolinic acid (QA) mimic the pattern of selective neuronal vulnerability seen in HD. QA lesions result in motor and cognitive deficits which are among the major symptoms seen in HD.
  • intrastriatal injections of QA have become a useful model of HD and can serve to evaluate novel therapeutic strategies aimed at preventing, attenuating, or reversing neuroanatomical and behavioral changes associated with HD.
  • GABA-ergic neurons are characteristically lost in Huntington's disease, we contemplate treatment of Huntington's patients by transplantation of cell cultures enriched in GABA-ergic neurons derived according to the methods of this invention.
  • Epilepsy is also associated with excitotoxicity. Accordingly, GABA-ergic neurons derived according to this invention are contemplated for transplantation into patients suffering from epilepsy.
  • oligodendrocytes or oligodendrocyte precursor or progenitors such cultures prepared and transplanted according to this invention to promote remyelination of demyelinated areas in the host.
  • the cells and methods of this invention are intended for use in a mammalian host, recipient, patient, subject or individual, preferably a primate, most preferably a human.
  • Proliferation medium was prepared with the following components in the indicated concentrations: Component Final Concentration 50/50 mix of DMEM/F-12 1 ⁇ glucose 0.6% w/v glutamine 2 mM NaHCO 3 3 mM HEPES 5 mM apo-human transferrin (Sigma T-2252) 100 ⁇ g/ml human insulin (Sigma I-2767) 25 ⁇ g/ml putrescine (Sigma P-7505) 60 ⁇ M selenium (Sigma S-9133) 30 nM progesterone (Sigma P-6149) 20 nM human EGF (Gibco 13247-010) 20 ng/ml human bFGF (Gibco 13256-029) 20 ng/ml human LIF (R&D Systems 250-L) 10 ng/ml heparin (Sigma H-3149) 2 ⁇ g/ml
  • Sample tissue from human embryonic forebrain was collected and dissected in Sweden and kindly provided by Huddinje Sjukhus. Blood samples from the donors were sent for viral testing. Dissections were performed in saline and the selected tissue was placed directly into proliferation medium (as described in Example 1). Tissue was stored at 4° C. until dissociated. The tissue was dissociated using a standard glass homogenizer, without the presence of any digesting enzymes. The dissociated cells were counted and seeded into flasks containing proliferation medium. After 5-7 days, the contents of the flasks are centrifuged at 1000 rpm for 2 min. The supernatant was aspirated and the pellet resuspended in 200 ⁇ l of proliferation medium.
  • the cell clusters were triturated using a P200 pipetman about 100 times to break up the clusters. Cells were reseeded at 75,000-100,000 cells/ml into proliferation medium. Cells were passaged every 6-21 days depending upon the mitogens used and the seeding density. Typically these cells incorporate BrdU, indicative of cell proliferation. For T75 flask cultures (initial volume 20 ml), cells are “fed” 3 times weekly by addition of 5 ml of proliferation medium. We prefer Nunc flasks for culturing.
  • nestin a measure of proliferating neurospheres
  • Cells were fixed for 20 min at room temperature with 4% paraformaldehyde. Cells were washed twice for 5 min with 0.1 M PBS, pH 7.4. Cells were permeabilized for 2 min with 100% EtOH. The cells were then washed twice for 5 min with 0.1 M PBS. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4 and 1% Triton X-100 (Sigma X-100) for 1 hr at room temperature with gentle shaking. Cells were incubated with primary antibodies to human nestin (from Dr.
  • NGS normal goat serum
  • FIG. 1 shows a picture of proliferating spheres (here called “neurospheres”) of human forebrain derived neural stem cells.
  • neurospheres proliferating spheres
  • LIF significantly increased the rate of cell proliferation.
  • the effect of LIF was most pronounced after about 60 days in vitro.
  • the proliferating neurospheres were induced to differentiate by removal of the growth factor mitogens and LIF, and provision of 1% serum, a substrate and a source of ionic charges (e.g., glass cover slip covered with poly-ornithine).
  • a source of ionic charges e.g., glass cover slip covered with poly-ornithine
  • the staining protocol for neurons, astrocytes and oligodendrocytes was as follows:
  • DAPI nuclear stain
  • This differentiation protocol produced cell cultures enriched in neurons as follows:
  • the proliferating neurospheres were induced to differentiate by removal of the growth factor mitogens and LIF, and provision of 1% serum, a substrate (e.g., glass cover slip or extracellular matrix components), a source of ionic charges (e.g., poly-ornithine) as well as a mixture of growth factors including 10 ng/ml PDGF A/B, 10 ng/ml CNTF, 10 ng/ml IGF-1, 10 ⁇ M forskolin, 30 ng/ml T3, 10 ng/ml LIF and 1 ng/ml NT-3.
  • This differentiation protocol produced cell cultures highly enriched in neurons (i.e., greater than 35% of the differentiated cell culture) and enriched in oligodendrocytes.
  • cell suspensions were initially cultured in a cocktail of hbFGF, EGF, and LIF, were then placed into altered growth media containing 20 ng/mL hEGF (GIBCO) and 10 ng/mL human leukemia inhibitory factor (hLIF) (R&D Systems), but without hbFGF.
  • hbFGF human leukemia inhibitory factor
  • the cells initially grew significantly more slowly than the cultures that also contained hbFGF (see FIG. 3 ). Nonetheless, the cells continued to grow and were passaged as many as 22 times.
  • Stem cells were removed from growth medium and induced to differentiate by plating on poly-ornithine coated glass coverslips in differentiation medium supplemented with a growth factor cocktail (hPDGF A/B, hCNTF, hGF-1, forskolin, T3 and hNT-3).
  • hPDGF A/B a growth factor cocktail
  • hCNTF a growth factor cocktail
  • hGF-1 a growth factor-1
  • forskolin T3 and hNT-3
  • this protocol produced differentiated cell cultures enrichment in oligodendrocytes. Neurons were only occasionally seen, had small processes, and appeared quite immature.
  • Mx-1 glioblast cell line derived from the human neural stem cells described herein, using the Mx-1 system described in WO 96/02646.
  • the Mx-1 promoter drives expression of the SV40 large T antigen.
  • the Mx-1 promoter is induced by interferon. When induced, large T is expressed, and quiescent cells proliferate.
  • Human glioblasts were derived from human forebrain neural stem cells as follows. Proliferating human neurospheres were removed from proliferation medium and plated onto poly-ornithine plastic (24 well plate) in a mixture of N2 with the mitogens EGF, bFGF and LIF, as well as 0.5% FBS. 0.5 ml of N2 medium and 1% FBS was added. The cells were incubated overnight. The cells were then transfected with p318 (a plasmid containing the Mx-1 promoter operably linked to the SV 40 large T antigen) using Invitrogen lipid kit (lipids 4 and 6). The transfection solution contained 6 ⁇ l/ml of lipid and 4 ⁇ l/ml DNA in optiMEM medium.
  • p318 a plasmid containing the Mx-1 promoter operably linked to the SV 40 large T antigen
  • the cells were incubated in transfection solution for 5 hours.
  • the transfection solution was removed and cells placed into N2 and 1% FBS and 500 U/ml A/D interferon.
  • the cells were fed twice a week. After ten weeks cells were assayed for large T antigen expression. The cells showed robust T antigen staining at this time. As FIG. 4 shows, cell number was higher in the presence of interferon than in the absence of interferon.
  • T expression was monitored using immunocytochemistry as follows. Cells were fixed for 20 min at room temperature with 4% paraformaldehyde. Cells were washed twice for 5 min with 0.1 M PBS, pH 7.4. Cells were permeabilized for 2 min with 100% EtOH. The cells were then washed twice for 5 min with 0.1 M PBS. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4. Cells were incubated with primary antibodies to large T antigen (used at 1:10) diluted in 1% NGS for 2 hr at room temperature. We prepared antibody to large T antigen in house by culturing PAB 149 cells and obtaining the conditioned medium.
  • NGS normal goat serum
  • Preparations were then washed twice for 5 min with 0.1 M PBS.
  • Cells were incubated with secondary antibodies (goat-anti-mouse biotinylated at 1:500 from Vector Laboratories, Vectastain Elite ABC mouse IgG kit, PK-6102) diluted in 1% NGS for 30 min at room temperature.
  • Preparations are washed twice for 5 min with 0.1 M PBS.
  • Preparations are incubated in ABC reagent diluted 1:500 in 0.1 M PBS, pH 7.4 for 30 min at room temperature.
  • Cells are washed twice for 5 min in 0.1 M PBS, pH 7.4, then washed twice for 5 min in 0.1 M Tris, pH 7.6.
  • Cells are incubated in DAB (nickel intensification) for 5 min at room temperature. The DAB solution is removed, and cells are washed three to five times with dH2O. Cells are stored in 50% glycerol/50% 0.1 M PBS, pH 7.4.
  • DAB nickel intensification
  • the hollow fibers are fabricated from a polyether sulfone (PES) with an outside diameter of 720 m and a wall thickness of a 100 m (AKZO-Nobel Wüppertal, Germany). These fibers are described in U.S. Pat. Nos. 4,976,859 and 4,968,733, herein incorporated by reference.
  • the fiber may be chosen for its molecular weight cutoff. We sometimes use a PES#5 membrane which has a MWCO of about 280 kd. In other studies we use a PES#8 membrane which has a MWCO of about 90 kd.
  • the devices typically comprise:
  • the semipermeable membrane used typically has the following characteristics: Internal Diameter 500 + 30 m Wall Thickness 100 + 15 m Force at Break 100 + 15 cN Elongation at Break 44 + 10% Hydraulic Permeability 63 + 8 (ml/min m 2 mmHg) nMWCO (dextrans) 280 + 20 kd
  • the components of the device are commercially available.
  • the LCM glue is available from Ablestik Laboratories (Newark, Del.); Luxtrak Adhesives LCM23 and LCM24).
  • the tether material is available from Specialty Silicone Fabricators (Robles, Calif.). The tether dimensions are 0.79 mm OD ⁇ 0.43 mm ID ⁇ length 202 mm.
  • the morphology of the device is as follows: The inner surface has a permselective skin.
  • the wall has an open cell foam structure.
  • the outer surface has an open structure, with pores up to 1.5 m occupying 30+5% of the outer surface.
  • Fiber material is first cut into 5 cm long segments and the distal extremity of each segment sealed with a photopolymerized acrylic glue (LCM-25, ICI). Following sterilization with ethylene oxide and outgassing, the fiber segments are loaded with a suspension of between 10 4 -10 7 cells, either in a liquid medium, or a hydrogel matrix (e.g., a collagen solution (Zyderm®), alginate, agarose or chitosan) via a Hamilton syringe and a 25 gauge needle through an attached injection port. The proximal end of the capsule is sealed with the same acrylic glue.
  • the volume of the device contemplated in the human studies is approximately 15-181.
  • a silicone tether (Specialty Silicone Fabrication, Taunton, Mass.) (ID: 690 m; OD: 1.25 mm) is placed over the proximal end of the fiber allowing easy manipulation and retrieval of the device.
  • Transplantation was performed according to standard techniques.
  • Adult rats were anesthetized with sodium pentobarbitol (45 mg/kg, i.p.) And positioned in a Kopf stereotaxic instrument. A midline incision was made in the scalp and a hole drilled for the injection of cells.
  • Rats received implants of unmodified, undifferentiated human neural stem cells into the left striatum using a glass capillary attached to a 10 ⁇ l Hamilton syringe. Each animal received a total of about 250,000-500,000 cells in a total volume of 2 ⁇ l.
  • Cells were transplanted 1-2 days after passaging and the cell suspension was made up of undifferentiated stem cell clusters of 5-20 cells. Following implantation, the skin was sutured closed.
  • Approximately 300,000 neural stem cells were transplanted as small neurospheres into the adult rat striatum close to the lateral ventricle using standard techniques.
  • osmotic minipumps releasing either EGF (400 ng/day) or vehicle were also implanted in the striatum.
  • the rats received EGF over a period of 7 days at a flow rate of 0.5 ⁇ L/hr, resulting in the delivery of 2.8 ⁇ g EGF in total into the lateral ventricle of each animal.
  • Subsets of implanted rats were additionally immunosuppressed by i.p. cyclosporin injections (10 mg/kg/day).
  • the animals received injections of BrdU every three hours (120 mg/kg).
  • neural stem cells described in this invention are suitable for replacement, because only a structural function would be required by the cells.
  • Neural stem cells are implanted in the spinal cord of injured patients to prevent syrinx formation. Outcomes are measured preferably by MRI imaging. Clinical trial protocols have been written and could easily be modified to include the described neural stem cells.
  • Neural stem cells are obtained from ventral mesencephalic tissue from a human fetus aged 8 weeks following routine suction abortion which is collected into a sterile collection apparatus. A 2 ⁇ 4 ⁇ 1 mm piece of tissue is dissected and dissociated as in Example 2. Neural stem cells are then proliferated. Neural stem cell progeny are used for neurotransplantation into a blood-group matched host with a neurodegenerative disease. Surgery is performed using a BRW computed tomographic (CT) stereotaxic guide. The patient is given local anesthesia suppiemencea with intravenously administered midazolam. The patient undergoes CT scanning to establish the coordinates of the region to receive the transplant.
  • CT computed tomographic
  • the injection cannula consists of a 17-gauge stainless steel outer cannula with a 19-gauge inner stylet. This is inserted into the brain to the correct coordinates, then removed and replaced with a 19-gauge infusion cannula that has been preloaded with 30 ⁇ l of tissue suspension. The cells are slowly infused at a rate of 3 ⁇ l/min as the cannula is withdrawn. Multiple stereotactic needle passes are made throughout the area of interest, approximately 4 mm apart. The patient is examined by CT scan postoperatively for hemorrhage or edema. Neurological evaluations are performed at various post-operative intervals, as well as PET scans to determine metabolic activity of the implanted cells.
  • Neural stem cell progeny are propagated as described in Example 2.
  • the cells are then transfected using a calcium phosphate transfection technique.
  • the cells are mechanically dissociated into a single cell suspension and plated on tissue culture-treated dishes at 50% confluence (50,000-75,000 cells/cm 2 ) and allowed to attach overnight.
  • the modified calcium phosphate transfection procedure is performed as follows: DNA (15-25 ⁇ g) in sterile TE buffer (10 mM Tris, 0.25 mM EDTA, pH 7.5) diluted to 4401 with TE, and 60 ⁇ l of 2M CaCl 2 (pH to 5.8 with 1M HEPES buffer) is added to the DNA/TE buffer. A total of 500 ⁇ l of 2 ⁇ HeBS (HEPES-Buffered saline; 275 mM NaCl, 10 mM KCl, 1.4 mM Na 2 HPO 4 , 12 mM dextrose, 40 mM HEPES buffer powder, pH 6.92) is added dropwise to this mix. The mixture is allowed to stand at room temperature for 20 minutes.
  • 2 ⁇ HeBS HPES-Buffered saline; 275 mM NaCl, 10 mM KCl, 1.4 mM Na 2 HPO 4 , 12 mM dextrose, 40 mM HEPES buffer
  • the cells are washed briefly with 1 ⁇ HeBS and 1 ml of the calcium phosphate precipitated DNA solution is added to each plate, and the cells are incubated at 37° for 20 minutes. Following this incubation, 10 mls of complete medium is added to the cells, and the plates are placed in an incubator (37° C., 9.5% CO 2 ) for an additional 3-6 hours. The DNA and the medium are removed by aspiration at the end of the incubation period, and the cells are washed 3 times with complete growth medium and then returned to the incubator.
  • Cells proliferated as in Examples 2 are transfected with expression vectors containing the genes for the FGF-2 receptor or the NGF receptor.
  • Vector DNA containing the genes are diluted in 0.1 ⁇ TE (1 mM Tris pH 8.0, 0.1 mM EDTA) to a concentration of 40 ⁇ g/ml. 22 ⁇ l of the DNA is added to 250 ⁇ l of 2 ⁇ HBS (280 mM NaCl, 10 mM KCl, 1.5 mM Na 2 HPO 4 2H 2 O, 12 mM dextrose, 50 mM HEPES) in a disposable, sterile 5 ml plastic tube. 31 ⁇ l of 2M CaCl 2 is added slowly and the mixture is incubated for 30 minutes at room temperature.
  • 2 ⁇ HBS 280 mM NaCl, 10 mM KCl, 1.5 mM Na 2 HPO 4 2H 2 O, 12 mM dextrose, 50 mM HEPES
  • the cells are centrifuged at 800 g for 5 minutes at 4° C.
  • the cells are resuspended in 20 volumes of ice-cold PBS and divided into aliquots of 1 ⁇ 10 7 cells, which are again centrifuged.
  • Each aliquot of cells is resuspended in 1 ml of the DNA-CaCl 2 suspension, and incubated for 20 minutes at room temperature.
  • the cells are then diluted in growth medium and incubated for 6-24 hours at 37° C. in 5%-7% CO 2 .
  • the cells are again centrifuged, washed in PBS and returned to 10 ml of growth medium for 48 hours.
  • transfected neural stem cell progeny are transplanted into a human patient using the procedure described in Example 8 or Example 11, or are used for drug screening procedures as described in the example below.
  • Precursor cells were propagated as described in Example 2 and differentiated as described in Example 4. At the time of plating the cells, BDNF was added at a concentration of 10 ng/ml. At 3, 7, 14, and 21 days in vitro (DIV), cells were processed for indirect immunocytochemistry. BrdU labeling was used to monitor proliferation of the neural stem cells. The effects of BDNF on neurons, oligodendrocytes and astrocytes were assayed by probing the cultures with antibodies that recognize antigens found on neurons (MAP-2, NSE, NF), oligodendrocytes (O4, GalC, MBP) or astrocytes (GFAP). Cell survival was determined by counting the number of immunoreactive cells at each time point and morphological observations were made. BDNF significantly increased the differentiation and survival of neurons over the number observed under control conditions. Astrocyte and oligodendrocyte numbers were not significantly altered from control values.
  • BDNF BDNF-treated culture conditions
  • neurons tested positive for the presence of substance P and GABA As well as an increase in numbers, neurons grown in BDNF showed a dramatic increase in neurite extension and branching when compared with control examples.
  • BDNF BDNF-derived neurotrophic factor
  • NSE neurotrophic factor
  • NF oligodendrocytes
  • GFAP astrocytes
  • Exposure to BDNF resulted in a selective increase in the expression of c-fos in neuronal cells.
  • Cells treated with BDNF according to the methods described in Part A are processed for analysis of the expression of regulatory factors, FGF-R1 or other markers.
  • Chlorpromazine a drug widely used in the treatment of psychiatric illness, is used in concentrations ranging from 10 ng/ml to 1000 ng/ml in place of BDNF in Examples 14A to 14D above.
  • the effects of the drug at various concentrations on stem cell proliferation and on stem cell progeny differentiation and survival is monitored. Alterations in gene expression and electrophysiological properties of differentiated neurons are determined.

Abstract

The invention provides a cell culture including proliferating human neural stem cells with a doubling rate faster than thirty days. The invention also provides a cell culture media for proliferating mammalian neural cells including a standard defined culture medium, a carbohydrate source, a buffer, a source of hormones, one or more growth factors that stimulate the proliferation of neural stem cells, and LIF. The invention also provides a method for protecting, repairing or replacing damaged tissue comprising transplanting mammalian neural stem cells formed into neurospheres. The invention also provides a cell culture of differentiated human neural stem cells where the cells are glioblasts. The invention also provides a method of differentiating human neural stem cells in culture media.

Description

    TECHNICAL FIELD OF THE INVENTION
  • This invention relates to isolation of human central nervous system stem cells, and methods and media for proliferating, differentiating and transplanting them.
  • BACKGROUND OF THE INVENTION
  • During development of the central nervous system (“CNS”), multipotent precursor cells, also known as neural stem cells, proliferate, giving rise to transiently dividing progenitor cells that eventually differentiate into the cell types that compose the adult brain. Stem cells (from other tissues) have classically been defined as having the ability to self-renew (i.e., form more stem cells), to proliferate, and to differentiate into multiple different phenotypic lineages. In the case of neural stem cells this includes neurons, astrocytes and oligodendrocytes. For example, Potten and Loeffler (Development, 110:1001, 1990) define stem cells as “undifferentiated cells capable of a) proliferation, b) self-maintenance, c) the production of a large number of differentiated functional progeny, d) regenerating the tissue after injury, and e) a flexibility in the use of these options.”
  • These neural stem cells have been isolated from several mammalian species, including mice, rats, pigs and humans. See, e.g., WO 93/01275, WO 94/09119, WO 94/10292, WO 94/16718 and Cattaneo et al., Mol. Brain. Res., 42, pp. 161-66 (1996), all herein incorporated by reference.
  • Human CNS neural stem cells, like their rodent homologues, when maintained in a mitogen-containing (typically epidermal growth factor or epidermal growth factor plus basic fibroblast growth factor), serum-free culture medium, grow in suspension culture to form aggregates of cells known as “neurospheres”. In the prior art, human neural stem cells have doubling rates of about 30 days. See, e.g., Cattaneo et al., Mol. Brain. Res., 42, pp. 161-66 (1996). Upon removal of the mitogen(s) and provision of a substrate, the stem cells differentiate into neurons, astrocytes and oligodendrocytes. In the prior art, the majority of cells in the differentiated cell population have been identified as astrocytes, with very few neurons (<10%) being observed.
  • There remains a need to increase the rate of proliferation of neural stem cell cultures. There also remains a need to increase the number of neurons in the differentiated cell population. There further remains a need to improve the viability of neural stem cell grafts upon implantation into a host.
  • SUMMARY OF THE INVENTION
  • This invention provides novel human central nervous system stem cells, and methods and media for proliferating, differentiating and transplanting them. In one embodiment, this invention provides novel human stem cells with a doubling rate of between 5-10 days, as well as defined growth media for prolonged proliferation of human neural stem cells. In another embodiment, this invention provides a defined media for differentiation of human neural stem cells so as to enrich for neurons, oligodendrocytes, astrocytes, or a combination thereof. The invention also provides differentiated cell populations of human neural stem cells that provide previously unobtainable large numbers of neurons, as well as astrocytes and oligodendrocytes. This invention also provides novel methods for transplanting neural stem cells that improve the viability of the graft upon implantation in a host.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows a representation of spheres of proliferating 9FBr human neural stem cells (passage 6) derived from human forebrain tissue.
  • FIG. 2, Panel A, shows a growth curve for a human neural stem cell line designated 6.5Fbr cultured in (a) defined media containing EGF, FGF and leukemia inhibitory factor (“LIF”) (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds); Panel B shows a growth curve for a human neural stem cell line designated 9Fbr cultured in (a) defined media containing EGF, FGF and LIF (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds); Panel C shows a growth curve for a human neural stem cell line designated 9.5Fbr cultured in (a) defined media containing EGF, FGF and LIF (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds); Panel D shows a growth curve for a human neural stem cell line designated 10.5Fbr cultured in (a) defined media containing EGF, FGF and leukemia inhibitory factor (“LIF”) (shown as closed diamonds), and (b) the same media but without LIF (shown as open diamonds).
  • FIG. 3 shows a growth curve for a human neural stem cell line designated 9Fbr cultured in (a) defined media containing EGF and basic fibroblast growth factor (“bFGF”) (shown as open diamonds), and (b) defined media with EGF but without bFGF (shown as closed diamonds).
  • FIG. 4 shows a graph of cell number versus days in culture for an Mx-1 conditionally immortalized human glioblast line derived from a human neural stem cell line. The open squares denote growth in the presence of interferon, the closed diamonds denote growth in the absence of interferon.
  • DETAILED DESCRIPTION OF THE INVENTION
  • This invention relates to isolation, characterization, proliferation, differentiation and transplantation of CNS neural stem cells.
  • The neural stem cells described and claimed in the applications may be proliferated in suspension culture or in adherent culture. When the neural stem cells of this invention are proliferating as neurospheres, human nestin antibody may be used as a marker to identify undifferentiated cells. The proliferating cells show little GFAP staining and little β-tubulin staining (although some staining might be present due to diversity of cells within the spheres).
  • When differentiated, most of the cells lose their nestin positive immunoreactivity. In particular, antibodies specific for various neuronal or glial proteins may be employed to identify the phenotypic properties of the differentiated cells. Neurons may be identified using antibodies to neuron specific enolase (“NSE”), neurofilament, tau, beta-tubulin, or other known neuronal markers. Astrocytes may be identified using antibodies to glial fibrillary acidic protein (“GFAP”), or other known astrocytic markers. Oligodendrocytes may be identified using antibodies to galactocerebroside, O4, myelin basic protein (“MBP”) or other known oligodendrocytic markers. Glial cells in general may be identified by staining with antibodies, such as the M2 antibody, or other known glial markers.
  • In one embodiment the invention provides novel human CNS stem cells isolated from the forebrain. We have isolated 4 neural stem cell lines from human forebrain, all of which exhibit neural stem cell properties; namely, the cells are self renewing, the cells proliferate for long periods in mitogen containing serum free medium, and the cells, when differentiated, comprise a cell population of neurons, astrocytes and oligodendrocytes. These cells are capable of doubling every 5-10 days, in contrast with the prior art diencephalon-derived human neural stem cells. Reported proliferation rates of diencephalon-derived human neural stem cells approximate one doubling every 30 days. See Cattaneo et al., Mol. Brain. Res., 42, pp. 161-66 (1996).
  • Any suitable tissue source may be used to derive the neural stem cells of this invention. Neural stem cells can be induced to proliferate and differentiate either by culturing the cells in suspension or on an adherent substrate. See, e.g., U.S. Pat. No. 5,750,376 and U.S. Pat. No. 5,753,506 (both incorporated herein by reference in their entirety), and prior art medium described therein. Both allografts and autografts are contemplated for transplantation purposes.
  • This invention also provides a novel growth media for proliferation of neural stem cells. Provided herein is a serum-free or serum-depleted culture medium for the short term and long term proliferation of neural stem cells.
  • A number of serum-free or serum-depleted culture media have been developed due to the undesirable effects of serum which can lead to inconsistent culturing results. See, e.g., WO 95/00632 (incorporated herein by reference), and prior art medium described therein.
  • Prior to development of the novel media described herein, neural stem cells have been cultured in serum-free media containing epidermal growth factor (“EGF”) or an analog of EGF, such as amphiregulin or transforming growth factor alpha (“TGF-α”), as the mitogen for proliferation. See, e.g., WO 93/01275, WO 94/16718, both incorporated herein by reference. Further, basic fibroblast growth factor (“bFGF”) has been used, either alone, or in combination with EGF, to enhance long term neural stem cell survival.
  • The improved medium according to this invention, which contains leukemia inhibitory factor (“LIF”), markedly and unexpectedly increases the rate of proliferation of neural stem cells, particularly human neural stem cells.
  • We have compared growth rates of the forebrain-derived stem cells described herein in the presence and absence of LIF; unexpectedly we have found that LIF dramatically increases the rate of cellular proliferation in almost all cases.
  • The medium according to this invention comprises cell viability and cell proliferation effective amounts of the following components:
      • (a) a standard culture medium being serum-free (containing 0-0.49% serum) or serum-depleted (containing 0.5-5.0% serum), known as a “defined” culture medium, such as Iscove's modified Dulbecco's medium (“IMDM”), RPM1, DMEM, Fischer's, alpha medium, Leibovitz's, L-15, NCTC, F-10, F-12, MEM and McCoy's;
      • (b) a suitable carbohydrate source, such as glucose;
      • (c) a buffer such as MOPS, HEPES or Tris, preferably HEPES;
      • (d) a source of hormones including insulin, transferrin, progesterone, selenium, and putrescine;
      • (e) one or more growth factors that stimulate proliferation of neural stem cells, such as EGF, bFGF, PDGF, NGF, and analogs, derivatives and/or combinations thereof, preferably EGF and bFGF in combination;
      • (f) LIF
  • Standard culture media typically contains a variety of essential components required for cell viability, including inorganic salts, carbohydrates, hormones, essential amino acids, vitamins, and the like. We prefer DMEM or F-12 as the standard culture medium, most preferably a 50/50 mixture of DMEM and F-12. Both media are commercially available (DMEM-Gibco 12100-046; F-12-Gibco 21700-075). A premixed formulation is also commercially available (N2-Gibco 17502-030). It is advantageous to provide additional glutamine, preferably at about 2 mM. It is also advantageous to provide heparin in the culture medium. Preferably, the conditions for culturing should be as close to physiological as possible. The pH of the culture medium is typically between 6-8, preferably about 7, most preferably about 7.4. Cells are typically cultured between 30-40° C., preferably between 32-38° C., most preferably between 35-37° C. Cells are preferably grown in 5% CO2. Cells are preferably grown in suspension culture.
  • In one exemplary embodiment, the neural stem cell culture comprises the following components in the indicated concentrations:
    Component Final Concentration
    50/50 mix of DMEM/F-12 0.5-2.0×, preferably 1×
    glucose 0.2-1.0%, preferably 0.6% w/v
    glutamine 0.1-10 mM, preferably 2 mM
    NaHCO3 0.1-10 mM, preferably 3 mM
    HEPES 0.1-10 mM, preferably 5 mM
    apo-human transferrin (Sigma 1-1000 μg/ml, preferably 100 μg/ml
    T-2252)
    human insulin (Sigma I-2767) 1-100, preferably 25 μg/ml
    putrescine (Sigma P-7505) 1-500, preferably 60 μM
    selenium (Sigma S-9133) 1-100, preferably 30 nM
    progesterone (Sigma P-6149) 1-100, preferably 20 nM
    human EGF (Gibco 13247-010) 0.2-200, preferably 20 ng/ml
    human bFGF (Gibco 13256-029) 0.2-200, preferably 20 ng/ml
    human LIF (R&D Systems 250-L) 0.1-500, preferably 10 ng/ml
    heparin (Sigma H-3149) 0.1-50, preferably 2 μg/ml
    CO2 preferably 5%

    Serum albumin may also be present in the instant culture medium—although the present medium is generally serum-depleted or serum-free (preferably serum-free), certain serum components which are chemically well defined and highly purified (>95%), such as serum albumin, may be included.
  • The human neural stem cells described herein may be cryopreserved according to routine procedures. We prefer cryopreserving about one to ten million cells in “freeze” medium which consists of proliferation medium (absent the growth factor mitogens), 10% BSA (Sigma A3059) and 7.5% DMSO. Cells are centrifuged. Growth medium is aspirated and replaced with freeze medium. Cells are resuspended gently as spheres, not as dissociated cells. Cells are slowly frozen, by, e.g., placing in a container at −80° C. Cells are thawed by swirling in a 37° C. bath, resuspended in fresh proliferation medium, and grown as usual.
  • In another embodiment, this invention provides a differentiated cell culture containing previously unobtainable large numbers of neurons, as well as astrocytes and oligodendrocytes. In the prior art, typically the differentiated human diencephalon-derived neural stem cell cultures formed very few neurons (i.e., less than 5-10%). According to this methodology, we have routinely achieved neuron concentrations of between 20% and 35% (and much higher in other cases) in differentiated human forebrain-derived neural stem cell cultures. This is highly advantageous as it permits enrichment of the neuronal population prior to implantation in the host in disease indications where neuronal function has been impaired or lost.
  • Further, according to the methods of this invention, we have achieved differentiated neural stem cell cultures that are highly enriched in GABA-ergic neurons. Such GABA-ergic neuron enriched cell cultures are particularly advantageous in the potential therapy of excitotoxic neurodegenerative disorders, such as Huntington's disease or epilepsy.
  • In order to identify the cellular phenotype either during proliferation or differentiation of the neural stem cells, various cell surface or intracellular markers may be used.
  • When the neural stem cells of this invention are proliferating as neurospheres, we contemplate using human nestin antibody as a marker to identify undifferentiated cells. The proliferating cells should show little GFAP staining and little β-tubulin staining (although some staining might be present due to diversity of cells within the spheres).
  • When differentiated, most of the cells lose their nestin positive immunoreactivity. In particular, antibodies specific for various neuronal or glial proteins may be employed to identify the phenotypic properties of the differentiated cells. Neurons may be identified using antibodies to neuron specific enolase (“NSE”), neurofilament, tau, β-tubulin, or other known neuronal markers. Astrocytes may be identified using antibodies to glial fibrillary acidic protein (“GFAP”), or other known astrocytic markers. Oligodendrocytes may be identified using antibodies to galactocerebroside, O4, myelin basic protein (“MBP”) or other known oligodendrocytic markers.
  • It is also possible to identify cell phenotypes by identifying compounds characteristically produced by those phenotypes. For example, it is possible to identify neurons by the production of neurotransmitters such as acetylcholine, dopamine, epinephrine, norepinephrine, and the like.
  • Specific neuronal phenotypes can be identified according to the specific products produced by those neurons. For example, GABA-ergic neurons may be identified by their production of glutamic acid decarboxylase (“GAD”) or GABA. Dopaminergic neurons may be identified by their production of dopa decarboxylase (“DDC”), dopamine or tyrosine hydroxylase (“TH”). Cholinergic neurons may be identified by their production of choline acetyltransferase (“ChAT”). Hippocampal neurons may be identified by staining with NeuN. It will be appreciated that any suitable known marker for identifying specific neuronal phenotypes may be used.
  • The human neural stem cells described herein can be genetically engineered or modified according to known methodology. The term “genetic modification” refers to the stable or transient alteration of the genotype of a cell by intentional introduction of exogenous DNA. DNA may be synthetic, or naturally derived, and may contain genes, portions of genes, or other useful DNA sequences. The term “genetic modification” is not meant to include naturally occurring alterations such as that which occurs through natural viral activity, natural genetic recombination, or the like.
  • A gene of interest (i.e., a gene that encodes a biologically active molecule) can be inserted into a cloning site of a suitable expression vector by using standard techniques. These techniques are well known to those skilled in the art. See, e.g., WO 94/16718, incorporated herein by reference.
  • The expression vector containing the gene of interest may then be used to transfect the desired cell line. Standard transfection techniques such as calcium phosphate co-precipitation, DEAE-dextran transfection, electroporation, biolistics, or viral transfection may be utilized. Commercially available mammalian transfection kits may be purchased from e.g., Stratagene. Human adenoviral transfection may be accomplished as described in Berg et al. Exp. Cell Res., 192, pp. (1991). Similarly, lipofectamine-based transfection may be accomplished as described in Cattaneo, Mol. Brain. Res., 42, pp. 161-66 (1996).
  • A wide variety of host/expression vector combinations may be used to express a gene encoding a biologically active molecule of interest. See, e.g., U.S. Pat. No. 5,545,723, herein incorporated by reference, for suitable cell-based production expression vectors.
  • Increased expression of the biologically active molecule can be achieved by increasing or amplifying the transgene copy number using amplification methods well known in the art. Such amplification methods include, e.g., DHFR amplification (see, e.g., Kaufman et al., U.S. Pat. No. 4,470,461) or glutamine synthetase (“GS”) amplification (see, e.g., U.S. Pat. No. 5,122,464, and European published application EP 338,841), all herein incorporated by reference.
  • In another embodiment, the genetically modified neural stem cells are derived from transgenic animals.
  • When the neural stem cells are genetic modified for the production of a biologically active substance, the substance will preferably be useful for the treatment of a CNS disorder. We contemplate genetically modified neural stem cells that are capable of secreting a therapeutically effective biologically active molecule in patients. We also contemplate producing a biologically active molecule with growth or trophic effect on the transplanted neural stem cells. We further contemplate inducing differentiation of the cells towards neural cell lineages. The genetically modified neural stem cells thus provide cell-based delivery of biological agents of therapeutic value.
  • The neural stem cells described herein, and their differentiated progeny may be immortalized or conditionally immortalized using known techniques. We prefer conditional immortalization of stem cells, and most preferably conditional immortalization of their differentiated progeny. Among the conditional immortalization techniques contemplated are Tet-conditional immortalization (see WO 96/31242, incorporated herein by reference), and Mx-1 conditional immortalization (see WO 96/02646, incorporated herein by reference).
  • This invention also provides methods for differentiating neural stem cells to yield cell cultures enriched with neurons to a degree previously unobtainable. According to one protocol, the proliferating neurospheres are induced to differentiate by removal of the growth factor mitogens and LIF, and provision of 1% serum, a substrate and a source of ionic charges (e.g., glass cover slip covered with poly-ornithine or extracellular matrix components). The preferred base medium for this differentiation protocol, excepting the growth factor mitogens and LIF, is otherwise the same as the proliferation medium. This differentiation protocol produces a cell culture enriched in neurons. According to this protocol, we have routinely achieved neuron concentrations of between 20% and 35% in differentiated human forebrain-derived neural stem cell cultures.
  • According to a second protocol, the proliferating neurospheres are induced to differentiate by removal of the growth factor mitogens, and provision of 1% serum, a substrate and a source of ionic charges (e.g., glass cover slip covered with poly-ornithine or extracellular matrix components), as well as a mixture of growth factors including PDGF, CNTF, IGF-1, LIF, forskolin, T-3 and NT-3. The cocktail of growth factors may be added at the same time as the neurospheres are removed from the proliferation medium, or may be added to the proliferation medium and the cells pre-incubated with the mixture prior to removal from the mitogens. This protocol produces a cell culture highly enriched in neurons and enriched in oligodendrocytes. According to this protocol, we have routinely achieved neuron concentrations of higher than 35% in differentiated human forebrain-derived neural stem cell cultures.
  • The presence of bFGF in the proliferation media unexpectedly inhibits oligodendrocyte differentiation capability. bFGF is trophic for the oligodendrocyte precursor cell line. Oligodendrocytes are induced under differentiation conditions when passaged with EGF and LIF in proliferating media, without bFGF.
  • The human stem cells of this invention have numerous uses, including for drug screening, diagnostics, genomics and transplantation. Stem cells can be induced to differentiate into the neural cell type of choice using the appropriate media described in this invention. The drug to be tested can be added prior to differentiation to test for developmental inhibition, or added post-differentiation to monitor neural cell-type specific reactions.
  • The cells of this invention may be transplanted “naked” into patients according to conventional techniques, into the CNS, as described for example, in U.S. Pat. Nos. 5,082,670 and 5,618,531, each incorporated herein by reference, or into any other suitable site in the body.
  • In one embodiment, the human stem cells are transplanted directly into the CNS. Parenchymal and intrathecal sites are contemplated. It will be appreciated that the exact location in the CNS will vary according to the disease state.
  • Implanted cells may be labeled with bromodeoxyuridine (BrdU) prior to transplantation. We have observed in various experiments that cells double stained for a neural cell marker and BrdU in the various grafts indicate differentiation of BrdU stained stem cells into the appropriate differentiated neural cell type (see Example 9). Transplantation of human forebrain derived neural stem cells to the hippocampus produced neurons that were predominantly NeuN staining but GABA negative. The NeuN antibody is known to stain neurons of the hippocampus. GABA-ergic neurons were formed when these same cell lines were transplanted into the striatum. Thus, transplanted cells respond to environmental clues in both the adult and the neonatal brain.
  • According to one aspect of this invention, provided herein is methodology for improving the viability of transplanted human neural stem cells. In particular, we have discovered that graft viability improves if the transplanted neural stem cells are allowed to aggregate, or to form neurospheres prior to implantation, as compared to transplantation of dissociated single cell suspensions. We prefer transplanting small sized neurospheres, approximately 10-500 μm in diameter, preferably 40-50 μm in diameter. Alternatively, we prefer spheres containing about 5-100, preferably 5-20 cells per sphere. We contemplate transplanting at a density of about 10,000-1,000,000 cells per μl, preferably 25,000-500,000 cells per μl.
  • The cells may also be encapsulated and used to deliver biologically active molecules, according to known encapsulation technologies, including microencapsulation (see, e.g., U.S. Pat. Nos. 4,352,883; 4,353,888; and 5,084,350, herein incorporated by reference), (b) macroencapsulation (see, e.g., U.S. Pat. Nos. 5,284,761, 5,158,881, 4,976,859 and 4,968,733 and published PCT patent applications WO92/19195, WO 95/05452, each incorporated herein by reference).
  • If the human neural stem cells are encapsulated, we prefer macroencapsulation, as described in U.S. Pat. Nos. 5,284,761; 5,158,881; 4,976,859; 4,968,733; 5,800,828 and published PCT patent application WO 95/05452, each incorporated herein by reference. Cell number in the devices can be varied; preferably each device contains between 103-109 cells, most preferably 105 to 107 cells. A large number of macroencapsulation devices may be implanted in the patient; we prefer between one to 10 devices.
  • In addition, we also contemplate “naked” transplantation of human stem cells in combination with a capsular device wherein the capsular device secretes a biologically active molecule that is therapeutically effective in the patient or that produces a biologically active molecule that has a growth or trophic effect on the transplanted neural stem cells, or that induces differentiation of the neural stem cells towards a particular phenotypic lineage.
  • The cells and methods of this invention may be useful in the treatment of various neurodegenerative diseases and other disorders. It is contemplated that the cells will replace diseased, damaged or lost tissue in the host. Alternatively, the transplanted tissue may augment the function of the endogenous affected host tissue. The transplanted neural stem cells may also be genetically modified to provide a therapeutically effective biologically active molecule.
  • Excitotoxicity has been implicated in a variety of pathological conditions including epilepsy, stroke, ischemia, and neurodegenerative diseases such as Huntington's disease, Parkinson's disease and Alzheimer's disease. Accordingly, neural stem cells may provide one means of preventing or replacing the cell loss and associated behavioral abnormalities of these disorders. Neural stem cells may replace cerebellar neurons lost in cerebellar ataxia, with clinical outcomes readily measurable by methods known in the medical arts.
  • Huntington's disease (HD) is an autosomal dominant neurodegenerative disease characterized by a relentlessly progressive movement disorder with devastating psychiatric and cognitive deterioration. HD is associated with a consistent and severe atrophy of the neostriatum which is related to a marked loss of the GABAergic medium-sized spiny projection neurons, the major output neurons of the striatum. Intrastriatal injections of excitotoxins such as quinolinic acid (QA) mimic the pattern of selective neuronal vulnerability seen in HD. QA lesions result in motor and cognitive deficits which are among the major symptoms seen in HD. Thus, intrastriatal injections of QA have become a useful model of HD and can serve to evaluate novel therapeutic strategies aimed at preventing, attenuating, or reversing neuroanatomical and behavioral changes associated with HD. Because GABA-ergic neurons are characteristically lost in Huntington's disease, we contemplate treatment of Huntington's patients by transplantation of cell cultures enriched in GABA-ergic neurons derived according to the methods of this invention.
  • Epilepsy is also associated with excitotoxicity. Accordingly, GABA-ergic neurons derived according to this invention are contemplated for transplantation into patients suffering from epilepsy.
  • We also contemplate use of the cells of this invention in the treatment of various demyelinating and dysmyelinating disorders, such as Pelizaeus-Merzbacher disease, multiple sclerosis, various leukodystrophies, post-traumatic demyelination, and cerebrovascular (CVS) accidents, as well as various neuritis and neuropathies, particularly of the eye. We contemplate using cell cultures enriched in oligodendrocytes or oligodendrocyte precursor or progenitors, such cultures prepared and transplanted according to this invention to promote remyelination of demyelinated areas in the host.
  • We also contemplate use of the cells of this invention in the treatment of various acute and chronic pains, as well as for certain nerve regeneration applications (such as spinal cord injury). We also contemplate use of human stem cells for use in sparing or sprouting of photoreceptors in the eye.
  • The cells and methods of this invention are intended for use in a mammalian host, recipient, patient, subject or individual, preferably a primate, most preferably a human.
  • The following examples are provided for illustrative purposes only, and are not intended to be limiting.
  • EXAMPLES Example 1 Media for Proliferating Neural Stem Cells
  • Proliferation medium was prepared with the following components in the indicated concentrations:
    Component Final Concentration
    50/50 mix of DMEM/F-12
    glucose 0.6% w/v
    glutamine
    2 mM
    NaHCO3 3 mM
    HEPES 5 mM
    apo-human transferrin (Sigma T-2252) 100 μg/ml
    human insulin (Sigma I-2767) 25 μg/ml
    putrescine (Sigma P-7505) 60 μM
    selenium (Sigma S-9133) 30 nM
    progesterone (Sigma P-6149) 20 nM
    human EGF (Gibco 13247-010) 20 ng/ml
    human bFGF (Gibco 13256-029) 20 ng/ml
    human LIF (R&D Systems 250-L) 10 ng/ml
    heparin (Sigma H-3149) 2 μg/ml
  • Example 2 Isolation of Human CNS Neural Stem Cells
  • Sample tissue from human embryonic forebrain was collected and dissected in Sweden and kindly provided by Huddinje Sjukhus. Blood samples from the donors were sent for viral testing. Dissections were performed in saline and the selected tissue was placed directly into proliferation medium (as described in Example 1). Tissue was stored at 4° C. until dissociated. The tissue was dissociated using a standard glass homogenizer, without the presence of any digesting enzymes. The dissociated cells were counted and seeded into flasks containing proliferation medium. After 5-7 days, the contents of the flasks are centrifuged at 1000 rpm for 2 min. The supernatant was aspirated and the pellet resuspended in 200 μl of proliferation medium. The cell clusters were triturated using a P200 pipetman about 100 times to break up the clusters. Cells were reseeded at 75,000-100,000 cells/ml into proliferation medium. Cells were passaged every 6-21 days depending upon the mitogens used and the seeding density. Typically these cells incorporate BrdU, indicative of cell proliferation. For T75 flask cultures (initial volume 20 ml), cells are “fed” 3 times weekly by addition of 5 ml of proliferation medium. We prefer Nunc flasks for culturing.
  • Nestin Staining for Proliferating Neurospheres
  • We stained for nestin (a measure of proliferating neurospheres) as follows. Cells were fixed for 20 min at room temperature with 4% paraformaldehyde. Cells were washed twice for 5 min with 0.1 M PBS, pH 7.4. Cells were permeabilized for 2 min with 100% EtOH. The cells were then washed twice for 5 min with 0.1 M PBS. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4 and 1% Triton X-100 (Sigma X-100) for 1 hr at room temperature with gentle shaking. Cells were incubated with primary antibodies to human nestin (from Dr. Lars Wahlberg, Karolinska, Sweden, rabbit polyclonal used at 1:500) diluted in 1% NGS and 1% Triton X-100 for 2 hr at room temperature. Preparations were then washed twice for 5 min with 0.1 M PBS. Cells were incubated with secondary antibodies (pool of GAM/FITC used at 1:128, Sigma F-0257; GAR/TRITC used at 1:80, Sigma T-5268) diluted in 1% NGS and 1% Triton X-100 for 30 min at room temperature in the dark. Preparations are washed twice for 5 min with 0.1 M PBS in the dark. Preparations are mounted onto slides face down with mounting medium (Vectashield Mounting Medium, Vector Labs., H-1000) and stored at 4° C.
  • FIG. 1 shows a picture of proliferating spheres (here called “neurospheres”) of human forebrain derived neural stem cells. We evaluated proliferation of 4 lines of human forebrain derived neural stem cells in proliferation medium as described above with LIF present of absent.
  • As FIG. 2 shows, in three of the four lines (6.5 Fbr, 9Fbr, and 10.5FBr), LIF significantly increased the rate of cell proliferation. The effect of LIF was most pronounced after about 60 days in vitro.
  • We also evaluated the effect of bFGF on the rate of proliferation of human forebrain-derived neural stem cells. As FIG. 3 shows, in the presence of bFGF, the stem cells proliferation was significantly enhanced.
  • Example 3 Differentiation of Human Neural Stem Cells
  • In a first differentiation protocol, the proliferating neurospheres were induced to differentiate by removal of the growth factor mitogens and LIF, and provision of 1% serum, a substrate and a source of ionic charges (e.g., glass cover slip covered with poly-ornithine).
  • The staining protocol for neurons, astrocytes and oligodendrocytes was as follows:
  • β-Tubulin Staining for Neurons
  • Cells were fixed for 20 min at room temperature with 4% paraformaldehyde. Cells were washed twice for 5 min with 0.1 M PBS, pH 7.4. Cells were permeabilized for 2 min with 100% EtOH. The cells were then washed twice for 5 min with 0.1 M PBS. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4. Cells were incubated with primary antibodies to β-tubulin (Sigma T-8660, mouse monoclonal; used at 1:1,000) diluted in 1% NGS for 2 hr at room temperature. Preparations were then washed twice for 5 min with 0.1 M PBS. Cells were incubated with secondary antibodies (pool of GAM/FITC used at 1:128, Sigma F-0257; GAR/TRITC used at 1:80, Sigma T-5268) diluted in 1% NGS for 30 min at room temperature in the dark. Preparations are washed twice for 5 min with 0.1 M PBS in the dark. Preparations are mounted onto slides face down with mounting medium (Vectashield Mounting Medium, Vector Labs., H-1000) and stored at 4° C.
  • In some instances we also stain with DAPI (a nuclear stain), as follows. Coverslips prepared as above are washed with DAPI solution (diluted 1:1000 in 100% MeOH, Boehringer Mannheim, #236 276). Coverslips are incubated in DAPI solution for 15 min at 37° C.
  • O4 Staining for Oligodendrocytes
  • Cells were fixed for 10 min at room temperature with 4% paraformaldehyde. Cells were washed three times for 5 min with 0.1 M PBS, pH 7.4. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4. Cells were incubated with primary antibodies to O4 (Boehringer Mannheim #1518 925, mouse monoclonal; used at 1:25) diluted in 1% NGS for 2 hr at room temperature. Preparations were then washed twice for 5 min with 0.1 M PBS. Cells were incubated with secondary antibodies, and further processed as described above for β-tubulin.
  • GFAP Staining for Astrocytes
  • Cells were fixed for 20 min at room temperature with 4% paraformaldehyde. Cells were washed twice for 5 min with 0.1 M PBS, pH 7.4. Cells were permeabilized for 2 min with 100% EtOH. The cells were then washed twice for 5 min with 0.1 M PBS. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4. Cells were incubated with primary antibodies to GFAP (DAKO Z 334, rabbit polyclonal; used at 1:500) diluted in 1% NGS for 2 hr at room temperature. Preparations were then washed twice for 5 min with 0.1 M PBS. Cells were incubated with secondary antibodies, and further processed as described above for β-tubulin.
  • This differentiation protocol produced cell cultures enriched in neurons as follows:
  • Cell Line Passage % GFAP Positive % β-tubulin positive % of neurons that are GABA positive
    6.5FBr 5 15 37 20
    9FBr 7 52 20 35
    10.5FBr  5 50 28 50
  • We also evaluated the ability of a single cell line to differentiate consistently as the culture aged (i.e., at different passages), using the above differentiation protocol. The data are as follows:
  • Cell Line Passage % GFAP Positive %β-tubulin positive % of neurons that are GABA positive
    9FBr 7 53 20.4 ND
    9FBr 9 ND 20.3 34.5
    9FBr 15 62 17.9 37.9

    We conclude from these data that cells will follow reproducible differentiation patterns irrespective of passage number or culture age.
  • Example 4 Differentiation of Human Neural Stem Cells
  • In a second differentiation protocol, the proliferating neurospheres were induced to differentiate by removal of the growth factor mitogens and LIF, and provision of 1% serum, a substrate (e.g., glass cover slip or extracellular matrix components), a source of ionic charges (e.g., poly-ornithine) as well as a mixture of growth factors including 10 ng/ml PDGF A/B, 10 ng/ml CNTF, 10 ng/ml IGF-1, 10 μM forskolin, 30 ng/ml T3, 10 ng/ml LIF and 1 ng/ml NT-3. This differentiation protocol produced cell cultures highly enriched in neurons (i.e., greater than 35% of the differentiated cell culture) and enriched in oligodendrocytes.
  • Example 5 Differentiation of Human Neural Stem Cells
  • In a third differentiation protocol, cell suspensions were initially cultured in a cocktail of hbFGF, EGF, and LIF, were then placed into altered growth media containing 20 ng/mL hEGF (GIBCO) and 10 ng/mL human leukemia inhibitory factor (hLIF) (R&D Systems), but without hbFGF. The cells initially grew significantly more slowly than the cultures that also contained hbFGF (see FIG. 3). Nonetheless, the cells continued to grow and were passaged as many as 22 times. Stem cells were removed from growth medium and induced to differentiate by plating on poly-ornithine coated glass coverslips in differentiation medium supplemented with a growth factor cocktail (hPDGF A/B, hCNTF, hGF-1, forskolin, T3 and hNT-3). Surprisingly, GalC immunoreactivity was seen in these differentiated cultures at levels that far exceeded the number of O4 positive cells seen in the growth factor induction protocol described in Example 4.
  • Hence, this protocol produced differentiated cell cultures enrichment in oligodendrocytes. Neurons were only occasionally seen, had small processes, and appeared quite immature.
  • Example 6 Genetic Modification
  • We have conditionally immortalized a glioblast cell line derived from the human neural stem cells described herein, using the Mx-1 system described in WO 96/02646. In the Mx-1 system, the Mx-1 promoter drives expression of the SV40 large T antigen. The Mx-1 promoter is induced by interferon. When induced, large T is expressed, and quiescent cells proliferate.
  • Human glioblasts were derived from human forebrain neural stem cells as follows. Proliferating human neurospheres were removed from proliferation medium and plated onto poly-ornithine plastic (24 well plate) in a mixture of N2 with the mitogens EGF, bFGF and LIF, as well as 0.5% FBS. 0.5 ml of N2 medium and 1% FBS was added. The cells were incubated overnight. The cells were then transfected with p318 (a plasmid containing the Mx-1 promoter operably linked to the SV 40 large T antigen) using Invitrogen lipid kit (lipids 4 and 6). The transfection solution contained 6 μl/ml of lipid and 4 μl/ml DNA in optiMEM medium. The cells were incubated in transfection solution for 5 hours. The transfection solution was removed and cells placed into N2 and 1% FBS and 500 U/ml A/D interferon. The cells were fed twice a week. After ten weeks cells were assayed for large T antigen expression. The cells showed robust T antigen staining at this time. As FIG. 4 shows, cell number was higher in the presence of interferon than in the absence of interferon.
  • Large T expression was monitored using immunocytochemistry as follows. Cells were fixed for 20 min at room temperature with 4% paraformaldehyde. Cells were washed twice for 5 min with 0.1 M PBS, pH 7.4. Cells were permeabilized for 2 min with 100% EtOH. The cells were then washed twice for 5 min with 0.1 M PBS. Cell preparations were blocked for 1 hr at room temperature in 5% normal goat serum (“NGS”) diluted in 0.1M PBS, pH 7.4. Cells were incubated with primary antibodies to large T antigen (used at 1:10) diluted in 1% NGS for 2 hr at room temperature. We prepared antibody to large T antigen in house by culturing PAB 149 cells and obtaining the conditioned medium. Preparations were then washed twice for 5 min with 0.1 M PBS. Cells were incubated with secondary antibodies (goat-anti-mouse biotinylated at 1:500 from Vector Laboratories, Vectastain Elite ABC mouse IgG kit, PK-6102) diluted in 1% NGS for 30 min at room temperature. Preparations are washed twice for 5 min with 0.1 M PBS. Preparations are incubated in ABC reagent diluted 1:500 in 0.1 M PBS, pH 7.4 for 30 min at room temperature. Cells are washed twice for 5 min in 0.1 M PBS, pH 7.4, then washed twice for 5 min in 0.1 M Tris, pH 7.6. Cells are incubated in DAB (nickel intensification) for 5 min at room temperature. The DAB solution is removed, and cells are washed three to five times with dH2O. Cells are stored in 50% glycerol/50% 0.1 M PBS, pH 7.4.
  • Example 7 Encapsulation
  • If the human neural stem cells are encapsulated, then the following procedure may be used:
  • The hollow fibers are fabricated from a polyether sulfone (PES) with an outside diameter of 720 m and a wall thickness of a 100 m (AKZO-Nobel Wüppertal, Germany). These fibers are described in U.S. Pat. Nos. 4,976,859 and 4,968,733, herein incorporated by reference. The fiber may be chosen for its molecular weight cutoff. We sometimes use a PES#5 membrane which has a MWCO of about 280 kd. In other studies we use a PES#8 membrane which has a MWCO of about 90 kd.
  • The devices typically comprise:
      • 1) a semipermeable poly (ether sulfone) hollow fiber membrane fabricated by AKZO Nobel Faser AG;
      • 2) a hub membrane segment;
      • 3) a light cured methacrylate (LCM) resin leading end; and
      • 4) a silicone tether.
  • The semipermeable membrane used typically has the following characteristics:
    Internal Diameter 500 + 30 m
    Wall Thickness 100 + 15 m
    Force at Break 100 + 15 cN
    Elongation at Break  44 + 10%
    Hydraulic Permeability  63 + 8 (ml/min m2 mmHg)
    nMWCO (dextrans) 280 + 20 kd

    The components of the device are commercially available. The LCM glue is available from Ablestik Laboratories (Newark, Del.); Luxtrak Adhesives LCM23 and LCM24). The tether material is available from Specialty Silicone Fabricators (Robles, Calif.). The tether dimensions are 0.79 mm OD×0.43 mm ID×length 202 mm. The morphology of the device is as follows: The inner surface has a permselective skin. The wall has an open cell foam structure. The outer surface has an open structure, with pores up to 1.5 m occupying 30+5% of the outer surface.
  • Fiber material is first cut into 5 cm long segments and the distal extremity of each segment sealed with a photopolymerized acrylic glue (LCM-25, ICI). Following sterilization with ethylene oxide and outgassing, the fiber segments are loaded with a suspension of between 104-107 cells, either in a liquid medium, or a hydrogel matrix (e.g., a collagen solution (Zyderm®), alginate, agarose or chitosan) via a Hamilton syringe and a 25 gauge needle through an attached injection port. The proximal end of the capsule is sealed with the same acrylic glue. The volume of the device contemplated in the human studies is approximately 15-181.
  • A silicone tether (Specialty Silicone Fabrication, Taunton, Mass.) (ID: 690 m; OD: 1.25 mm) is placed over the proximal end of the fiber allowing easy manipulation and retrieval of the device.
  • Example 8 Transplantation of Neural Stem Cells
  • We have transplanted human neural stem cells into rat brain and assessed graft viability, integration, phenotypic fate of the grafted cells, as well as behavioral changes associated with the grafted cells in lesioned animals.
  • Transplantation was performed according to standard techniques. Adult rats were anesthetized with sodium pentobarbitol (45 mg/kg, i.p.) And positioned in a Kopf stereotaxic instrument. A midline incision was made in the scalp and a hole drilled for the injection of cells. Rats received implants of unmodified, undifferentiated human neural stem cells into the left striatum using a glass capillary attached to a 10 μl Hamilton syringe. Each animal received a total of about 250,000-500,000 cells in a total volume of 2 μl. Cells were transplanted 1-2 days after passaging and the cell suspension was made up of undifferentiated stem cell clusters of 5-20 cells. Following implantation, the skin was sutured closed.
  • Animals were behaviorally tested and then sacrificed for histological analysis.
  • Example 9 Intraventricular EGF Delivery with Transplantation of Neural Stem Cells
  • Approximately 300,000 neural stem cells were transplanted as small neurospheres into the adult rat striatum close to the lateral ventricle using standard techniques. During the same surgery session, osmotic minipumps releasing either EGF (400 ng/day) or vehicle were also implanted in the striatum. The rats received EGF over a period of 7 days at a flow rate of 0.5 μL/hr, resulting in the delivery of 2.8 μg EGF in total into the lateral ventricle of each animal. Subsets of implanted rats were additionally immunosuppressed by i.p. cyclosporin injections (10 mg/kg/day). During the last 16 hours of pump infusion, the animals received injections of BrdU every three hours (120 mg/kg).
  • One week after transplantation, the animals were perfused with 4% para-formaldehyde and serial sections cut on a freezing microtome at 30 μm thickness. Brain sections were stained for astrocytes, oligodendrocytes, neuron, and undifferentiated progenitor cell markers. Minimal migration was demonstrated in adult CNS in the absence of EGF. Excellent survival of the 7 day old grafts was seen in rats receiving EGF as demonstrated by M2 immunoreactivity, and grafts in EGF-treated animals were more extensive than in animals treated with vehicle alone. Furthermore, proliferation of host cells was observed upon EGF treatment. Animals receiving BrdU injections before sacrifice demonstrated an increased number of dividing cells in the treated ventricle, but not the adjoining ventricles.
  • Example 10 Treatment of Syringomyelia
  • Primary fetal transplants have been used to obliterate the syrinx formed around spinal cord injuries in patients. The neural stem cells described in this invention are suitable for replacement, because only a structural function would be required by the cells. Neural stem cells are implanted in the spinal cord of injured patients to prevent syrinx formation. Outcomes are measured preferably by MRI imaging. Clinical trial protocols have been written and could easily be modified to include the described neural stem cells.
  • Example 11 Treatment of Neurodegenerative Disease Using Progent of Human Neural Stem Cells Proliferated In Vitro
  • Cells are obtained from ventral mesencephalic tissue from a human fetus aged 8 weeks following routine suction abortion which is collected into a sterile collection apparatus. A 2×4×1 mm piece of tissue is dissected and dissociated as in Example 2. Neural stem cells are then proliferated. Neural stem cell progeny are used for neurotransplantation into a blood-group matched host with a neurodegenerative disease. Surgery is performed using a BRW computed tomographic (CT) stereotaxic guide. The patient is given local anesthesia suppiemencea with intravenously administered midazolam. The patient undergoes CT scanning to establish the coordinates of the region to receive the transplant. The injection cannula consists of a 17-gauge stainless steel outer cannula with a 19-gauge inner stylet. This is inserted into the brain to the correct coordinates, then removed and replaced with a 19-gauge infusion cannula that has been preloaded with 30 μl of tissue suspension. The cells are slowly infused at a rate of 3 μl/min as the cannula is withdrawn. Multiple stereotactic needle passes are made throughout the area of interest, approximately 4 mm apart. The patient is examined by CT scan postoperatively for hemorrhage or edema. Neurological evaluations are performed at various post-operative intervals, as well as PET scans to determine metabolic activity of the implanted cells.
  • Example 12 Genetic Modification of Neural Stem Cell Progeny Using Calcium Phosphate Transfection
  • Neural stem cell progeny are propagated as described in Example 2. The cells are then transfected using a calcium phosphate transfection technique. For standard calcium phosphate transfection, the cells are mechanically dissociated into a single cell suspension and plated on tissue culture-treated dishes at 50% confluence (50,000-75,000 cells/cm2) and allowed to attach overnight.
  • The modified calcium phosphate transfection procedure is performed as follows: DNA (15-25 μg) in sterile TE buffer (10 mM Tris, 0.25 mM EDTA, pH 7.5) diluted to 4401 with TE, and 60 μl of 2M CaCl2 (pH to 5.8 with 1M HEPES buffer) is added to the DNA/TE buffer. A total of 500 μl of 2×HeBS (HEPES-Buffered saline; 275 mM NaCl, 10 mM KCl, 1.4 mM Na2HPO4, 12 mM dextrose, 40 mM HEPES buffer powder, pH 6.92) is added dropwise to this mix. The mixture is allowed to stand at room temperature for 20 minutes. The cells are washed briefly with 1×HeBS and 1 ml of the calcium phosphate precipitated DNA solution is added to each plate, and the cells are incubated at 37° for 20 minutes. Following this incubation, 10 mls of complete medium is added to the cells, and the plates are placed in an incubator (37° C., 9.5% CO2) for an additional 3-6 hours. The DNA and the medium are removed by aspiration at the end of the incubation period, and the cells are washed 3 times with complete growth medium and then returned to the incubator.
  • Example 13 Genetic Modification of Neural Stem Cell Progeny
  • Cells proliferated as in Examples 2 are transfected with expression vectors containing the genes for the FGF-2 receptor or the NGF receptor. Vector DNA containing the genes are diluted in 0.1×TE (1 mM Tris pH 8.0, 0.1 mM EDTA) to a concentration of 40 μg/ml. 22 μl of the DNA is added to 250 μl of 2×HBS (280 mM NaCl, 10 mM KCl, 1.5 mM Na2HPO42H2O, 12 mM dextrose, 50 mM HEPES) in a disposable, sterile 5 ml plastic tube. 31 μl of 2M CaCl2 is added slowly and the mixture is incubated for 30 minutes at room temperature. During this 30 minute incubation, the cells are centrifuged at 800 g for 5 minutes at 4° C. The cells are resuspended in 20 volumes of ice-cold PBS and divided into aliquots of 1×107 cells, which are again centrifuged. Each aliquot of cells is resuspended in 1 ml of the DNA-CaCl2 suspension, and incubated for 20 minutes at room temperature. The cells are then diluted in growth medium and incubated for 6-24 hours at 37° C. in 5%-7% CO2. The cells are again centrifuged, washed in PBS and returned to 10 ml of growth medium for 48 hours.
  • The transfected neural stem cell progeny are transplanted into a human patient using the procedure described in Example 8 or Example 11, or are used for drug screening procedures as described in the example below.
  • Example 14 Screening of Drugs or Other Biological Agents for Effects on Multipotent Neural Stem Cells and Neural Stem Cell Progeny
  • A. Effects of BDNF on Neuronal and Glial Cell Differentiation and Survival
  • Precursor cells were propagated as described in Example 2 and differentiated as described in Example 4. At the time of plating the cells, BDNF was added at a concentration of 10 ng/ml. At 3, 7, 14, and 21 days in vitro (DIV), cells were processed for indirect immunocytochemistry. BrdU labeling was used to monitor proliferation of the neural stem cells. The effects of BDNF on neurons, oligodendrocytes and astrocytes were assayed by probing the cultures with antibodies that recognize antigens found on neurons (MAP-2, NSE, NF), oligodendrocytes (O4, GalC, MBP) or astrocytes (GFAP). Cell survival was determined by counting the number of immunoreactive cells at each time point and morphological observations were made. BDNF significantly increased the differentiation and survival of neurons over the number observed under control conditions. Astrocyte and oligodendrocyte numbers were not significantly altered from control values.
  • B. Effects of BDNF on the Differentiation of Neural Phenotypes
  • Cells treated with BDNF according to the methods described in Part A were probed with antibodies that recognize neural transmitters or enzymes involved in the synthesis of neural transmitters. These included TH, ChAT, substance P, GABA, somatostatin, and glutamate. In both control and BDNF-treated culture conditions, neurons tested positive for the presence of substance P and GABA. As well as an increase in numbers, neurons grown in BDNF showed a dramatic increase in neurite extension and branching when compared with control examples.
  • C. Identification of Growth-Factor Responsive Cells
  • Cells were differentiated as described in Example 4, and at 1 DIV approximately 100 ng/ml of BDNF was added. At 1, 3, 6, 12 and 24 hours after the addition of BDNF the cells were fixed and processed for dual label immunocytochemistry. Antibodies that recognize neurons (MAP-2, NSE, NF), oligodendrocytes (O4, GalC, MBP) or astrocytes (GFAP) were used in combination with an antibody that recognizes c-fos and/or other immediate early genes. Exposure to BDNF resulted in a selective increase in the expression of c-fos in neuronal cells.
  • D. Effects of BDNF on the Expression of Markers and Regulatory Factors During Proliferation and Differentiation
  • Cells treated with BDNF according to the methods described in Part A are processed for analysis of the expression of regulatory factors, FGF-R1 or other markers.
  • E. Effects of Chlorpromazine on the Proliferation, Differentiation, and Survival of Growth Factor Generated Stem Cell Progeny
  • Chlorpromazine, a drug widely used in the treatment of psychiatric illness, is used in concentrations ranging from 10 ng/ml to 1000 ng/ml in place of BDNF in Examples 14A to 14D above. The effects of the drug at various concentrations on stem cell proliferation and on stem cell progeny differentiation and survival is monitored. Alterations in gene expression and electrophysiological properties of differentiated neurons are determined.

Claims (8)

1. A method for implanting neural stem cells in the central nervous system of a patient comprising transplanting a therapeutically effective amount of human neural stem cell cultures comprising human neural stem cells that have a doubling rate faster than 30 days, wherein the cultures comprise cells which stain positive for nestin, and wherein in the presence of differentiation-inducing conditions said cells produce progeny cells that differentiate into neurons, astrocytes, or oligodendrocytes.
2. The method of claim 1, wherein the human neural stem cells have a doubling rate of 5-10 days.
3. The method of claim 1, wherein the human neural stem cells are substantially formed into neurospheres of a diameter between 10-500 μm.
4. The method of claim 1, wherein the cultures comprise between 10,000 and 1,000,000 cells/μl.
5. The method of claim 1, wherein the cells are proliferated in suspension culture.
6. The method of claim 1, wherein the cells are proliferated in an adherent culture.
7. The method of claim 1, wherein the progeny of said neural stem cells are genetically modified.
8. A method for implanting neural stem cells in the central nervous system of a patient comprising:
a) obtaining a culture comprising human neural stem cells having a doubling rate faster than 30 days, wherein the cultures comprise cells which stain positive for nestin, and wherein in the presence of differentiation-inducing conditions said cells produce progeny cells that differentiate into neurons, astrocytes, or oligodendrocytes;
b) exposing said culture to differentiation inducing-conditions; and
c) transplanting a therapeutically effective amount of said neurons, astrocytes, or oligodendrocytes to the central nervous system of the patient.
US11/903,408 1997-09-05 2007-09-21 Cultures of human CNS neural stem cells Abandoned US20080107633A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/903,408 US20080107633A1 (en) 1997-09-05 2007-09-21 Cultures of human CNS neural stem cells

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US08/926,313 US5968829A (en) 1997-09-05 1997-09-05 Human CNS neural stem cells
PCT/US1998/018597 WO1999011758A2 (en) 1997-09-05 1998-09-04 Cultures of human cns neural stem cells
US09/486,302 US6498018B1 (en) 1997-09-05 1998-09-04 Cultures of human CNS neural stem cells
US10/328,644 US20030166276A1 (en) 2000-10-16 2002-12-23 Cultures of human CNS neural stem cells
US11/356,281 US20060240553A1 (en) 1997-09-05 2006-02-15 Cultures of human CNS neural stem cells
US11/903,408 US20080107633A1 (en) 1997-09-05 2007-09-21 Cultures of human CNS neural stem cells

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/356,281 Continuation US20060240553A1 (en) 1997-09-05 2006-02-15 Cultures of human CNS neural stem cells

Publications (1)

Publication Number Publication Date
US20080107633A1 true US20080107633A1 (en) 2008-05-08

Family

ID=25453045

Family Applications (6)

Application Number Title Priority Date Filing Date
US08/926,313 Expired - Lifetime US5968829A (en) 1997-09-05 1997-09-05 Human CNS neural stem cells
US09/486,302 Expired - Lifetime US6498018B1 (en) 1997-09-05 1998-09-04 Cultures of human CNS neural stem cells
US09/178,035 Expired - Lifetime US6103530A (en) 1997-09-05 1998-10-23 Cultures of human CNS neural stem cells
US10/134,234 Expired - Lifetime US6777233B2 (en) 1997-09-05 2002-04-29 Cultures of human CNS Neural stem cells
US11/356,281 Abandoned US20060240553A1 (en) 1997-09-05 2006-02-15 Cultures of human CNS neural stem cells
US11/903,408 Abandoned US20080107633A1 (en) 1997-09-05 2007-09-21 Cultures of human CNS neural stem cells

Family Applications Before (5)

Application Number Title Priority Date Filing Date
US08/926,313 Expired - Lifetime US5968829A (en) 1997-09-05 1997-09-05 Human CNS neural stem cells
US09/486,302 Expired - Lifetime US6498018B1 (en) 1997-09-05 1998-09-04 Cultures of human CNS neural stem cells
US09/178,035 Expired - Lifetime US6103530A (en) 1997-09-05 1998-10-23 Cultures of human CNS neural stem cells
US10/134,234 Expired - Lifetime US6777233B2 (en) 1997-09-05 2002-04-29 Cultures of human CNS Neural stem cells
US11/356,281 Abandoned US20060240553A1 (en) 1997-09-05 2006-02-15 Cultures of human CNS neural stem cells

Country Status (6)

Country Link
US (6) US5968829A (en)
EP (1) EP1007636B1 (en)
JP (2) JP4848538B2 (en)
AU (1) AU758270B2 (en)
CA (1) CA2302484C (en)
WO (1) WO1999011758A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012164137A1 (en) 2011-05-30 2012-12-06 Fundación Investigación En Regeneración Del Sistema Nervioso Stem cells and neural crest cells derived from olfactory ensheathing glia, and uses thereof
CN105420105A (en) * 2015-12-25 2016-03-23 北京工业大学 Biochip and manufacturing method thereof

Families Citing this family (233)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7544511B2 (en) 1996-09-25 2009-06-09 Neuralstem Biopharmaceuticals Ltd. Stable neural stem cell line methods
US5968829A (en) * 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
US6093531A (en) * 1997-09-29 2000-07-25 Neurospheres Holdings Ltd. Generation of hematopoietic cells from multipotent neural stem cells
US5958767A (en) * 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
US7410798B2 (en) * 2001-01-10 2008-08-12 Geron Corporation Culture system for rapid expansion of human embryonic stem cells
US20020168766A1 (en) * 2000-01-11 2002-11-14 Gold Joseph D. Genetically altered human pluripotent stem cells
US6667176B1 (en) * 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
US6488930B1 (en) * 1999-01-15 2002-12-03 Millennium Pharmaceuticals, Inc. Anti-CCR4 antibodies and methods of use therefor
US7105150B2 (en) * 1999-02-12 2006-09-12 Stemcells California, Inc. In vivo screening methods using enriched neural stem cells
US6468794B1 (en) * 1999-02-12 2002-10-22 Stemcells, Inc. Enriched central nervous system stem cell and progenitor cell populations, and methods for identifying, isolating and enriching for such populations
ES2351170T3 (en) * 1999-02-12 2011-02-01 Stemcells California, Inc. ENRICHED POPULATIONS OF CENTRAL NERVOUS SYSTEM CELLS.
US7037719B1 (en) 1999-02-12 2006-05-02 Stemcells California, Inc. Enriched central nervous system stem cell and progenitor cell populations, and methods for identifying, isolating and enriching for such populations
US7217565B2 (en) * 1999-02-12 2007-05-15 Stemcells California, Inc. Enriched central nervous system stem cell and progenitor cell populations, and methods for identifying, isolating and enriching for such populations
EP1165771A2 (en) * 1999-03-17 2002-01-02 The University Of Tennessee Research Corporation Rt-pcr of clonal neurospheres
JP2003524396A (en) * 1999-03-18 2003-08-19 ウェルスタット バイオロジクス コーポレイション Cells, cell populations and methods of making and using them.
US20050265977A1 (en) * 1999-04-30 2005-12-01 Elliott Robert B Xenotransplant for CNS therapy
EP1176970B1 (en) * 1999-04-30 2010-05-19 NeurotrophinCell Pty Ltd Xenotransplant for cns therapy
US20090047325A1 (en) * 1999-04-30 2009-02-19 Neurotrophincell Pty. Limited Xenotransplant for cns therapy
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
US8252280B1 (en) 1999-08-05 2012-08-28 Regents Of The University Of Minnesota MAPC generation of muscle
US10638734B2 (en) 2004-01-05 2020-05-05 Abt Holding Company Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US8609412B2 (en) 1999-08-05 2013-12-17 Regents Of The University Of Minnesota Mapc generation of lung tissue
US7015037B1 (en) 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US6749850B1 (en) 1999-08-18 2004-06-15 The General Hospital Corporation Methods, compositions and kits for promoting recovery from damage to the central nervous system
JP2003512333A (en) * 1999-10-20 2003-04-02 ステム セルズ, インコーポレイテッド Methods for inducing the in vivo proliferation and migration of transplanted precursor cells in the brain
US6878543B1 (en) 1999-10-25 2005-04-12 Nsgene Sa Cultures of GFAP+ nestin+ cells that differentiate to neurons
CA2393071A1 (en) * 1999-12-07 2001-06-14 Monash University Long-term cell culture compositions and genetically modified animals derived therefrom
AU779273B2 (en) * 1999-12-07 2005-01-13 Monash University Long-term cell culture compositions and genetically modified animals derived therefrom
EP1107003A1 (en) * 1999-12-09 2001-06-13 Trophos Methods for screening compounds active on neurons
US7468277B2 (en) * 1999-12-23 2008-12-23 Cornell Research Foundation, Inc. Enriched preparation of human fetal multipotential neural stem cells
US7455983B2 (en) * 2000-01-11 2008-11-25 Geron Corporation Medium for growing human embryonic stem cells
US20050042749A1 (en) * 2001-05-16 2005-02-24 Carpenter Melissa K. Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
EP1248640B1 (en) * 2000-01-20 2006-10-04 Diabcell Pty Limited Preparation and xenotransplantation of porcine islets
ATE473751T1 (en) * 2000-02-11 2010-07-15 Schepens Eye Res Inst ISOLATION AND TRANSPLANTATION OF RETINAL STEM CELLS
CA2401989A1 (en) * 2000-03-03 2001-09-07 The Walter And Eliza Hall Institute Of Medical Research A method of treatment
US7011828B2 (en) * 2000-03-14 2006-03-14 Es Cell International Pte. Ltd. Implanting neural progenitor cells derived for human embryonic stem cells
WO2001068815A1 (en) 2000-03-14 2001-09-20 Es Cell International Pte Ltd Embryonic stem cells and neural progenitor cells derived therefrom
US6808702B2 (en) 2000-04-13 2004-10-26 Board Of Regents, The University Of Texas System Treatment of disorders by implanting stem cells and/or progeny thereof into gastrointestinal organs
US7037493B2 (en) 2000-05-01 2006-05-02 Cornell Research Foundation, Inc. Method of inducing neuronal production in the brain and spinal cord
US6828145B2 (en) 2000-05-10 2004-12-07 Cedars-Sinai Medical Center Method for the isolation of stem cells by immuno-labeling with HLA/MHC gene product marker
GB2379447B (en) * 2000-05-17 2004-12-29 Geron Corp Neural progenitor cell populations
US7250294B2 (en) * 2000-05-17 2007-07-31 Geron Corporation Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
US6897061B1 (en) * 2000-06-16 2005-05-24 Spinal Cord Society Transdifferentiation of glial cells
US6602680B2 (en) 2000-07-14 2003-08-05 The Regents Of The University Of California Production of gabaergic cells
DK1333846T3 (en) * 2000-10-17 2012-08-06 Diatranz Otsuka Ltd Preparation and xenotransplantation of porcine islands
IL155728A0 (en) * 2000-11-22 2003-11-23 Geron Corp Tolerizing allografts of pluripotent stem cells
US6576464B2 (en) 2000-11-27 2003-06-10 Geron Corporation Methods for providing differentiated stem cells
US6921665B2 (en) * 2000-11-27 2005-07-26 Roslin Institute (Edinburgh) Selective antibody targeting of undifferentiated stem cells
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
EP1362095B1 (en) 2001-02-14 2015-05-27 Anthrogenesis Corporation 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
WO2002069976A2 (en) * 2001-03-02 2002-09-12 Stem Cell Therapeutics Inc. Use of ovarian hormone for increasing neural stem cell number
EP1370134B1 (en) * 2001-03-23 2016-03-23 University Of Ottawa Methods and compositions for cryopreservation of dissociated primary animal cells
JP3763749B2 (en) * 2001-03-28 2006-04-05 独立行政法人科学技術振興機構 Central nervous system progenitor cells that induce synaptogenic neurons in the spinal cord
US7838292B1 (en) 2001-03-29 2010-11-23 University Of Louisville Research Foundation, Inc. Methods for obtaining adult human olfactory progenitor cells
US20020169102A1 (en) * 2001-04-03 2002-11-14 Frey William H. Intranasal delivery of agents for regulating development of implanted cells in the CNS
WO2002086106A1 (en) * 2001-04-23 2002-10-31 Nsgene A/S Method and culture medium for producing neural cells expressing tyrosine hydroxylase
US7943376B2 (en) 2001-04-27 2011-05-17 Stem Cell Therapuetics Inc. Platelet derived growth factor (PDGF)-derived neurospheres define a novel class of progenitor cells
US7132287B2 (en) * 2001-06-18 2006-11-07 Psychiatric Genomics, Inc. Method for neural stem cell differentiation using 5HT-1A agonists
WO2002102989A2 (en) * 2001-06-18 2002-12-27 Psychiatric Genomics, Inc. Method for neural stem cell differentiation using valproate
WO2003000852A2 (en) * 2001-06-22 2003-01-03 The University Of Texas System Method of producing region-specific neurons from human neuronal stem cells
CN1549924A (en) * 2001-07-16 2004-11-24 �����ʸ���˹�ع�˾ Arrays of buffers for analysing biomolecules by their isoelectric point
WO2003014317A2 (en) * 2001-08-08 2003-02-20 Celmed Biosciences Usa Compositions and methods for isolation, propagation, and differentiation of human stem cells and uses thereof
WO2003014320A2 (en) * 2001-08-10 2003-02-20 Cornell Research Foundation, Inc. Telomerase immortalized human neutral stem cells and phenotypically-restricted progenitor cells
ATE535601T1 (en) * 2001-09-28 2011-12-15 Diabcell Pty Ltd BREEDING OF FOREIGN TRANSPLANTATION MATERIAL IN CULTURE
WO2003029418A2 (en) * 2001-10-02 2003-04-10 Becton, Dickinson And Company Proliferation and differentiation of stem cells using extracellular matrix and other molecules
US8153424B2 (en) * 2001-10-03 2012-04-10 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US7588937B2 (en) * 2001-10-03 2009-09-15 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US7129034B2 (en) * 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
NZ515310A (en) * 2001-11-07 2004-08-27 Diabcell Pty Ltd Methods of treatment and delivery modes
WO2003044158A1 (en) * 2001-11-15 2003-05-30 Becton, Dickinson And Company Methods and devices for the integrated discovery of cell culture environments
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
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
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
US7285415B2 (en) 2002-07-11 2007-10-23 The Regents Of The University Of California Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US20050101014A1 (en) * 2002-07-11 2005-05-12 Keirstead Hans S. Oligodendrocytes derived from human embryonic stem cells for remyelination and treatment of spinal cord injury
US7312025B2 (en) * 2002-07-12 2007-12-25 University Of Washington Methods and systems for extended in vitro culture of neuronal cells
CA2493768A1 (en) * 2002-07-23 2004-01-29 Nanodiagnostics, Inc. Embryonic stem cell markers and uses thereof
WO2004029203A2 (en) * 2002-09-25 2004-04-08 Bresagen, Inc. Compositions and methods for enrichment of neural stem cells using ceramide analogs
AU2003273332A1 (en) * 2002-09-26 2004-04-19 Children's Medical Center Corporation Method of enhancing proliferation and/or hematopoietic differentiation of stem cells
US20040147020A1 (en) * 2002-11-01 2004-07-29 The Regents Of The University Of Colorado, A Body Corporate Dopamine neurons from human embryonic stem cells
US20050265983A1 (en) * 2002-11-17 2005-12-01 Eldad Melamed Methods, nucleic acid constructs and cells for treating neurodegenerative disorders
DK1583541T3 (en) * 2002-11-20 2011-04-11 Neuronova Ab Compounds and Methods to Increase Neurogenesis
WO2004047770A2 (en) 2002-11-26 2004-06-10 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
CN1745168A (en) * 2002-12-02 2006-03-08 安增子摩祺株式会社 Method for culturing neural stem cells using hepatocyte growth factor
US8293488B2 (en) 2002-12-09 2012-10-23 Neuralstem, Inc. Method for screening neurogenic agents
AU2003293409A1 (en) 2002-12-09 2004-06-30 Karl K. Johe Method for discovering neurogenic agents
US20060211111A1 (en) * 2002-12-18 2006-09-21 Maisam Mitalipova Compositions and methods for neural cell production and stabilization
ATE446791T1 (en) 2003-05-16 2009-11-15 Univ Laval CNS CHLORIDE MODULATION AND USE THEREOF
WO2006040615A2 (en) * 2003-06-11 2006-04-20 Jan Remmereit Differenciation of stem cells for therapeutic use
ES2584428T3 (en) 2003-06-20 2016-09-27 Axiogenesis Ag Tissue modeling in an embryonic stem cell system (ES)
CN1791670A (en) * 2003-06-24 2006-06-21 戴伯塞尔有限公司 Porcine pancreatic islet cultured with porcine sertoli cells for xenotransplantation
WO2005003320A2 (en) 2003-07-02 2005-01-13 Regents Of The University Of Minnesota Neuronal differentiation of stem cells
EP1644485B9 (en) 2003-07-08 2012-01-04 Axiogenesis Ag Secreted proteins as markers for cell differentiation
ATE505539T1 (en) * 2003-09-09 2011-04-15 Univ Keio METHOD FOR PROMOTING SURVIVAL AND/OR PROLIFERATION OF A NEURAL STEM CELL AND FOR PROMOTING NEURAL STEM CELL EXTENSION, PROMOTER THEREOF, PHARMACEUTICAL COMPOSITION CONTAINING THE NEURAL STEM CELL, TESTING METHODS AND SCREENING METHODS
ATE493504T1 (en) * 2003-09-12 2011-01-15 Stemcell Technologies Inc TEST FOR NEURAL COLONEY FORMATION
WO2005033297A1 (en) * 2003-09-19 2005-04-14 The Rockefeller University Compositions, methods and kits relating to reprogramming adult differentiated cells and production of embryonic stem cell-like cells
US20050148069A1 (en) * 2003-09-24 2005-07-07 Gage Fred H. IGF-1 instructs multipotent adult CNS neural stem cells to an oligodendroglial lineage
JP2005139164A (en) * 2003-11-10 2005-06-02 Toudai Tlo Ltd Pharmaceutical composition for treating neuropathy and neurodegenerative disease
JP4676442B2 (en) * 2003-12-02 2011-04-27 セラヴィー バイオサイエンシズ エルエルシー Compositions and methods for growing neural progenitor cells
US20070269412A1 (en) * 2003-12-02 2007-11-22 Celavie Biosciences, Llc Pluripotent cells
US20050196864A1 (en) * 2004-02-10 2005-09-08 Goldman Steven A. Induction and high-yield preparative purification of mesencephalic dopaminergic neuronal progenitor cells and dopaminergic neurons from human embryonic stem cells
WO2005079250A2 (en) * 2004-02-13 2005-09-01 Cornell Research Foundation, Inc. Purines are self-renewal signals for neural stem cells, and purine receptor antagonists promote neuronal and glial differentiation therefrom
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
ES2557772T3 (en) * 2004-06-09 2016-01-28 The University Court Of The University Of Edinburgh Neural stem cells
ES2294650T3 (en) 2004-09-30 2008-04-01 Reneuron Limited CELLPHONE LINE.
CN101084303A (en) 2004-11-17 2007-12-05 神经干公司 Transplantation of human neural cells for treatment of neurodegenerative conditions
US20090010873A1 (en) * 2004-11-29 2009-01-08 Yeda Research And Development Co. Ltd. Methods of Cell Therapy, Neurogenesis and Oligodendrogenesis
CA2589343A1 (en) 2004-12-01 2006-06-08 Stem Cell Therapeutics Corporation Platelet-derived growth factor-responsive neural precursor cells and progeny thereof
US7018804B1 (en) 2004-12-03 2006-03-28 Orion Biosolutions, Inc. Determination of cell viability and phenotype
US20060121443A1 (en) * 2004-12-03 2006-06-08 Orion Biosolutions, Inc. Determination of cell viability
SG158851A1 (en) * 2005-01-04 2010-02-26 Stemcells California Inc Methods for the treatment of lysosomal storage disorders
AU2006210425A1 (en) * 2005-01-31 2006-08-10 Cognate Therapeutics, Inc. Adipose derived adult stromal cells exhibiting characteristics of endothelial cells
NZ540597A (en) * 2005-06-08 2007-02-23 Neurotrophincell Pty Ltd A method for preventing the onset of type I diabetes comprising administering an implantable composition comprising living choroid plexus cells
ES2524996T3 (en) 2005-06-16 2014-12-16 Ramot At Tel Aviv University Ltd. Isolated cells and populations that comprise them for the treatment of CNS diseases
GB2441488C (en) * 2005-06-22 2011-10-05 Geron Corp Suspension culture of human embryonic stem cells
JP2009500043A (en) * 2005-07-08 2009-01-08 ブレインセルス,インコーポレイティド Methods for identifying agents and conditions that modulate neurogenesis
TW200726474A (en) * 2005-07-15 2007-07-16 Cognate Therapeutics Inc The immunophenotype and immunogenicity of human adipose derived cells
US20090148417A1 (en) * 2005-07-28 2009-06-11 Brainguard Co., Ltd Carbon nanotubes serving as stem cell scaffold
US8357666B2 (en) * 2005-08-01 2013-01-22 Nupotential, Inc. Reprogramming a cell by inducing a pluripotent gene through RNA interference
US9095573B2 (en) * 2005-08-01 2015-08-04 University Of Central Florida Research Foundation, Inc. Method of biasing implanted human neural stem cells away from differentiation into glial cells by (+)phenserine to modulate the concentration of soluble βAPP in tissue or CSF
WO2008020815A1 (en) 2006-08-15 2008-02-21 Agency For Science, Technology And Research Mesenchymal stem cell conditioned medium
EP2530145A1 (en) 2005-10-13 2012-12-05 Anthrogenesis Corporation Immunomodulation using placental stem cells
US7803364B2 (en) 2005-11-17 2010-09-28 The Cleveland Clinic Foundation Multipotent neural stem cells
ATE517178T1 (en) * 2005-11-17 2011-08-15 Cleveland Clinic Foundation MULTIPOTENT NEURAL STEM CELLS
EP1976978A2 (en) 2005-12-29 2008-10-08 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
EP2422803A3 (en) 2006-03-07 2013-03-13 Geeta Shroff Compositions comprising human embryonic stem cells and their derivatives, methods of use, and methods of preparation
JP2009532369A (en) * 2006-03-30 2009-09-10 リサーチ ファウンデーション オブ シティ ユニバーシティ オブ ニューヨーク Stimulation of nerve regeneration by secretory leukocyte protease inhibitor
US20100297087A1 (en) * 2006-04-11 2010-11-25 Nanodiagnostics Israel., Ltd Pluripotent stem cells characterized by expression of germline specific genes
US7727763B2 (en) 2006-04-17 2010-06-01 Bioe, Llc Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
WO2008020329A2 (en) 2006-05-11 2008-02-21 Regenics As Administration of cells and cellular extracts for rejuvenation
US8877253B2 (en) * 2006-05-11 2014-11-04 Regenics As Cellular extracts
US20080003676A1 (en) * 2006-06-26 2008-01-03 Millipore Corporation Growth of embryonic stem cells
EP2049043B1 (en) * 2006-07-24 2018-10-10 International Stem Cell Corporation Synthetic cornea from retinal stem cells
EP2061876A2 (en) * 2006-08-31 2009-05-27 The University of Louisville Research Foundation, Inc. Transcription factors for differentiation of adult human olfactory progenitor cells
PL3255061T3 (en) 2006-11-03 2021-12-06 The Board Of Trustees Of The Leland Stanford Junior University Selective immunodepletion of endogenous stem cell niche for engraftment
WO2008066658A2 (en) * 2006-11-03 2008-06-05 The Trustees Of Princeton University Engineered cellular pathways for programmed autoregulation of differentiation
CA2670497A1 (en) 2006-11-24 2008-05-29 Regents Of The University Of Minnesota Endodermal progenitor cells
US20090136456A1 (en) * 2006-12-22 2009-05-28 Yadong Huang Methods of treating neurodegenerative disorders
NZ597779A (en) 2007-02-12 2013-07-26 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
US20080213892A1 (en) * 2007-03-02 2008-09-04 The Board Of Trustees Of The Leland Stanford Junior University Self-renewal of neural stem cells is promoted by wnt proteins
JP2010520286A (en) * 2007-03-05 2010-06-10 ザ ボード オブ トラスティーズ オブ ザ リーランド スタンフォード ジュニア ユニバーシティ Wnt composition and method of use thereof
EP2136646A1 (en) * 2007-03-08 2009-12-30 Raymond F. Sekula Jr. A method of producing purified neural stem cells and related methods of treating a patient
WO2008134406A1 (en) * 2007-04-25 2008-11-06 The Board Of Trustees Of The Leland Stanford Junior University Ischemia-induced neovascularization is enhanced by hcns-sc transplantation
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
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
US9200253B1 (en) 2007-08-06 2015-12-01 Anthrogenesis Corporation Method of producing erythrocytes
EP2028268A1 (en) 2007-08-20 2009-02-25 Université Libre De Bruxelles Generation of neuronal cells from pluripotent stem cells
CN102316888A (en) * 2007-08-23 2012-01-11 利兰斯坦福青年大学托管委员会 Synaptogenic regulation and control
US9700582B2 (en) 2007-09-11 2017-07-11 Sapporo Medical University Cell growth method and pharmaceutical preparation for tissue repair and regeneration
EP3524253A1 (en) 2007-09-28 2019-08-14 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
US20090090214A1 (en) * 2007-10-04 2009-04-09 Chung Yuan Christian University Method for forming nano-scale metal particles
SG188096A1 (en) 2008-01-30 2013-03-28 Geron Corp Synthetic surfaces for culturing stem cell derived oligodendrocyte progenitor cells
WO2009116951A2 (en) 2008-03-17 2009-09-24 Agency For Science, Technology And Research Microcarriers for stem cell culture
JP2011522514A (en) 2008-04-07 2011-08-04 ニューポテンシャル,インコーポレイテッド Cell reprogramming by inducing pluripotency genes via RNA interference
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
ES2729557T3 (en) 2008-05-28 2019-11-04 Univ Ramot Mesenchymal stem cells for the treatment of CNS diseases
WO2010008486A2 (en) 2008-06-24 2010-01-21 Parkinsons Institute Pluripotent cell lines and methods of use thereof
KR20180108887A (en) 2008-08-20 2018-10-04 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
EP3539380A3 (en) 2008-08-20 2019-12-18 Celularity, Inc. Improved cell composition and methods of making the same
CA2734446C (en) 2008-08-22 2017-06-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
RU2562154C2 (en) 2008-11-19 2015-09-10 Антродженезис Корпорейшн Amniotic adhesive cells
GB0822246D0 (en) * 2008-12-05 2009-01-14 Reneuron Ltd Composition
CN102325878A (en) 2008-12-23 2012-01-18 斯特姆塞尔思加利福尼亚有限公司 Target populations of oligodendrocyte precursor cells and methods of making and using same
CA2756283A1 (en) 2009-02-03 2010-08-12 Keio University Culture method of embryoid bodies and/or neural stem cells derived from human differentiated cell-derived pluripotent stem cells
GB0902034D0 (en) * 2009-02-06 2009-03-11 Reneuron Ltd Method
US20100209399A1 (en) * 2009-02-13 2010-08-19 Celavie Biosciences, Llc Brain-derived stem cells for repair of musculoskeletal system in vertebrate subjects
US9101570B1 (en) 2009-02-13 2015-08-11 Endocellutions, Inc. Adult and neonatal stem cell therapy to treat diabetes through the repair of the gastrointestinal tract
US9511093B2 (en) * 2009-03-23 2016-12-06 The Texas A & M University System Compositions of mesenchymal stem cells to regenerate bone
JP5783504B2 (en) * 2009-03-30 2015-09-24 国立研究開発法人国立環境研究所 Methods for assessing impact on embryonic programming
WO2010129917A2 (en) 2009-05-08 2010-11-11 Vaccinex, Inc. Anti-cd100 antibodies and methods for using the same
CA2767014C (en) 2009-07-02 2022-01-25 Anthrogenesis Corporation Method of producing erythrocytes without feeder cells
SG10201913618WA (en) 2009-07-21 2020-03-30 Abt Holding Co Use of stem cells to reduce leukocyte extravasation
DK2456853T3 (en) 2009-07-21 2021-02-01 Abt Holding Co USE OF STEM CELLS TO REDUCE LEUCOCYT EXTRAVASATION
DK2430454T3 (en) 2009-08-22 2013-02-11 Univ Leland Stanford Junior Imaging and assessment of embryos, oocytes and stem cells
US9234179B2 (en) 2009-12-18 2016-01-12 Shanghai Icell Biotechnology Co., Ltd. Materials and methods for generating pluripotent stem cells
ES2646750T3 (en) 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
US8927276B2 (en) * 2010-02-17 2015-01-06 Cellin Technologies Oue Ex vivo progenitor and stem cell expansion and differentiation for use in the treatment of disease of the nervous system
SG183497A1 (en) 2010-02-25 2012-09-27 Abt Holding Co Modulation of macrophage activation
TWI578993B (en) 2010-04-07 2017-04-21 安瑟吉納西斯公司 Angiogenesis using placental stem cells
TW201138792A (en) 2010-04-08 2011-11-16 Anthrogenesis Corp Treatment of sarcoidosis using placental stem cells
EP3502254A1 (en) 2010-04-23 2019-06-26 Cold Spring Harbor Laboratory Novel structurally designed shrnas
WO2011138687A2 (en) 2010-05-06 2011-11-10 Regenics As Use of cellular extracts for skin rejuvenation
KR20130093091A (en) 2010-07-13 2013-08-21 안트로제네시스 코포레이션 Methods of generating natural killer cells
CA2806904C (en) 2010-07-28 2018-11-27 Neuralstem, Inc. Methods for treating and/or reversing neurodegenerative diseases and/or disorders
MX348419B (en) 2010-08-19 2017-06-12 F Hoffmann-La Roche Ag * Conversion of somatic cells to induced reprogrammed neural stem cells (irnscs).
WO2012034070A1 (en) 2010-09-09 2012-03-15 The Board Of Trustees Of The Leland Stanford Junior University Use of liposomal wnt compositions to enhance osseointegration
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
EP2658557A1 (en) 2010-12-31 2013-11-06 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
WO2012106281A2 (en) 2011-01-31 2012-08-09 The General Hospital Corporation Multimodal trail molecules and uses in cellular therapies
EP2678675B1 (en) 2011-02-23 2017-10-11 The Board of Trustees of the Leland Stanford Junior University Methods of detecting aneuploidy in human embryos
TWI602570B (en) 2011-06-01 2017-10-21 安瑟吉納西斯公司 Treatment of pain using placental stem cells
JP6182135B2 (en) 2011-06-06 2017-08-16 レゲネシス ベーフェーベーアー Amplification of stem cells in a hollow fiber bioreactor
US20130046093A1 (en) * 2011-08-18 2013-02-21 Korea Institute Of Science And Technology Pharmaceutical compositions for preventing or treating degenerative brain disease and method of screening the same
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
BR112014008885B1 (en) 2011-10-11 2021-02-17 Vaccinex, Inc use of an isolated antibody or antigen-binding fragment that specifically binds to semaphorin-4d to decrease blood-brain barrier (bbb) permeability in an individual with increased bbb permeability and a neuroinflammatory disorder
EP2782587B1 (en) 2011-11-21 2018-11-07 Ramot at Tel Aviv University, Ltd. Stem cell-derived neural cells for cell therapy in neurological disorders
EP2814947B1 (en) 2012-02-17 2018-11-21 Schepens Eye Research Institute Phenotype profile of human retinal progenitor cells
CN102604894B (en) * 2012-02-29 2014-07-30 中国科学院广州生物医药与健康研究院 Culture medium for preparing neural stem cells and application thereof
EP3470073A1 (en) 2012-04-03 2019-04-17 Reneuron Limited Stem cell microparticles
GB201302468D0 (en) 2013-02-12 2013-03-27 Reneuron Ltd Stem cell product
EP2840133B1 (en) * 2012-04-18 2017-06-14 Jeong Chan Ra Method for manufacturing stem cell having appropriate size for intravascular administration
US10494440B2 (en) 2012-05-11 2019-12-03 Vaccinex, Inc. Use of semaphorin-4D binding molecules to promote neurogenesis following stroke
ES2717478T3 (en) 2012-05-31 2019-06-21 Ares Trading Sa Methods of prediction of embryonic blastocysts in vitro
KR101523040B1 (en) * 2012-06-26 2015-05-26 라정찬 Method for Preparing High Concentration of Stem Cells
WO2014013258A1 (en) 2012-07-19 2014-01-23 Reneuron Limited Stem cell microparticles
EP2890720B1 (en) 2012-08-30 2019-07-17 The General Hospital Corporation Compositions and methods for treating cancer
US10357517B1 (en) 2012-10-01 2019-07-23 University Of South Florida Methods of treating epilepsy using neural stem cells that express nanog, SSEA-4, OCT-4, MIR-34B, MIR-34C and MIR-592
EP3459522B1 (en) 2012-12-10 2021-02-17 Regenics AS Use of egg cellular extracts for wound treatment
US10241108B2 (en) 2013-02-01 2019-03-26 Ares Trading S.A. Abnormal syngamy phenotypes observed with time lapse imaging for early identification of embryos with lower development potential
EP3622960A1 (en) 2013-02-05 2020-03-18 Celularity, Inc. Natural killer cells from placenta
AU2014217615B2 (en) 2013-02-12 2018-11-15 Reneuron Limited Method of producing microparticles
SG10201914136UA (en) 2013-04-12 2020-03-30 Saverio Lafrancesca Improving organs for transplantation
US20140322135A1 (en) 2013-04-30 2014-10-30 Katholieke Universiteit Leuven Cell therapy for myelodysplastic syndromes
GB201317887D0 (en) 2013-10-09 2013-11-20 Reneuron Ltd Product
GB201317889D0 (en) 2013-10-09 2013-11-20 Reneuron Ltd Product and use
NZ630892A (en) 2013-10-21 2016-03-31 Vaccinex Inc Use of semaphorin-4d binding molecules for treating neurodegenerative disorders
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
WO2015140005A1 (en) 2014-03-19 2015-09-24 Ifom Fondazione Istituto Firc Di Oncologia Molecolare Method of generation of pluripotent cells
WO2015143350A1 (en) 2014-03-20 2015-09-24 Auxogyn, Inc. Quantitative measurement of human blastocyst and morula morphology developmental kinetics
US10406179B2 (en) 2014-08-26 2019-09-10 The Board Of Trustees Of The Leland Stanford Junior University Engraftment of stem cells with a combination of an agent that targets stem cells and modulation of immunoregulatory signaling
US9750769B2 (en) 2014-10-20 2017-09-05 Neuralstem, Inc. Stable neural stem cells comprising an exogenous polynucleotide coding for a growth factor and methods of use thereof
EP3288570A4 (en) 2015-04-29 2018-11-21 Fred Hutchinson Cancer Research Center Modified stem cells and uses thereof
US20190216891A1 (en) 2016-03-06 2019-07-18 Yeda Research And Development Co., Ltd. Method for modulating myelination
US10513689B2 (en) 2016-04-29 2019-12-24 Hope Biosciences, Llc Culture media for multipotent stem cells
BR112019015342A2 (en) 2017-01-30 2020-03-10 The Board Of Trustees Of The Leland Stanford Junior University NON-GENOToxic CONDITIONING REGIME FOR STEM CELL TRANSPLANTATION
GB201919021D0 (en) 2019-12-20 2020-02-05 Reneuron Ltd Nucleic acid constructs for delivering polynucleotides into exosomes
CN115698262A (en) 2020-03-30 2023-02-03 阿卜杜拉国王科技大学 Compositions and methods for controlling cell identity
CN112080470B (en) * 2020-09-14 2022-07-12 中国科学院遗传与发育生物学研究所 In-vitro culture method for differentiation of neural stem cells into neurons
GB202114441D0 (en) 2021-10-08 2021-11-24 Reneuron Ltd Proteins and extracellular vesicles

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4753635A (en) * 1986-05-23 1988-06-28 Jacqueline Sagen Inducing analgesia by implantation of cells releasing neuroactive substances
US4980174A (en) * 1988-12-23 1990-12-25 Jacqueline Sagen Method for alleviating depression
US5082670A (en) * 1988-12-15 1992-01-21 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage or the central nervous system
US5175103A (en) * 1991-10-21 1992-12-29 Trustees Of University Of Pennsylvania Preparation of pure cultures of post-mitotic human neurons
US5411883A (en) * 1989-12-26 1995-05-02 Somatix Therapy Corporation Proliferated neuron progenitor cell product and process
US5580777A (en) * 1987-10-29 1996-12-03 Amrad Corporation Limited Generation of neural precursor cell lines
US5612211A (en) * 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
US5639618A (en) * 1994-05-13 1997-06-17 Plurion, Inc. Method of isolating a lineage specific stem cell in vitro
US5672499A (en) * 1992-07-27 1997-09-30 California Institute Of Technology Immoralized neural crest stem cells and methods of making
US5688692A (en) * 1990-02-20 1997-11-18 Ludwig Institute For Cancer Research Transgenic mouse cells expressing ts SV40 large T
US5750376A (en) * 1991-07-08 1998-05-12 Neurospheres Holdings Ltd. In vitro growth and proliferation of genetically modified multipotent neural stem cells and their progeny
US5753506A (en) * 1996-05-23 1998-05-19 Cns Stem Cell Technology, Inc. Isolation propagation and directed differentiation of stem cells from embryonic and adult central nervous system of mammals
US5766948A (en) * 1993-01-06 1998-06-16 The Regents Of The University Of California Method for production of neuroblasts
US5851832A (en) * 1991-07-08 1998-12-22 Neurospheres, Ltd. In vitro growth and proliferation of multipotent neural stem cells and their progeny
US5968829A (en) * 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
US5980885A (en) * 1991-07-08 1999-11-09 Neurospheres Holdings Ltd. Growth factor-induced proliferation of neural precursor cells in vivo

Family Cites Families (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU6174490A (en) * 1989-08-04 1991-03-11 Board Of Regents, The University Of Texas System Methods and compositions; purified preparation of neural progenitor regulatory factor
WO1991009936A1 (en) * 1989-12-26 1991-07-11 Hana Biologics, Inc. Proliferated neuron progenitor cell product and process
US5292918A (en) * 1990-04-26 1994-03-08 Schering Corporation Diastereoselective process for preparing n-substituted amino acids and derivatives
WO1994016718A1 (en) * 1991-07-08 1994-08-04 Neurospheres Ltd. Genetic modification of neural stem cells
CA2113118C (en) * 1991-07-08 2002-09-17 Samuel Weiss Novel growth factor-responsive progenitor cells which can be proliferated in vitro
ES2133333T3 (en) * 1991-11-22 1999-09-16 Genentech Inc TGF-BETA TO IMPROVE THE RECOVERY OF NEURONS.
CA2147162C (en) * 1992-10-16 2002-04-30 Samuel Weiss Remyelination using neural stem cells
WO1994010292A1 (en) * 1992-10-28 1994-05-11 Neurospheres Ltd. Biological factors and neural stem cells
US5405772A (en) * 1993-06-18 1995-04-11 Amgen Inc. Medium for long-term proliferation and development of cells
KR970706490A (en) * 1994-09-23 1997-11-03 스코트 디. 코르맥 Laboratory model of central nervous system function and dysfunction (IN VITRO MODELS OF CNS FUNCTION AND DYSFUNCTION)
KR970707272A (en) * 1994-11-14 1997-12-01 스코트 디. 코르맥 REGULATION OF NEURAL STEM CELL PROLIFERATION
JP2997995B2 (en) 1995-09-13 2000-01-11 日本コーンスターチ株式会社 Aqueous dispersion of biodegradable resin composition
WO1997016534A1 (en) * 1995-11-01 1997-05-09 Genentech, Inc. Normal neural epithelial precursor cells

Patent Citations (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4753635A (en) * 1986-05-23 1988-06-28 Jacqueline Sagen Inducing analgesia by implantation of cells releasing neuroactive substances
US5580777A (en) * 1987-10-29 1996-12-03 Amrad Corporation Limited Generation of neural precursor cell lines
US5082670A (en) * 1988-12-15 1992-01-21 The Regents Of The University Of California Method of grafting genetically modified cells to treat defects, disease or damage or the central nervous system
US4980174A (en) * 1988-12-23 1990-12-25 Jacqueline Sagen Method for alleviating depression
US5411883A (en) * 1989-12-26 1995-05-02 Somatix Therapy Corporation Proliferated neuron progenitor cell product and process
US5688692A (en) * 1990-02-20 1997-11-18 Ludwig Institute For Cancer Research Transgenic mouse cells expressing ts SV40 large T
US5612211A (en) * 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
US5851832A (en) * 1991-07-08 1998-12-22 Neurospheres, Ltd. In vitro growth and proliferation of multipotent neural stem cells and their progeny
US5980885A (en) * 1991-07-08 1999-11-09 Neurospheres Holdings Ltd. Growth factor-induced proliferation of neural precursor cells in vivo
US5750376A (en) * 1991-07-08 1998-05-12 Neurospheres Holdings Ltd. In vitro growth and proliferation of genetically modified multipotent neural stem cells and their progeny
US5175103A (en) * 1991-10-21 1992-12-29 Trustees Of University Of Pennsylvania Preparation of pure cultures of post-mitotic human neurons
US5672499A (en) * 1992-07-27 1997-09-30 California Institute Of Technology Immoralized neural crest stem cells and methods of making
US5766948A (en) * 1993-01-06 1998-06-16 The Regents Of The University Of California Method for production of neuroblasts
US5639618A (en) * 1994-05-13 1997-06-17 Plurion, Inc. Method of isolating a lineage specific stem cell in vitro
US5753506A (en) * 1996-05-23 1998-05-19 Cns Stem Cell Technology, Inc. Isolation propagation and directed differentiation of stem cells from embryonic and adult central nervous system of mammals
US5968829A (en) * 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
US6103530A (en) * 1997-09-05 2000-08-15 Cytotherapeutics, Inc. Cultures of human CNS neural stem cells

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012164137A1 (en) 2011-05-30 2012-12-06 Fundación Investigación En Regeneración Del Sistema Nervioso Stem cells and neural crest cells derived from olfactory ensheathing glia, and uses thereof
CN105420105A (en) * 2015-12-25 2016-03-23 北京工业大学 Biochip and manufacturing method thereof

Also Published As

Publication number Publication date
US6777233B2 (en) 2004-08-17
WO1999011758A2 (en) 1999-03-11
US6498018B1 (en) 2002-12-24
JP4848538B2 (en) 2011-12-28
CA2302484A1 (en) 1999-03-11
JP2009106301A (en) 2009-05-21
EP1007636B1 (en) 2014-06-18
AU9305998A (en) 1999-03-22
US20060240553A1 (en) 2006-10-26
US5968829A (en) 1999-10-19
AU758270B2 (en) 2003-03-20
US6103530A (en) 2000-08-15
CA2302484C (en) 2013-12-24
EP1007636A2 (en) 2000-06-14
US20020164309A1 (en) 2002-11-07
JP2002518990A (en) 2002-07-02
WO1999011758A3 (en) 1999-05-27

Similar Documents

Publication Publication Date Title
US6777233B2 (en) Cultures of human CNS Neural stem cells
CA2113118C (en) Novel growth factor-responsive progenitor cells which can be proliferated in vitro
US8501467B2 (en) Cultures of GFAP+ nestin+ cells that differentiate to neurons
US6787355B1 (en) Multipotent neural stem cells from peripheral tissues and uses thereof
AU2002324645C1 (en) Compositions and methods for isolation, propagation, and differentiation of human stem cells and uses thereof
AU755657B2 (en) Lineage-restricted neuronal precursors
WO2006055841A2 (en) Indefinite culture of human adult glia without immortalization and therapeutic uses thereof
US6787353B1 (en) Lineage-restricted neuronal precursors and methods of isolation
US20030166276A1 (en) Cultures of human CNS neural stem cells
AU2118501A (en) Methods for inducing in vivo proliferation and migration of transplanted progenitor cells in the brain
US20040103448A1 (en) Methods for inducing in vivo proliferation and migration of transplanted progenitor cells in the brain
MXPA00000225A (en) Lineage-restricted neuronal precursors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION