US20020009743A1 - Neural progenitor cell populations - Google Patents

Neural progenitor cell populations Download PDF

Info

Publication number
US20020009743A1
US20020009743A1 US09/859,351 US85935101A US2002009743A1 US 20020009743 A1 US20020009743 A1 US 20020009743A1 US 85935101 A US85935101 A US 85935101A US 2002009743 A1 US2002009743 A1 US 2002009743A1
Authority
US
United States
Prior art keywords
cells
cell
cell population
neural
population
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
US09/859,351
Other languages
English (en)
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.)
Asterias Biotherapeutics Inc
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=26900583&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20020009743(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US09/859,351 priority Critical patent/US20020009743A1/en
Application filed by Individual filed Critical Individual
Assigned to GERON CORPORATION reassignment GERON CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CARPENTER, MELISSA K.
Priority to US09/872,183 priority patent/US6833269B2/en
Publication of US20020009743A1 publication Critical patent/US20020009743A1/en
Priority to US10/157,288 priority patent/US7250294B2/en
Priority to US10/873,414 priority patent/US20050042749A1/en
Priority to US11/009,504 priority patent/US7763463B2/en
Priority to US11/281,040 priority patent/US8148148B2/en
Priority to US12/332,783 priority patent/US8252585B2/en
Priority to US13/558,078 priority patent/US9803174B2/en
Assigned to ASTERIAS BIOTHERAPEUTICS, INC. reassignment ASTERIAS BIOTHERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GERON CORPORATION
Priority to US14/217,699 priority patent/US20140242691A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0618Cells of the nervous system
    • C12N5/0623Stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6881Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for tissue or cell typing, e.g. human leukocyte antigen [HLA] probes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/01Modulators of cAMP or cGMP, e.g. non-hydrolysable analogs, phosphodiesterase inhibitors, cholera toxin
    • 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
    • C12N2501/10Growth factors
    • C12N2501/119Other fibroblast growth factors, e.g. FGF-4, FGF-8, FGF-10
    • 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
    • C12N2501/13Nerve growth factor [NGF]; Brain-derived neurotrophic factor [BDNF]; Cilliary neurotrophic factor [CNTF]; Glial-derived neurotrophic factor [GDNF]; Neurotrophins [NT]; Neuregulins
    • 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
    • C12N2501/135Platelet-derived growth factor [PDGF]
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers

Definitions

  • This invention relates generally to the field of cell biology of embryonic cells and neural progenitor cells. More specifically, this invention relates to the directed differentiation of human pluripotent stem cells to form cells of the neuronal and glial lineages, using special culture conditions and selection techniques.
  • neural progenitor cells For this reason, there is a great deal of evolving interest in neural progenitor cells. Up until the present time, it was generally thought that multipotent neural progenitor cells commit early in the differentiation pathway to either neural restricted cells or glial restricted cells. These in turn are thought to give rise to mature neurons, or to mature astrocytes and oligodendrocytes. Multipotent neural progenitor cells in the neural crest also differentiate to neurons, smooth muscle, and Schwann cells. It is hypothesized that various lineage-restricted precursor cells renew themselves and reside in selected sites of the central nervous system, such as the spinal chord. Cell lineage in the developing neural tube has been reviewed in the research literature by Kalyani et al. (Biochem. Cell Biol. 6:1051, 1998).
  • NEP cells neuroepithelial cells
  • Kalyani et al. (Dev. Biol. 186:202, 1997) reported NEP cells in the rat.
  • Mujtaba et al. (Dev. Biol. 214:113, 1999) reported NEP cells in the mouse. Differentiation of NEP cells is thought to result in formation of restricted precursor cells having characteristic surface markers.
  • NPP neural restricted precursors
  • GFPs glial restricted precursors
  • Mujtaba et al. reported isolation of NRP and GRP cells from mouse embryonic stem (mES) cells.
  • the NRPs were PS-NCAM immunoreactive, underwent self-renewal in defined medium, and differentiated into multiple neuronal phenotypes. They apparently did not form glial cells.
  • the GRPs were A2B5-immunoreactive, and reportedly differentiated into astrocytes and oligodendrocytes, but not neurons.
  • hES and hEG cells have the long-sought characteristics of human pluripotent stem (hPS) cells: they are capable of ongoing proliferation in vitro without differentiating, they retain a normal karyotype, and they retain the capacity to differentiate to produce all adult cell types.
  • hPS human pluripotent stem
  • Reubinoff et al. (Nature Biotechnol. 18:399, 2000) reported somatic differentiation of human blastocysts. The cells differentiated spontaneously in culture, with no consistent pattern of structural organization. After culturing for 4-7 weeks to high density, multicellular aggregates formed above the plane of the monolayer. Different cells in the culture expressed a number of different phenotypes, including expression of ⁇ -actin, desmin, and NCAM.
  • This invention provides a system for efficient production of primate cells that have differentiated from pluripotent cells into cells of the neuronal or glial lineage.
  • Populations of cells are described which contain precursors for either lineage, which provide a source for generating additional precursor cells, the mature cells of the central nervous system: neurons, astrocytes, or oligodendrocytes.
  • Certain embodiments of the invention have the ability to generate cells of both lineages.
  • the precursor and mature cells of this invention can be used a number of important applications, including drug testing and therapy to restore nervous system function.
  • One embodiment of this invention is a cell population that proliferates in an in vitro culture, obtained by differentiating primate pluripotent stem (pPS) cells, wherein at least about 30% of the cells in the population are committed to form neuronal cells, glial cells, or both.
  • a second embodiment is a cell population that proliferates in an in vitro culture, comprising at least about 60% neural progenitor cells, wherein at least 10% of the cells can differentiate into neuronal cells, and at least 10% of the cells can differentiate into glial cells.
  • a third embodiment is a cell population that proliferates in an in vitro culture, comprising at least about 60% neural progenitor cells, wherein at least 10% of the cells express A2B5, and at least 10% of the cells express NCAM.
  • Certain cell populations of the invention are obtained by differentiating primate pluripotent stem cells, such as human embryonic stem cells. Some are obtained by differentiating stem cells in a medium containing at least two or more ligands that bind growth factor receptors. Some are obtained by differentiating pPS cells in a medium containing growth factors, sorting the differentiated cells for expression of NCAM or A2B5, and then 0 collecting the sorted cells. Certain cell populations are enriched such that at least 70% of the cells express NCAM or A2B5.
  • Another embodiment of this invention is a cell population comprising mature neurons, astrocytes, oligodendrocytes, or any combination thereof, obtained by further differentiating a precursor cell population of this invention.
  • Some such populations are obtained by culturing neural precursors in a medium containing an activator of cAMP, a neurotrophic factor, or a combination of such factors.
  • neurons produced by such methods may be capable of exhibiting an action potential, may show gated sodium and potassium channels, and may show calcium flux when administered with neurotransmitters or their equivalents. Included are populations of cells containing a substantial proportion of dopaminergic neurons, detectable for example by staining for tyrosine hydroxylase.
  • Also embodied in the invention are isolated neural precursor cells, neurons, astrocytes, and oligodendrocytes, obtained by selecting a cell for the desired phenotype from one of the cell populations.
  • the cell populations and isolated cells of this invention will typically have the same genome as the line from which they are derived. This means that the chromosomal DNA will be over 90% identical between the pPS cells and the neural cells, which can be inferred if the neural cells are obtained from the undifferentiated line through the course of normal mitotic division.
  • Neural cells that have been treated by recombinant methods to introduce a transgene (such as TERT) or knock out an endogenous gene are still considered to have the same genome as the line from which they are derived, since all non-manipulated genetic elements are preserved.
  • a further embodiment of the invention is a method of screening a compound for neural cell toxicity or modulation, in which a culture is prepared containing the compound and a neural cell or cell population of this invention, and any phenotypic or metabolic change in the cell that results from contact with the compound is determined.
  • Yet another embodiment of the invention is a method for obtaining a polynucleotide comprising a nucleotide sequence contained in an mRNA more highly expressed in neural progenitor cells or differentiated cells, as described and exemplified further on in this disclosure.
  • the nucleotide sequence can in turn be used to produce recombinant or synthetic polynucleotides, proteins, and antibodies for gene products enriched or suppressed in neural cells.
  • Antibodies can also be obtained by using the cells of this invention as an immunogen or an adsorbent to identify markers enriched or suppressed in neural cells.
  • a further embodiment of the invention is a method of reconstituting or supplementing central nervous system (CNS) function in an individual, in which the individual is administered with an isolated cell or cell population of this invention.
  • the isolated cells and cell populations can be used in the preparation of a medicament for use in clinical and veterinary treatment.
  • Medicaments comprising the cells of this invention can be formulated for use in such therapeutic applications.
  • Other embodiments of the invention are methods for obtaining the neural precursor cells and fully differentiated cells of this invention, using the techniques outlined in this disclosure on a suitable stem cell population. Included are methods for producing cell populations containing dopaminergic cells at a frequency of 1%, 3% or 5%—and populations of progenitor cells capable of generating dopaminergic cells at this frequency—from primate embryonic stem cells. This is particularly significant in view of the loss in dopamine neuron function that occurs in Parkinson's disease.
  • the compositions, methods, and techniques described in this disclosure hold considerable promise for use in diagnostic, drug screening, and therapeutic applications.
  • FIG. 1 is a graph representing the growth of cells bearing neural markers that were derived from human embryonic stem cells.
  • the upper panel shows growth of cells maintained in the presence of CNTF, bFGF, and NT3, and then sorted for expression of NCAM.
  • the lower panel shows growth of cells maintained in the presence of EGF, bFGF, PDGF, and IGF-1, and then sorted for expression of A2B5.
  • Four different hES cell lines were used: H1, H7, H9, and H13.
  • the A2B5 selected population has been passaged over 7 times, and can be further differentiated into both neuronal and glial cells.
  • FIG. 2 is a schematic diagram outlining an exemplary procedure for obtaining A2B5-positive cells.
  • FIG. 3 is a half-tone reproduction of a fluorescence micrograph of the brains from neonatal rats administered with cells that express green fluorescent protein.
  • Left panels parental hES cell line.
  • Middle panels embryoid body cells formed from the parental line.
  • Right panels differentiated cells sorted for expression of NCAM. Undifferentiated hES cells and embryoid body cells remain in the area of administration and show evidence of necrosis. In contrast, the differentiated NCAM + cells appear as single cells, and have migrated away from the injection site.
  • FIG. 4 is a photocopy reproduction of a fluorescence micrograph showing a cell staining for tyrosine hydroxylase (TH), a marker for dopaminergic cells.
  • TH tyrosine hydroxylase
  • Embryoid bodies made from human ES cells were maintained in 10 ⁇ m retinoic acid for 4 days, plated into a neural-supportive cocktail, and then passaged into medium containing 10 ng/mL NT-3 and 10 ng/mL BDNF. Certain populations of this invention contain >1% TH-positive cells.
  • FIG. 5 is a series of graphs showing response of the neural-restricted precursors to various neurotransmitters.
  • Panel A shows the ratio of emission data from single cells on two different coverslips. Both cells responded to GABA, elevated potassium, acetylcholine and ATP.
  • Panel B shows the frequency of cells tested that responded to specific neurotransmitters.
  • Panel C shows the combinations of neurotransmitter responses observed.
  • FIG. 6 is a series of graphs showing electrophysiology of neural-restricted precursors.
  • Panel A shows sodium and potassium currents observed in two cells depolarized to test potentials between ⁇ 80 and 80 mV from a holding potential of ⁇ 100 mV.
  • Panel B shows the inward (Na + ) and outward (K) peak current-voltage relationships observed.
  • Panel C shows action potentials generated by the same cells n response to depolarizing stimuli.
  • This invention provides a system for preparing and characterizing neural progenitor cells, suitable for use for therapeutic administration and drug screening.
  • pluripotent stem cells when pluripotent stem cells are cultured in the presence of selected differentiating agents, a population of cells is derived that has a remarkably high proportion of cells with phenotypic characteristics of neural cells.
  • the proportion of neural cells can be enhanced by sorting differentiated cells according to cell-surface markers. Since certain types of pluripotent stem cells (such as embryonic stem cells) can proliferate in culture for a year or more, the invention described in this disclosure provides an almost limitless supply of neural precursors. Certain cell populations of this invention are capable of generating cells of the neuronal or glial lineage, and themselves can be replicated through a large number of passages in culture.
  • FIG. 1 shows the growth curve of cells that have been cultured with differentiating agents, and then selected according to whether they bear polysialylated NCAM, or the A2B5 epitope. Either of these cell populations can be proliferated through a large number of cell doublings.
  • Differentiated cells positively selected for A2B5 expression comprise cells that appear to express A2B5 without NCAM, and cells that express A2B5 and NCAM simultaneously. In one of the experiments described below, maturation of these cells produced 13% oligodendrocytes, and 38% neurons. Since these cells proliferate in long-term culture without losing their phenotype, the population can provide a reserve of multipotential cells. Upon administration to a subject with CNS dysfunction, the population would comprise cells that may repopulate both the neuronal and glial cell lineage, as needed.
  • the neural precursor cells can be further differentiated ex vivo, either by culturing with a maturation factor, such as a neurotrophic factor, or by withdrawing one or more factors that sustain precursor cell renewal.
  • a maturation factor such as a neurotrophic factor
  • Neurons, astrocytes, and oligodendrocytes are mature differentiated cells of the neural lineage that can be obtained by culturing the precursor cells in this fashion.
  • the neurons obtained by these methods have extended processes characteristic of this cell type, show staining for neuron-specific markers like neurofilament and MAP-2, and show evidence of synapse formation, as detected by staining for synaptophysin.
  • FIG. 5 shows that these cells respond to a variety of neurotransmitter substances.
  • FIG. 6 shows that these cells are capable of action potentials as measured in a standard patch-clamp system. In all these respects, the cells are apparently capable of full neurological function.
  • FIG. 4 Of particular interest is the ability of this system to generate a supply of dopaminergic neurons (FIG. 4).
  • Cells of this type are particularly desirable for the treatment of Parkinson's disease, for which the best current modality is an allograft of fetal brain tissue.
  • the use of fetal tissue as a clinical therapy is fraught with supply and procedural issues, but no other source described previously can supply the right kind of cells with sufficient abundance.
  • the neural precursor cells of this invention are capable of generating differentiated cells in which several percent of the neurons have a dopaminergic phenotype. This is believed to be a sufficient proportion for cell replacement therapy in Parkinson's disease, and warrants the development of the progenitor populations of this invention for therapeutic use.
  • neural progenitor cell or “neural precursor cell” mean a cell that can generate progeny that are either neuronal cells (such as neuronal precursors or mature neurons) or glial cells (such as glial precursors, mature astrocytes, or mature oligodendrocytes).
  • the cells express some of the phenotypic markers that are characteristic of the neural lineage. Typically, they do not produce progeny of other embryonic germ layers when cultured by themselves in vitro, unless dedifferentiated or reprogrammed in some fashion.
  • a “neuronal progenitor cell” or “neuronal precursor cell” is a cell that can generate progeny that are mature neurons. These cells may or may not also have the capability to generate glial cells.
  • a “glial progenitor cell” or “glial precursor cell” is a cell that can generate progeny that are mature astrocytes or mature oligodendrocytes. These cells may or may not also have the capability to generate neuronal cells.
  • a “multipotent neural progenitor cell population” is a cell population that has the capability to generate both progeny that are neuronal cells (such as neuronal precursors or mature neurons), and progeny that are glial cells (such as glial precursors, mature astrocytes, or mature oligodendrocytes), and sometimes other types of cells.
  • the term does not require that individual cells within the population have the capability of forming both types of progeny, although individual cells that are multipotent neural progenitors may be present.
  • pluripotent embryonic stem cells can differentiate to lineage-restricted precursor cells, such as hematopoetic cells, which are pluripotent for blood cell types; hepatocyte progenitors, which are pluripotent for hepatocytes; and various types of neural progenitors listed above. These in turn can be differentiated further to other types of precursor cells further down the pathway, or to an end-stage differentiated cell, which plays a characteristic role in a certain tissue type, and may or may not retain the capacity to proliferate further. Neurons, astrocytes, and oligodendrocytes are all examples of terminally differentiated cells.
  • a “differentiation agent”, as used in this disclosure, refers to one of a collection of compounds that are used in culture systems of this invention to produce differentiated cells of the neural lineage (including precursor cells and terminally differentiated cells). No limitation is intended as to the mode of action of the compound.
  • the agent may assist the differentiation process by inducing or assisting a change in phenotype, promoting growth of cells with a particular phenotype or retarding the growth of others, or acting in concert with other agents through unknown mechanisms.
  • Prototype “primate Pluripotent Stem cells” are pluripotent cells derived from pre-embryonic, embryonic, or fetal tissue at any time after fertilization, and have the characteristic of being capable under appropriate conditions of producing progeny of several different cell types that are derivatives of all of the three germinal layers (endoderm, mesoderm, and ectoderm), according to a standard art-accepted test, such as the ability to form a teratoma in 8-12 week old SCID mice.
  • pPS cells include embryonic cells of various types, exemplified by human embryonic stem (hES) cells, described by Thomson et al. (Science 282:1145, 1998); embryonic stem cells from other primates, such as Rhesus stem cells (Thomson et al., Proc. Nati. Acad. Sci. USA 92:7844, 1995), marmoset stem cells (Thomson et al., Biol. Reprod. 55:254, 1996) and human embryonic germ (hEG) cells (Shamblott et al., Proc. Natl. Acad. Sci. USA 95:13726, 1998). Other types of pluripotent cells are also included in the term.
  • hES human embryonic stem
  • Any cells of primate origin that are capable of producing progeny that are derivatives of all three germinal layers are included, regardless of whether they were derived from embryonic tissue, fetal tissue, or other sources.
  • the pPS cells are not derived from a malignant source. It is desirable (but not always necessary) that the cells be karyotypically normal.
  • pPS cell cultures are described as “undifferentiated” when a substantial proportion of stem cells and their derivatives in the population display morphological characteristics of undifferentiated cells, clearly distinguishing them from differentiated cells of embryo or adult origin. Undifferentiated pPS cells are easily recognized by those skilled in the art, and typically appear in the two dimensions of a microscopic view in colonies of cells with high nuclear/cytoplasmic ratios and prominent nucleoli. It is understood that colonies of undifferentiated cells within the population will often be surrounded by neighboring cells that are differentiated. “Feeder cells” or “feeders” are terms used to describe cells of one type that are co-cultured with cells of another type, to provide an environment in which the cells of the second type can grow.
  • pPS cells can be supported by primary mouse embryonic fibroblasts, immortalized mouse embryonic fibroblasts, or human fibroblast-like cells differentiated from hES cell.
  • pPS cell populations are said to be “essentially free” of feeder cells if the cells have been grown through at least one round after splitting in which fresh feeder cells are not added to support the growth of the pPS.
  • embryoid bodies is a term of art synonymous with “aggregate bodies”.
  • the terms refer to aggregates of differentiated and undifferentiated cells that appear when pPS cells overgrow in monolayer cultures, or are maintained in suspension cultures.
  • Embryoid bodies are a mixture of different cell types, typically from several germ layers, distinguishable by morphological criteria and cell markers detectable by immunocytochemistry.
  • a “growth environment” is an environment in which cells of interest will proliferate, differentiate, or mature in vitro.
  • the environment include the medium in which the cells are cultured, any growth factors or differentiation-inducing factors that may be present, and a supporting structure (such as a substrate on a solid surface) if present.
  • a cell is said to be “genetically altered”, “transfected”, or “genetically transformed” when a polynucleotide has been transferred into the cell by any suitable means of artificial manipulation, or where the cell is a progeny of the originally altered cell that has inherited the polynucleotide.
  • the polynucleotide will often comprise a transcribable sequence encoding a protein of interest, which enables the cell to express the protein at an elevated level.
  • the genetic alteration is said to be “inheritable” if progeny of the altered cell have the same alteration.
  • antibody refers to both polyclonal and monoclonal antibody.
  • the ambit of the term deliberately encompasses not only intact immunoglobulin molecules, but also such fragments and derivatives of immunoglobulin molecules (such as single chain Fv constructs, diabodies, and fusion constructs) as may be prepared by techniques known in the art, and retaining a desired antibody binding specificity.
  • This invention can be practiced using stem cells of various types, which may include the following non- limiting examples.
  • U.S. Pat. No. 5,851,832 reports multipotent neural stem cells obtained from brain tissue.
  • U.S. Pat. No. 5,766,948 reports producing neuroblasts from newborn cerebral hemispheres.
  • U.S. Pat. Nos. 5,654,183 and 5,849,553 report the use of mammalian neural crest stem cells.
  • U.S. Pat. No. 6,040,180 reports in vitro generation of differentiated neurons from cultures of mammalian multipotential CNS stem cells.
  • WO 98/50526 and WO 99/01159 report generation and isolation of neuroepithelial stem cells, oligodendrocyte-astrocyte precursors, and lineage-restricted neuronal precursors.
  • U.S. Pat. No. 5,968,829 reports neural stem cells obtained from embryonic forebrain and cultured with a medium comprising glucose, transferrin, insulin, selenium, progesterone, and several other growth factors.
  • stem cells of any vertebrate species include stem cells from humans; as well as non-human primates, domestic animals, livestock, and other non-human mammals.
  • the stem cells suitable for use in this invention are primate pluripotent stem (pPS) cells derived from tissue formed after gestation, such as a blastocyst, or fetal or embryonic tissue taken any time during gestation.
  • pPS pluripotent stem
  • Non-limiting examples are primary cultures or established lines of embryonic stem cells or embryonic germ cells.
  • Embryonic stem cells can be isolated from blastocysts of members of the primate species (Thomson et al., Proc. Natl. Acad. Sci. USA 92:7844, 1995).
  • Human embryonic stem (hES) cells can be prepared from human blastocyst cells using the techniques described by Thomson et al. (U.S. Pat. No. 5,843,780; Science 282:1145, 1998; Curr. Top. Dev. Biol. 38:133 ff., 1998) and Reubinoff et al, Nature Biotech. 18:399,2000.
  • human blastocysts are obtained from human in vivo preimplantation embryos.
  • in vitro fertilized (IVF) embryos can be used, or one-cell human embryos can be expanded to the blastocyst stage (Bongso et al., Hum Reprod 4: 706,1989).
  • Embryos are cultured to the blastocyst stage in G1.2 and G2.2 medium (Gardner et al., Fertil. Steril. 69:84, 1998).
  • the zona pellucida is removed from developed blastocysts by brief exposure to pronase (Sigma).
  • the inner cell masses are isolated by immunosurgery, in which blastocysts are exposed to a 1:50 dilution of rabbit anti-human spleen cell antiserum for 30 min, then washed for 5 min three times in DMEM, and exposed to a 1:5 dilution of Guinea pig complement (Gibco) for 3 min (Solter et al., Proc. Natl. Acad. Sci. USA 72:5099, 1975). After two further washes in DMEM, lysed trophectoderm cells are removed from the intact inner cell mass (ICM) by gentle pipetting, and the ICM plated on mEF feeder layers.
  • ICM inner cell mass
  • inner cell mass-derived outgrowths are dissociated into clumps, either by exposure to calcium and magnesium-free phosphate-buffered saline (PBS) with 1 mM EDTA, by exposure to dispase or trypsin, or by mechanical dissociation with a micropipette; and then replated on mEF in fresh medium.
  • PBS calcium and magnesium-free phosphate-buffered saline
  • EDTA calcium and magnesium-free phosphate-buffered saline
  • dispase or trypsin or by mechanical dissociation with a micropipette
  • ES-like morphology is characterized as compact colonies with apparently high nucleus to cytoplasm ratio and prominent nucleoli.
  • ES cells are then routinely split every 1-2 weeks by brief trypsinization, exposure to Dulbecco's PBS (containing 2 mM EDTA), exposure to type IV collagenase ( ⁇ 200 U/mL; Gibco) or by selection of individual colonies by micropipette. Clump sizes of about 50 to 100 cells are optimal.
  • Human Embryonic Germ (hEG) cells can be prepared from primordial germ cells present in human fetal material taken about 8-11 weeks after the last menstrual period. Suitable preparation methods are described in Shamblott et al., Proc. Natl. Acad. Sci. USA 95:13726,1998 and U.S. Pat. No. 6,090,622.
  • genital ridges are rinsed with isotonic buffer, then placed into 0.1 mL 0.05% trypsin/0.53 mM sodium EDTA solution (BRL) and cut into ⁇ 1 mm 3 chunks.
  • the tissue is then pipetted through a 100 ⁇ L tip to further disaggregate the cells. It is incubated at 37° C. for ⁇ 5 min, then ⁇ 3.5 mL EG growth medium is added.
  • EG growth medium is DMEM, 4500 mg/L D-glucose, 2200 mg/L mM NaHCO 3 ; 15% ES qualified fetal calf serum (BRL); 2 mM glutamine (BRL); 1 mM sodium pyruvate (BRL); 1000-2000 U/mL human recombinant leukemia inhibitory factor (LIF, Genzyme); 1-2 ng/ml human recombinant bFGF (Genzyme); and 10 ⁇ M forskolin (in 10% DMSO).
  • EG cells are isolated using hyaluronidase/collagenase/DNAse.
  • Gonadal anlagen or genital ridges with mesenteries are dissected from fetal material, the genital ridges are rinsed in PBS, then placed in 0.1 ml HCD digestion solution (0.01% hyaluronidase type V, 0.002% DNAse I, 0.1% collagenase type IV, all from Sigma prepared in EG growth medium). Tissue is minced, incubated 11 h or overnight at 37° C., resuspended in 1-3 mL of EG growth medium, and plated onto a feeder layer.
  • HCD digestion solution 0.01% hyaluronidase type V, 0.002% DNAse I, 0.1% collagenase type IV, all from Sigma prepared in EG growth medium.
  • feeder cells e.g., STO cells, ATCC No. CRL 1503
  • modified EG growth medium free of LIF, bFGF or forskolin inactivated with 5000 rad ⁇ -irradiation.
  • PSC primary germ cell
  • the first passage is done after 7-10 days in EG growth medium, transferring each well to one well of a 24-well culture dish previously prepared with irradiated STO mouse fibroblasts.
  • the cells are cultured with daily replacement of medium until cell morphology consistent with EG cells is observed, typically after 7-30 days or 1-4 passages.
  • pPS cells can be propagated continuously in culture, using culture conditions that promote proliferation without promoting differentiation.
  • Exemplary serum-containing ES medium is made with 80% DMEM (such as Knock-Out DMEM, Gibco), 20% of either defined fetal bovine serum (FBS, Hyclone) or serum replacement (WO 98/30679), 1% non-essential amino acids, 1 mM L-glutamine, and 0.1 mM ⁇ -mercaptoethanol.
  • FBS defined fetal bovine serum
  • FBS defined fetal bovine serum
  • WO 98/30679 serum replacement
  • human bFGF is added to 4 ng/mL (WO 99/20741, Geron Corp.).
  • ES cells are cultured on a layer of feeder cells, typically fibroblasts derived from embryonic or fetal tissue. Embryos are harvested from a CF1 mouse at 13 days of pregnancy, transferred to 2 mL trypsin/EDTA, finely minced, and incubated 5 min at 37° C. 10% FBS is added, debris is allowed to settle, and the cells are propagated in 90% DMEM, 10% FBS, and 2 mM glutamine. To prepare a feeder cell layer, cells are irradiated to inhibit proliferation but permit synthesis of factors that support ES cells ( ⁇ 4000 rads ⁇ -irradiation). Culture plates are coated with 0.5% gelatin overnight, plated with 375,000 irradiated mEFs per well, and used 5 h to 4 days after plating. The medium is replaced with fresh hES medium just before seeding pPS cells.
  • feeder cells typically fibroblasts derived from embryonic or fetal tissue. Embryos are harvested from a CF1 mouse
  • pPS cells can alternatively be maintained in an undifferentiated state even without feeder cells.
  • the environment for feeder-free cultures includes a suitable culture substrate, particularly an extracellular matrix such as Matrigel® or laminin.
  • the pPS cells are plated at >15,000 cells cm ⁇ 2 (optimally 90,000 cm ⁇ 2 to 170,000 cm ⁇ 2 ).
  • enzymatic digestion is halted before cells become completely dispersed (say, ⁇ 5 min with collagenase IV).
  • Clumps of ⁇ 10-2000 cells are then plated directly onto the substrate without further dispersal.
  • Feeder-free cultures are supported by a nutrient medium typically conditioned by culturing irradiated primary mouse embryonic fibroblasts, telomerized mouse fibroblasts, or fibroblast-like cells derived from pPS cells.
  • Medium can be conditioned by plating the feeders at a density of ⁇ 5.6 ⁇ 10 4 cm ⁇ 2 in a serum free medium such as KO DMEM supplemented with 20% serum replacement and 4 ng/mL bFGF.
  • a serum free medium such as KO DMEM supplemented with 20% serum replacement and 4 ng/mL bFGF.
  • Medium that has been conditioned for 1-2 days is supplemented with further bFGF, and used to support pPS cell culture for 1-2 days.
  • ES cells Under the microscope, ES cells appear with high nuclear/cytoplasmic ratios, prominent nucleoli, and compact colony formation with poorly discernable cell junctions. Primate ES cells express stage-specific embryonic antigens (SSEA) 3 and 4, and markers detectable using antibodies designated Tra-1-60 and Tra-1-81 (Thomson et al., Science 282:1145, 1998). Mouse ES cells can be used as a positive control for SSEA-1, and as a negative control for SSEA-4, Tra-1-60, and Tra-1-81. SSEA-4 is consistently present on human embryonal carcinoma (hEC) cells. Differentiation of pPS cells in vitro results in the loss of SSEA-4, Tra-1-60, and Tra-1-81 expression and increased expression of SSEA-1. SSEA-1 is also found on hEG cells.
  • SSEA stage-specific embryonic antigens
  • the factors are supplied to the cells in a nutrient medium, which is any medium that supports the proliferation or survival of the desired cell type. It is often desirable to use a defined medium that supplies nutrients as free amino acids rather than serum. It is also beneficial to supplement the medium with additives developed for sustained cultures of neural cells. Exemplary are N2 and B27 additives, available commercially from Gibco.
  • the stem cells are pPS cells
  • the cells obtained from feeder cell supported or feeder-free cultures
  • pPS cells are plated directly onto a suitable substrate, such as an adherent glass or plastic surface, such as coverslips coated with a polyamine.
  • a suitable substrate such as an adherent glass or plastic surface, such as coverslips coated with a polyamine.
  • the cells are then cultured in a suitable nutrient medium that is adapted to promote differentiation towards the desired cell lineage. This is referred to as the “direct differentiation” method.
  • pPS cells are first let differentiate into a heterogeneous cell population.
  • embryoid bodies are formed from the pPS cells by culturing them in suspension.
  • one or more of the differentiation agents listed earlier can be included in the medium to promote differentiation within the embryoid body.
  • the embryoid bodies After the embryoid bodies have reached sufficient size (typically 3-4 days), they are plated onto the substrate of the differentiation culture.
  • the embryoid bodies can be plated directly onto the substrate without dispersing the cells. This allows neural cell precursors to migrate out of the embryoid bodies and on to the extracellular matrix. Subsequent passaging of these cultures into an appropriate medium helps select out the neural progenitor cells.
  • Cells prepared according to these procedures have been found to be capable of further proliferation (Example 1). As many as 30%, 50%, 75% or more of the cells express either polysialylated NCAM or the A2B5 epitope, or both. Typically, at least about 10%, 20%, 30% or 50% of the cells express NCAM, and at least about 10%, 20%, 30% or 50% of the cells express A2B5—which implies that they have the capacity to form cells of the neuronal lineage, and the glial lineage, respectively.
  • the differentiated cells can be sorted based on phenotypic features to enrich for certain populations. Typically, this will involve contacting each cell with an antibody or ligand that binds to a marker characteristic of neural cells, followed by separation of the specifically recognized cells from other cells in the population.
  • One method is immunopanning, in which specific antibody is coupled to a solid surface. The cells are contacted with the surface, and cells not expressing the marker are washed away. The bound cells are then recovered by more vigorous elution. Variations of this are affinity chromatography and antibody-mediated magnetic cell sorting.
  • the cells are contacted with a specific primary antibody, and then captured with a secondary anti-immunoglobulin reagent bound to a magnetic bead.
  • the adherent cells are then recovered by collecting the beads in a magnetic field.
  • cells selected positively for polysialylated NCAM can provide a population that is 60%, 70%, 80%, or even 90% NCAM positive (Example 1). This implies that they are capable of forming some type of neural cell, including neurons.
  • cells selected positively for A2B5 can provide a population that is 60%, 70%, 80%, or even 90% A2B5 positive (Example 2). This implies that they are capable of forming some type of neural cell, possibly including both neurons and glial cells.
  • the A2B5 positive cells can be sorted again into two separate populations: one that is A2B5 positive and NCAM negative, and one that is both A2B5 positive and NCAM positive.
  • Differentiated or separated cells prepared according to this procedure can be maintained or proliferated further in any suitable culture medium.
  • the medium will contain most of the ingredients used initially to differentiate the cells.
  • neural precursor cells prepared according to these procedures can be further differentiated to mature neurons, astrocytes, or oligodendrocytes. This can be effected by culturing the cells in a maturation factor, such as forskolin or other compound that elevates intracellular cAMP levels, such as cholera toxin, isobutylmethylxanthine, dibutyladenosine cyclic monophosphate, or other factors such as c-kit ligand, retinoic acid, or neurotrophins. Particularly effective are neurotrophin-3 (NT-3) and brain-derived neurotrophic factor (BDNF). Other candidates are GDNF, BMP-2, and BMP-4. Alternatively or in addition, maturation can be enhanced by withdrawing some or all of the factors that promote neural precursor proliferation, such as EGF or FGF.
  • a maturation factor such as forskolin or other compound that elevates intracellular cAMP levels, such as cholera toxin, isobutylmethylxanthine, dibutyladen
  • populations of precursor or mature neurological cells be substantially free of undifferentiated pPS cells.
  • One way of depleting undifferentiated stem cells from the population is to transfect them with a vector in which an effector gene under control of a promoter that causes preferential expression in undifferentiated cells.
  • Suitable promoters include the TERT promoter and the OCT-4 promoter.
  • the effector gene may be directly lytic to the cell (encoding, for example, a toxin or a mediator of apoptosis). Alternatively, the effector gene may render the cell susceptible to toxic effects of an external agent, such as an antibody or a prodrug.
  • Exemplary is a herpes simplex thymidine kinase (tk) gene, which causes cells in which it is expressed to be susceptible to ganciclovir.
  • tk herpes simplex thymidine kinase
  • Suitable pTERT-tk constructs are provided in International Patent Publication WO 98/14593 (Morin et al.).
  • Cells can be characterized according to a number of phenotypic criteria.
  • the criteria include but are not limited to microscopic observation of morphological features, detection or quantitation of expressed cell markers, enzymatic activity, or neurotransmitters and their receptors, and electrophysiological function.
  • Certain cells embodied in this invention have morphological features characteristic of neuronal cells or glial cells. The features are readily appreciated by those skilled in evaluating the presence of such cells. For example, characteristic of neurons are small cell bodies, and multiple processes pronounced of axons and dendrites. Cells of this invention can also be characterized according to whether they express phenotypic markers characteristic of neural cells of various kinds.
  • Markers of interest include but are not limited to ⁇ -tubulin III, microtubule-associated protein 2 (MAP-2), or neurofilament, characteristic of neurons; glial fibrillary acidic protein (GFAP), present in astrocytes; galactocerebroside (GaIC) or myelin basic protein (MBP), characteristic of oligodendrocytes; Oct-4, characteristic of undifferentiated hES cells; Nestin, characteristic of neural precursors and other cells; and both A2B5 and polysialylated NCAM, as already described. While A2B5 and NCAM are instructive markers when studying neural lineage cells, it should be appreciated that these markers can sometimes be displayed on other cell types, such as liver or muscle cells.
  • GFAP glial fibrillary acidic protein
  • GaIC galactocerebroside
  • MBP myelin basic protein
  • Nestin characteristic of undifferentiated hES cells
  • Nestin characteristic of neural precursors and other cells
  • A2B5 and NCAM are
  • ⁇ -Tubulin III was previously thought to be specific for neural cells, but it has been discovered that a subpopulation of hES cells is also ⁇ -tubulin III positive.
  • MAP-2 is a more stringent marker for fully differentiated neurons of various types.
  • Tissue-specific markers listed in this disclosure and known in the art can be detected using any suitable immunological technique—such as flow immunocytochemistry for cell-surface markers, immunohistochemistry (for example, of fixed cells or tissue sections) for intracellular or cell-surface markers, Western blot analysis of cellular extracts, and enzyme-linked immunoassay, for cellular extracts or products secreted into the medium.
  • suitable immunological technique such as flow immunocytochemistry for cell-surface markers, immunohistochemistry (for example, of fixed cells or tissue sections) for intracellular or cell-surface markers, Western blot analysis of cellular extracts, and enzyme-linked immunoassay, for cellular extracts or products secreted into the medium.
  • an antigen by a cell is said to be “antibody-detectable” if a significantly detectable amount of antibody will bind to the antigen in a standard immunocytochemistry or flow cytometry assay, optionally after fixation of the cells, and optionally using a labeled secondary antibody or other conjugate (such as a biotin-avidin conjugate) to amplify labeling.
  • a labeled secondary antibody or other conjugate such as a biotin-avidin conjugate
  • tissue-specific gene products can also be detected at the mRNA level by Northern blot analysis, dot-blot hybridization analysis, or by reverse transcriptase initiated polymerase chain reaction (RT-PCR) using sequence-specific primers in standard amplification methods. See U.S. Pat. No. 5,843,780 for further details. Sequence data for the particular markers listed in this disclosure can be obtained from public databases such as GenBank (URL www.ncbi.nlm.nih.gov: 80/entrez).
  • Expression at the mRNA level is said to be “detectable” according to one of the assays described in this disclosure if the performance of the assay on cell samples according to standard procedures in a typical controlled experiment results in clearly discernable hybridization or amplification product.
  • Expression of tissue-specific markers as detected at the protein or mRNA level is considered positive if the level is at least 2-fold, and preferably more than 10- or 50-fold above that of a control cell, such as an undifferentiated pPS cell, a fibroblast, or other unrelated cell type.
  • Evidence of synapse formation can be obtained by staining for synaptophysin.
  • Evidence for receptivity to certain neurotransmitters can be obtained by detecting receptors for ⁇ -amino butyric acid (GABA), glutamate, dopamine, 3,4-dihydroxyphenylalanine (DOPA), noradrenaline, acetylcholine, and serotonin.
  • GABA ⁇ -amino butyric acid
  • DOPA 3,4-dihydroxyphenylalanine
  • noradrenaline acetylcholine
  • serotonin serotonin.
  • Differentiation of particular neural precursor cell populations of this invention can generate cell populations that are at least 20%, 30%, or 40% MAP-2 positive.
  • a substantial proportion, say 5%, 10%, 25%, or more of the NCAM or MAP-2 positive cells will be capable of synthesizing a neurotransmitter, such as acetylcholine, glycine, glutamate, norepinephrine, serotonin, or GABA.
  • Certain populations of the invention contain NCAM or MAP-2 positive cells that are 0.1%, and possibly 1%, 3%, or 5% or more (on a cell count basis) that are positive for tyrosine hydroxylase (TH), measured by immunocytochemistry or mRNA expression. This generally considered in the art to be a marker for dopamine synthesizing cells.
  • the cells can be tested according to functional criteria. For example, calcium flux can be measured by any standard technique, in response to a neurotransmitter, or other environmental condition known to affect neurons in vivo.
  • a neurotransmitter or other environmental condition known to affect neurons in vivo.
  • neuron-like cells in the population are identified by morphological criteria, or by a marker such as NCAM.
  • the neurotransmitter or condition is then applied to the cell, and the response is monitored (Example 6).
  • the cells can also be subjected to standard patch-clamp techniques, to determine whether there is evidence for an action potential, and what the lag time is between applied potential and response.
  • Differentiation of neural precursor populations of this invention can generate cultures that contain subpopulations that have morphological characteristics of neurons, are NCAM or MAP-2 positive, and show responses with the following frequency: a response to GABA, acetylcholine, ATP, and high sodium concentration in at least about 40%, 60% or 80% of the cells; a response to glutamate, glycine, ascorbic acid, dopamine, or norepinephrine in at least about 5%, 10% or 20% of the cells.
  • a substantial proportion of the NCAM or MAP-2 positive cells (at least about 25%, 50%, or 75%) can also show evidence of an action potential in a patch-clamp system.
  • neural precursors have the ability to replicate in certain drug screening and therapeutic applications, and to provide a reservoir for the generation of differentiated neuronal and glial cells.
  • the cells of this invention can optionally be telomerized to increase their replication potential, either before or after they progress to restricted developmental lineage cells or terminally differentiated cells.
  • pPS cells that are telomerized may be taken down the differentiation pathway described earlier; or differentiated cells can be telomerized directly.
  • telomere catalytic component typically under a heterologous promoter that increases telomerase expression beyond what occurs under the endogenous promoter.
  • hTERT human telomerase
  • hTERT human telomerase
  • hTERT clones (WO 98/14592) are used as a source of hTERT encoding sequence, and spliced into an EcoRI site of a PBBS212 vector under control of the MPSV promoter, or into the EcoRi site of commercially available pBABE retrovirus vector, under control of the LTR promoter.
  • Differentiated or undifferentiated pPS cells are genetically altered using vector containing supernatants over a 8-16 h period, and then exchanged into growth medium for 1-2 days. Genetically altered cells are selected using 0.5-2.5 ⁇ g/mL puromycin, and recultured. They can then be assessed for hTERT expression by RT-PCR, telomerase activity (TRAP assay), immunocytochemical staining for hTERT, or replicative capacity.
  • TRAP assay telomerase activity
  • the following assay kits are available commercially for research purposes: TRAPeze® XL Telomerase Detection Kit (Cat. s7707; Intergen Co., Purchase N.Y.); and TeloTAGGG Telomerase PCR ELISApIus (Cat.
  • TERT expression can also be evaluated at the mRNA by RT-PCR.
  • Available commercially for research purposes is the LightCycler TeloTAGGG hTERT quantification kit (Cat. 3,012,344; Roche Diagnostics). Continuously replicating colonies will be enriched by further culturing under conditions that support proliferation, and cells with desirable phenotypes can optionally be cloned by limiting dilution.
  • pPS cells are differentiated into multipotent or committed neural precursors, and then genetically altered to express TERT.
  • pPS cells are genetically altered to express TERT, and then differentiated into neural precursors or terminally differentiated cells.
  • Successful modification to increase TERT expression can be determined by TRAP assay, or by determining whether the replicative capacity of the cells has improved.
  • telomeres are of particular interest in applications of this invention where it is advantageous to have cells that can proliferate and maintain their karyotype—for example, in pharmaceutical screening, and in therapeutic protocols where differentiated cells are administered to an individual in order to augment CNS function.
  • This invention provides a method to produce large numbers of neural precursor cells and mature neuronal and glial cells. These cell populations can be used for a number of important research, development, and commercial purposes.
  • the cells of this invention can be used to prepare a cDNA library relatively uncontaminated with cDNA preferentially expressed in cells from other lineages.
  • multipotent neural progenitor cells are collected by centrifugation at 1000 rpm for 5 min, and then mRNA is prepared from the pellet by standard techniques (Sambrook et al., supra). After reverse transcribing into cDNA, the preparation can be subtracted with cDNA from any or all of the following cell types: cells committed to the neuronal or glial cell lineage, mature neurons, astrocytes, oligodendrocytes, or other cells of undesired specificity.
  • cDNA libraries can be made that represent transcripts preferentially expressed in neuronal or glial precursors, or mature neurons, astrocytes, and oligodendrocytes.
  • the differentiated cells of this invention can also be used to prepare antibodies that are specific for markers of multipotent neural progenitors, cells committed to the neuronal or glial cell lineage, and mature neurons, astrocytes, and oligodendrocytes.
  • This invention provides an improved way of raising such antibodies because cell populations are enriched for particular cell types compared with pPS cell cultures, and neuronal or glial cell cultures extracted directly from CNS tissue.
  • Polyclonal antibodies can be prepared by injecting a vertebrate animal with cells of this invention in an immunogenic form. Production of monoclonal antibodies is described in such standard references as Harrow & Lane (1988), U.S. Pat. Nos. 4,491,632, 4,472,500 and 4,444,887, and Methods in Enzymology 73B: 3 (1981). Other methods of obtaining specific antibody molecules (optimally in the form of single-chain variable regions) involve contacting a library of immunocompetent cells or viral particles with the target antigen, and growing out positively selected clones. See Marks et al., New Eng. J. Med.
  • the cells of this invention are also of interest in identifying expression patterns of transcripts and newly synthesized proteins that are characteristic for neural precursor cells, and may assist in directing the differentiation pathway or facilitating interaction between cells.
  • Expression patterns of the differentiated cells are obtained and compared with control cell lines, such as undifferentiated pPS cells, other types of committed precursor cells (such as pPS cells differentiated towards other lineages, cells committed to the neuronal or glial cell lineage), other types of putative neural stem cells such as those obtained from neural crest, neurospheres, or spinal chord, or terminally differentiated cells, such as mature neurons, astrocytes, oligodendrocytes, smooth muscle cells, and Schwann cells.
  • control cell lines such as undifferentiated pPS cells, other types of committed precursor cells (such as pPS cells differentiated towards other lineages, cells committed to the neuronal or glial cell lineage), other types of putative neural stem cells such as those obtained from neural crest, neurospheres, or spinal chord, or terminally differentiated
  • Suitable methods for comparing expression at the protein level include the immunoassay or immunohistochemistry techniques described above. Suitable methods for comparing expression at the level of transcription include methods of differential display of mRNA (Liang, Peng, et al., Cancer Res. 52:6966, 1992), whole-scale sequencing of cDNA libraries, and matrix array expression systems.
  • microarray in analyzing gene expression is reviewed generally by Fritz et al Science 288:316, 2000; “ Microarray Biochip Technology”, L Shi, www.Gene-Chips.com.
  • Systems and reagents for performing microarray analysis are available commercially from companies such as Affymetrix, Inc., Santa Clara CA; Gene Logic Inc., Columbia MD; HySeq Inc., Sunnyvale CA; Molecular Dynamics Inc., Sunnyvale CA; Nanogen, San Diego CA; and Synteni Inc., Fremont CA (acquired by Incyte Genomics, Palo Alto CA).
  • Solid-phase arrays are manufactured by attaching the probe at specific sites either by synthesizing the probe at the desired position, or by presynthesizing the probe fragment and then attaching it to the solid support (U.S. Pat. Nos. 5,474,895 and 5,514,785).
  • the probing assay is typically conducted by contacting the array by a fluid potentially containing the nucleotide sequences of interest under suitable conditions for hybridization conditions, and then determining any hybrid formed.
  • Microarrays are prepared by first amplifying cDNA fragments encoding marker sequences to be analyzed, and spotted directly onto glass slides To compare mRNA preparations from two cells of interest, one preparation is converted into Cy3-labeled cDNA, while the other is converted into Cy5-labeled cDNA. The two cDNA preparations are hybridized simultaneously to the microarray slide, and then washed to eliminate non-specific binding. he slide is then scanned at wavelengths appropriate for each of the labels, the resulting fluorescence is quantified, and the results are formatted to give an indication of the relative abundance of mRNA for each marker on the array.
  • Identifying expression products for use in characterizing and affecting differentiated cells of this invention involves analyzing the expression level of RNA, protein, or other gene product in a first cell type, such as a pluripotent neuronal precursor cell of this invention, or a cell capable of differentiating along the neuronal or glial pathway; then analyzing the expression level of the same product in a control cell type; comparing the relative expression level between the two cell types, (typically normalized by total protein or RNA in the sample, or in comparison with another gene product expected to be expressed at a similar level in both cell types, such as a house-keeping gene); and then identifying products of interest based on the comparative expression level.
  • a first cell type such as a pluripotent neuronal precursor cell of this invention, or a cell capable of differentiating along the neuronal or glial pathway
  • Neural precursor cells of this invention can be used to screen for factors (such as solvents, small molecule drugs, peptides, polynucleotides) or environmental conditions (such as culture conditions or manipulation) that affect the characteristics of neural precursor cells and their various progeny.
  • factors such as solvents, small molecule drugs, peptides, polynucleotides
  • environmental conditions such as culture conditions or manipulation
  • pPS cells (undifferentiated or differentiated) are used to screen factors that promote maturation into neural cells, or promote proliferation and maintenance of such cells in long-term culture. For example, candidate maturation factors or growth factors are tested by adding them to cells in different wells, and then determining any phenotypic change that results, according to desirable criteria for further culture and use of the cells.
  • screening applications of this invention relate to the testing of pharmaceutical compounds for their effect on neural tissue or nerve transmission. Screening may be done either because the compound is designed to have a pharmacological effect on neural cells, or because a compound designed to have effects elsewhere may have unintended side effects on the nervous system.
  • the screening can be conducted using any of the neural precursor cells or terminally differentiated cells of the invention, such as dopaminergic, serotonergic, cholinergic, sensory, and motor neurons, oligodendrocytes, and astrocytes.
  • Cytotoxicity can be determined in the first instance by the effect on cell viability, survival, morphology, and the expression of certain markers and receptors. Effects of a drug on chromosomal DNA can be determined by measuring DNA synthesis or repair. [ 3 H]-thymidine or BrdU incorporation, especially at unscheduled times in the cell cycle, or above the level required for cell replication, is consistent with a drug effect. Unwanted effects can also include unusual rates of sister chromatid exchange, determined by metaphase spread. The reader is referred to A. Vickers (pp 375-410 in “In vitro Methods in Pharmaceutical Research,” Academic Press, 1997) for further elaboration.
  • Effect of cell function can be assessed using any standard assay to observe phenotype or activity of neural cells, such as receptor binding, neurotransmitter synthesis, release or uptake, electrophysiology, and the growing of neuronal processes or myelin sheaths—either in cell culture or in an appropriate model.
  • This invention also provides for the use of neural precursor cells to restore a degree of central nervous system (CNS) function to a subject needing such therapy, perhaps due to an inborn error in function, the effect of a disease condition, or the result of an injury.
  • CNS central nervous system
  • the cells can first be tested in a suitable animal model. At one level, cells are assessed for their ability to survive and maintain their phenotype in vivo. Neural precursor cells are administered to immunodeficient animals (such as nude mice, or animals rendered immunodeficient chemically or by irradiation) at an observable site, such as in the cerebral cavity or in the spinal chord. Tissues are harvested after a period of a few days to several weeks or more, and assessed as to whether pPS derived cells are still present.
  • immunodeficient animals such as nude mice, or animals rendered immunodeficient chemically or by irradiation
  • a detectable label such as green fluorescent protein, or ⁇ -galactosidase
  • a constitutive cell marker for example, using human-specific antibody
  • the presence and phenotype of the administered cells can be assessed by immunohistochemistry or ELISA using human-specific antibody, or by RT-PCR analysis using primers and hybridization conditions that cause amplification to be specific for human polynucleotide sequences.
  • Suitable markers for assessing gene expression at the mRNA or protein level are provided elsewhere in this disclosure.
  • Differentiated cells of this invention can also be used for tissue reconstitution or regeneration in a human patient in need thereof.
  • the cells are administered in a manner that permits them to graft or migrate to the intended tissue site and reconstitute or regenerate the functionally deficient area.
  • Certain neural progenitor cells embodied in this invention are designed for treatment of acute or chronic damage to the nervous system.
  • excitotoxicity has been implicated in a variety of conditions including epilepsy, stroke, ischemia, Huntington's disease, Parkinson's disease and Alzheimer's disease.
  • Certain differentiated cells of this invention may also be appropriate for treating dysmyelinating disorders, such as Pelizaeus-Merzbacher disease, multiple sclerosis, leukodystrophies, neuritis and neuropathies. Appropriate for these purposes are cell cultures enriched in oligodendrocytes or oligodendrocyte precursors to promote remyelination.
  • neural stem cells are transplanted directly into parenchymal or intrathecal sites of the central nervous system, according to the disease being treated. Grafts are done using single cell suspension or small aggregates at a density of 25,000-500,000 cells per ⁇ L (U.S. Pat. No. 5,968,829). The efficacy of transplants of motor neurons or their precursors can be assessed in a rat model for acutely injured spinal cord as described by McDonald et al . (Nat. Med. 5:1410, 1999).
  • a successful transplant will show transplant-derived cells present in the lesion 2-5 weeks later, differentiated into astrocytes, oligodendrocytes, and/or neurons, and migrating along the cord from the lesioned end, and an improvement in gate, coordination, and weight-bearing.
  • the neural progenitor cells and terminally differentiated cells according to this invention can be supplied in the form of a pharmaceutical composition, comprising an isotonic excipient prepared under sufficiently sterile conditions for human administration.
  • a pharmaceutical composition comprising an isotonic excipient prepared under sufficiently sterile conditions for human administration.
  • the reader is referred to Cell Therapy. Stem Cell Transplantation, Gene Therapy, and Cellular Immunotherapy , by G. Morstyn & W. Sheridan eds, Cambridge University Press, 1996; and Hematopoletic Stem Cell Therapy, E. D. Ball, J. Lister & P. Law, Churchill Livingstone, 2000.
  • composition may optionally be packaged in a suitable container with written instructions for a desired purpose, such as the reconstitution of CNS function to improve some neurological abnormality.
  • hES cells are maintained either on primary mouse embryonic fibroblasts, or in a feeder-free system.
  • the hES cells are seeded as small clusters either on irradiated mouse embryonic fibroblasts, or on plates coated with Matrigel® (1:10 to 1:30 in culture medium).
  • hES cell cultures on feeder cells are maintained in a medium composed of 80% KO DMEM (Gibco) and 20% Serum Replacement (Gibco), supplemented with 1% non-essential amino acids, 1 mM glutamine, 0.1 mM ⁇ -mercaptoethanol and 4 ng/mL human bFGF (Gibco).
  • Cultures free of feeder cells are maintained in the same medium that has previously been conditioned by culturing with embryonic fibroblasts, and resupplemented with 4 ng/mL bFGF (replaced daily).
  • Cells are expanded by serial passaging.
  • the monolayer culture of ES colonies is treated with 1 mg/mL collagenase for 5-20 minutes at 37° C.
  • the cultures are then gently scraped to remove the cells.
  • the clusters are gently dissociated, and replated as small clusters onto fresh feeder cells.
  • Embryoid bodies are produced as follows. Confluent monolayer cultures of hES cells are harvested by incubating in 1 mg/mL collagenase for 5-20 min, following which the cells are scraped from the plate. The cells are then dissociated into clusters and plated in non-adherent cell culture plates (Costar) in a medium composed of 80% KO (“knockout”) DMEM (Gibco) and 20% non-heat-inactivated FBS (Hyclone), supplemented with 1% non-essential amino acids, 1 mM glutamine, 0.1 mM ⁇ -mercaptoethanol. The cells are seeded at a 1:1 or 1:2 ratio in 2 mL medium per well (6 well plate). The EBs are fed every other day by the addition of 2 mL of medium per well.
  • the EBs are collected and resuspended in fresh medium. After 4-8 days in suspension, the EBs are plated onto a substrate and allowed to differentiate further, in the presence of selected differentiation factors.
  • fibronectin (Sigma) at a final concentration of 20 ⁇ g/mL in PBS. Using 1 mL/well (9.6 cm 2 ), plates are incubated at 4° C. overnight or at room temperature for 4 h. The fibronectin is then removed, and the plates are washed with PBS or KO DMEM once before use.
  • Immunocytochemistry for NCAM and A2B5 expression is conducted as follows: Live cells are incubated in primary antibody diluted in culture medium with 1% goat serum for 15 minutes at 37° C., washed once with medium, and then incubated with labeled secondary antibody for 15 min. After washing, the cells are then fixed for 15-20 min in 2% paraformaldehyde. For other markers, cultures are fixed for 10-20 min with 4% paraformaldehyde in PBS, washed 3 times with PBS, permeabilized for 2 min in 100% ethanol, and washed with 0.1 M PBS. Cultures are then incubated in a blocking solution of 0.1 M PBS with 5% NGS (normal goat serum) for at least 1 hour at room temperature.
  • NGS normal goat serum
  • Cultures are then incubated in primary antibody diluted in 0.1M PBS containing 1% NGS for at least 2 h at room temperature. They are then washed in PBS before a 30 min incubation with secondary antibody in the same buffer. Antibodies used include those shown in Table 1.
  • Bead immunosorting is conducted using the following reagents and equipment: magnetic cell separator; Midi MACsTM column; buffer of PBS CMF containing 0.5% BSA and 2 mM EDTA; primary antibody against NCAM or A2B5; rat anti-mouse IgG (or IgM) microbeads; pre-separation filter; rat anti-mouse kappa PE; and a FACScan device.
  • Cells are harvested using trypsinlEDTA (Gibco) and dissociated. After removing the trypsin, the cells are resuspended in MACsTM buffer.
  • Cells are then labeled with primary antibody for 6-8 min at room temp., and washed 2 times in MACsTM buffer by spinning cells at 300 ⁇ g for 10 min and aspirating the buffer. The cells are then resuspended in 80 ⁇ I (minimum vol.) per 10 7 cells. 20 ⁇ I (minimum vol.) MACs ramTM IgG microbeads per 10 7 cells are added for 15 min at 6-12° C. The sample is then washed 2 times in MACsTM buffer before magnetic separation. With the column in the magnetic cell separator, the cell suspension is applied to the column (LS+Midi) in ⁇ 3-5 mL buffer. Negative cells are passed through by washing 3 times with 3 mL of MACsTM buffer. The column is then removed from the magnetic field, and positive cells are eluted with 5 mL of MACsTM buffer.
  • A2B5+ or NCAM+ cells are maintained on plates coated with poly-lysine and laminin in DMEM/F12 (Biowhittaker) supplemented with N2 (Gibco 17502-014), B27 (Gibco 17504-010) and the factors indicated.
  • Source of the factors is shown in Table 2.
  • TABLE 2 Factors used for Neural Cell Culture Working Growth Factor Source Concentration human EGF R & D Systems 10 ng/mL human bFGF Gibco 10-25 ng/mL human CNTF R & D Systems 1-10 ng/mL human PDGF R & D Systems 1 ng/mL human IGF-I R & D Systems 1 ng/mL
  • RNA is denatured for 10 min at 70° C., followed by annealing at room temperature for 10 min.
  • dNTPs are added at a final concentration of 1 mM along with 0.5 ⁇ L of Superscript II RT (Gibco), incubated at 42 ° C. for 50 minutes, and then heat-inactivated at 80 ° C. for 10 min. Samples are then stored at ⁇ 20 ° C. till they are processed for PCR analysis.
  • PCR Standard polymerase chain reaction
  • cDNA 1.0, ⁇ L, 10 ⁇ PCR buffer (Gibco) 2.5 ⁇ L, 10 ⁇ MgCI 2 2.5 ⁇ L, 2.5 mM dNTP 3.0 ⁇ L, 5 ⁇ M 3′-primer 1.0 ⁇ L, 5 ⁇ M 5′-primer, 1.0 ⁇ L, Taq 0.4, ⁇ L, DEPC-water 13.6 ⁇ L.
  • This experiment focused on determining whether the human embryonic stem cells (hES) could undergo directed differentiation to NCAM-positive progenitor cells.
  • the hES cells were harvested either from mEF-supported cultures or feeder-free cultures, and then differentiated via embryoid body (EB) formation in suspension culture using medium containing 20% FBS.
  • the EBs were then plated intact onto fibronectin in DMEM/F12 medium, supplemented with N2 supplement (Gibco) and 25 ng/mL human bFGF. After culturing for about 2-3 days, NCAM-positive cells and A2B5-positive cells were identified by immunostaining.
  • NCAM positive cells retrieved from the sort were plated on poly-L-lysine/laminin in DMEM/F12 with N2 and B27 supplements and 2 mg/mL BSA, 10 ng/mL human CNTF, 10 ng/mL human bFGF and 1 ng/mL human NT-3.
  • cells were maintained in DMEM/F12 with N2 and B27 supplements and 10 ng/mL EGF, 10 ng/mL bFGF, 1 ng/mL PDGF, and 1 ng/mL IGF-1.
  • FIG. 1 (Upper Panel) shows the growth curves for the NCAM positive cells.
  • the cells studied in this experiment were prepared by forming embryoid bodies in 20% FBS for 4 days in suspension, then plating onto a fibronectin matrix in DMEM/F12 with N2 and B27 supplements and 25 ng/mL bFGF for 2-3 days. The cells were then positively sorted for NCAM expression, and maintained in a medium containing CNTF, bFGF, and NT3. The sorted cells did not show increased survival relative to the unsorted population. It was found that some of the NCAM positive cells also express ⁇ -tubulin 111 , indicating that these cells have the capacity to form neurons. They also had morphology characteristic of neuronal cells.
  • A2B5 positive cells within this population, which may represent glial progenitor cells.
  • GFAP a marker for astrocytes.
  • this cell population proliferated in culture, the proportion of NCAM positive cells (and the capacity to form neurons) diminished after several passages.
  • FIG. 2 shows an exemplary procedure for obtaining A2B5-positive cells.
  • FIG. 1 (Lower Panel) shows the growth curves for the sorted A2B5-positive cells.
  • the cells were maintained in the same media formulation on poly-l-lysine coated plates. The cells proliferate when serially passaged.
  • A2B5-positive cells were induced to differentiate by the addition of forskolin. These cells have been assessed through different culture passages, as shown in Table 5. TABLE 5 Phenotypic Features of Mature Neural Cells Neuron-like No. of passages Method of morphology Cells Staining Positively for: after A2B5 sort Maturation visible ⁇ -tubulin GFAP GalC A2B5 NCAM 1 PICNT + Fk yes 38 ⁇ 9% 13 ⁇ 7% 79 ⁇ 3% 28 ⁇ 6% 4 days 3 PICNT + Fk yes +++ + +++++ ++ 2 days 7 +/ ⁇ EF yes + + ++ +++ ⁇ +/ ⁇ serum
  • H1 a genetically altered line designated H7NHG.
  • H7NHG a genetically altered line carries an expression cassette that permits the cells to constitutively express green fluorescent protein (GFP).
  • GFP green fluorescent protein
  • Neonatal Sprague Dawley rats received unilateral intrastriatal implants of one of the following cell populations:
  • Control animals received grafts of irradiated mouse embryonic fibroblasts upon which the undifferentiated hES cells were maintained. To determine if cell proliferation occurred after grafting, some animals were pulsed with intraperitoneal injections of BrdU, commencing 48 h prior to sacrifice. Fourteen days after transplantation, the rats were transcardially perfused with 4% paraformaldehyde and the tissue was processed for immunohistochemical analysis.
  • FIG. 3 shows the fluorescence observed in sections from animals administered cells expressing GFP.
  • the first stage of differentiation was induced by forming embryoid bodies in FBS medium with or without 10 ⁇ M retinoic acid (RA). After 4 days in suspension, embryoid bodies were plated onto fibronectin-coated plates in defined medium supplemented with 10 ng/mL human EGF, 10 ng/mL human bFGF, 1 ng/mL human PDGF-AA, and 1 ng/mL human IGF-1. The embryoid bodies adhered to the plates, and cells began to migrate onto the plastic, forming a monolayer.
  • RA retinoic acid
  • neuronal morphology After 3 days, many cells with neuronal morphology were observed. The neural precursors were identified as cells positive for BrdU incorporation, nestin staining, and the absence of lineage specific differentiation markers. Putative neuronal and glial progenitor cells were identified as positive for polysialylated NCAM and A2B5. Forty one to sixty percent of the cells expressed NCAM, and 20-66% expressed A2B5, as measured by flow cytometry. A subpopulation of the NCAM-positive cells was found to express ⁇ -tubulin III and MAP-2. There was no co-localization with glial markers such as GFAP or GaIC. The A2B5 positive cells appeared to generate both neurons and glia.
  • the cell populations were further differentiated by replating the cells in a medium containing none of the mitogens, but containing 10 ng/mL Neurotrophin-3 (NT-3) and 10 ng/mL brain-derived neurotrophic factor (BDNF). Neurons with extensive processes were seen after about 7 days. Cultures derived from embryoid bodies maintained in retinoic acid (RA) showed more MAP-2 positive cells ( ⁇ 26%) than those maintained without RA ( ⁇ 5%). GFAP positive cells were seen in patches. GaIC positive cells were identified, but the cells were large and flat rather than having complex processes.
  • RA retinoic acid
  • GABA-immunoreactive cells were identified that co-expressed ⁇ -tubulin IlIl or MAP2, and had morphology characteristic of neuronal cells. Occasional GABA-positive cells were identified that did not co-express neuronal markers, but had an astrocyte-like morphology. Neuronal cells were identified that expressed both tyrosine hydroxylase (TH) and MAP-2. Synapse formation was identified by staining with synaptophysin antibody.
  • TH tyrosine hydroxylase
  • FIG. 4 shows TH staining in cultures differentiated from the H9 line of human ES cells.
  • Embryoid bodies were maintained in 10 ⁇ M retinoic acid for 4 days, then plated onto fibronectin coated plates in EGF, basic FGF, PDGF and IGF for 3 days. They were next passaged onto laminin in N2 medium supplemented with 10 ng/mL NT-3 and 10 ng/mL BDNF, and allowed to differentiate further for 14 days.
  • the differentiated cells were fixed with 2% formaldehyde for 20 min at room temp, and then developed using antibody to TH, a marker for dopaminergic cells.
  • Standard fura-2 imaging of calcium flux was used to investigate the functional properties of the hES cell derived neurons.
  • Neurotransmitters studied included GABA, glutamate (E), glycine (G), elevated potassium (50 mM K + instead of 5 mM K + ), ascorbic acid (control), dopamine, acetylcholine (ACh) and norepinephrine.
  • the solutions contained 0.5 mM of the neurotransmitter (except ATP at 10 pM) in rat Ringers (RR) solution: 140 mM NaCI, 3 mM KCI, 1 mM MgCI 2 , 2 mM CaCI 2 , 10 mM HEPES buffer, and 10 mM glucose.
  • FIG. 5 shows the response of neural-restricted precursors to various neurotransmitters.
  • Panel A shows the ratio of emission data from single cells on two different coverslips. Addition of the neurotransmitters is indicated above by labeled triangles.
  • Panel B shows the frequency of cells tested that responded to specific neurotransmitters.
  • Panel C shows the combinations of neurotransmitter responses observed. Of the 53 cells tested, 26 responded to GABA, acetylcholine, ATP and elevated potassium. Smaller subsets of the population responded to other combinations of agonists. Only 2 of the cells failed to respond to any of the agonists applied.
  • Standard whole-cell patch-clamp technique was conducted on the hES cell derived neurons, to record ionic currents generated in voltage-clamp mode and the action potential generated in current-clamp mode.
  • the external bath solution was rat Ringers solution (Example 6).
  • the internal solution was 75 mM potassium-aspartate, 50 mM KF, 15 mM NaCI, 11 mM EGTA, and 10 mM HEPES buffer, set to pH 7.2 using KOH.
  • FIG. 6 shows results from a typical experiment.
  • Panel A shows sodium and potassium currents observed in two cells depolarized to test potentials between ⁇ 80 and 80 mV from a holding potential of ⁇ 100 mV.
  • Panel B shows the inward (Na + ) and outward (K + ) peak current-voltage relationships observed.
  • Sodium current activates between ⁇ 30 and 0 mV, reaching a peak at ⁇ 10 or 0 mV.
  • Potassium current activates above ⁇ 10 mV, becoming equal or larger in magnitude than the sodium current at voltages between 20 and 40 mV.
  • Panel C shows action potentials generated by the same cells n response to depolarizing stimuli. Cell membranes were held at voltages between ⁇ 60 and ⁇ 100 mV in ⁇ 80 or ⁇ 150 pA of current, and depolarized for short durations
  • Embryoid bodies were cultured in suspension with 10 ⁇ M retinoic acid for 4 days, then plated into defined medium supplemented with EGF, bFGF, PDGF, and IGF-1 for 3-4 days. Cells were then separated by magnetic bead sorting or immunopanning into A2B5-positive or NCAM-positive enriched populations.
  • the immuno-selected cells were maintained in defined medium supplemented with 10 ng/mL NT-3 and 10 ng/mL BDNF. After 14 days, 25 ⁇ 4% of the NCAM-sorted cells were MAP-2 positive—of which 1.9 ⁇ 0.8% were GABA-positive, and 3 ⁇ 1% were positive for tyrosine hydroxylase (TH): the rate-limiting enzyme for dopamine synthesis, generally considered to be representative of dopamine-synthesizing cells.
  • TH tyrosine hydroxylase
  • glial markers such as GFAP or GaIC.
  • Cells sorted for A2B5 have the capacity to generate both neurons and astrocytes. After the enrichment, the cells were placed into defined media supplemented with NT-3 and BDNF and allowed to differentiate for 14 days. Within the first 1-2 days after plating, cells in the A2B5 enriched population began to extend processes. After two weeks, cells took on the morphology of mature neurons, and 32 ⁇ 3% of the cells were MAP-2 positive. Importantly, 3 ⁇ 1% of the MAP-2 cells were TH-positive, while only 0.6 ⁇ 0.3% were GABA immunoreactive. These data indicate that a population of cells can be obtained from hES cells that comprise progenitors for both astrocytes and neurons, including those that synthesize dopamine.
  • Embryoid bodies were generated from confluent hES cells of the H7 line at passage 32 by incubating in 1 mg/mL collagenase (37° C., 5-20 min), scraping the dish, and placing the cells into non-adherent culture plates (Costar®).
  • the resulting EBs were cultured in suspension in media containing FBS and 10 ⁇ M all-trans retinoic acid. After four days, the aggregates were collected and allowed to settle in a centrifuge tube.
  • the supernatant was then aspirated, and the aggregates were plated onto poly L-lysine and fibronectin coated plates in proliferation medium (DMEM/F12 1:1 supplemented with N2, half-strength B27, 10 ng/mL EGF (R&D Systems), 10 ng/mL bFGF (Gibco), 1 ng/mL PDGF-AAA (R&D Systems), and 1 ng/mL IGF-1 (R & D Systems).
  • proliferation medium DMEM/F12 1:1 supplemented with N2, half-strength B27, 10 ng/mL EGF (R&D Systems), 10 ng/mL bFGF (Gibco), 1 ng/mL PDGF-AAA (R&D Systems), and 1 ng/mL IGF-1 (R & D Systems).
  • the EBs were allowed to attach and proliferate for three days; then collected by trypsinizing ⁇ 1 min (Sigma) and plated at 1.5 ⁇ 10 5 cells/well onto poly l-lysine and laminin coated 4-well chamber slides in proliferation medium for one day. The medium was then changed to Neural Basal medium supplemented with B27, and one of the following growth cocktails:
  • Results are shown in Table 7. Initial culturing in bFGF, BDNF and NT-3 yielded the highest proportion of TH positive cells. TABLE 7 Conditions for Producing Dopaminergic Neurons Culture conditions % MAP-2 % MAP-2 cells that are days 1-6 days 6-12 positive TH positive B, N, F B, N 26% 5.5% B, N, F CA, AA 35% 4.0% B, N, F CA, AA, B, N 25% 8.7% F, F8, S B, N 37% 3.7% F, F8, S CA, AA 34% 3.9% F, F8, S CA, AA, B, N 21% 5.8% F B, N 28% 3.5% F CA, AA 26% 4.1% F CA, AA, B, N 22% 5.7%

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Biotechnology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Immunology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Psychology (AREA)
  • Pain & Pain Management (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Psychiatry (AREA)
US09/859,351 2000-05-17 2001-05-16 Neural progenitor cell populations Abandoned US20020009743A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US09/859,351 US20020009743A1 (en) 2000-05-17 2001-05-16 Neural progenitor cell populations
US09/872,183 US6833269B2 (en) 2000-05-17 2001-05-31 Making neural cells for human therapy or drug screening from human embryonic stem cells
US10/157,288 US7250294B2 (en) 2000-05-17 2002-05-28 Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
US10/873,414 US20050042749A1 (en) 2001-05-16 2004-06-21 Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
US11/009,504 US7763463B2 (en) 2000-05-17 2004-12-10 Use of cyclic AMP and ascorbic acid to produce dopaminergic neurons from embryonic stem cells
US11/281,040 US8148148B2 (en) 2000-05-17 2005-11-16 Neural progenitor cell populations
US12/332,783 US8252585B2 (en) 2000-05-17 2008-12-11 Neural progenitor cell populations
US13/558,078 US9803174B2 (en) 2000-05-17 2012-07-25 Neural progenitor cell populations
US14/217,699 US20140242691A1 (en) 2000-05-17 2014-03-18 Neural Progenitor Cell Populations

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US20560000P 2000-05-17 2000-05-17
US25760800P 2000-12-22 2000-12-22
US09/859,351 US20020009743A1 (en) 2000-05-17 2001-05-16 Neural progenitor cell populations

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/872,183 Continuation-In-Part US6833269B2 (en) 2000-05-17 2001-05-31 Making neural cells for human therapy or drug screening from human embryonic stem cells

Related Child Applications (6)

Application Number Title Priority Date Filing Date
PCT/US2001/015861 Division WO2001088104A2 (en) 2000-05-17 2001-05-16 Neural progenitor cell populations
US09/872,183 Continuation-In-Part US6833269B2 (en) 2000-05-17 2001-05-31 Making neural cells for human therapy or drug screening from human embryonic stem cells
US09/872,183 Division US6833269B2 (en) 2000-05-17 2001-05-31 Making neural cells for human therapy or drug screening from human embryonic stem cells
US10/157,288 Continuation-In-Part US7250294B2 (en) 2000-05-17 2002-05-28 Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
US10/873,414 Continuation-In-Part US20050042749A1 (en) 2000-01-11 2004-06-21 Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
US11/281,040 Continuation US8148148B2 (en) 2000-05-17 2005-11-16 Neural progenitor cell populations

Publications (1)

Publication Number Publication Date
US20020009743A1 true US20020009743A1 (en) 2002-01-24

Family

ID=26900583

Family Applications (6)

Application Number Title Priority Date Filing Date
US09/859,351 Abandoned US20020009743A1 (en) 2000-05-17 2001-05-16 Neural progenitor cell populations
US09/872,183 Expired - Lifetime US6833269B2 (en) 2000-05-17 2001-05-31 Making neural cells for human therapy or drug screening from human embryonic stem cells
US11/281,040 Expired - Fee Related US8148148B2 (en) 2000-05-17 2005-11-16 Neural progenitor cell populations
US12/332,783 Expired - Lifetime US8252585B2 (en) 2000-05-17 2008-12-11 Neural progenitor cell populations
US13/558,078 Expired - Fee Related US9803174B2 (en) 2000-05-17 2012-07-25 Neural progenitor cell populations
US14/217,699 Abandoned US20140242691A1 (en) 2000-05-17 2014-03-18 Neural Progenitor Cell Populations

Family Applications After (5)

Application Number Title Priority Date Filing Date
US09/872,183 Expired - Lifetime US6833269B2 (en) 2000-05-17 2001-05-31 Making neural cells for human therapy or drug screening from human embryonic stem cells
US11/281,040 Expired - Fee Related US8148148B2 (en) 2000-05-17 2005-11-16 Neural progenitor cell populations
US12/332,783 Expired - Lifetime US8252585B2 (en) 2000-05-17 2008-12-11 Neural progenitor cell populations
US13/558,078 Expired - Fee Related US9803174B2 (en) 2000-05-17 2012-07-25 Neural progenitor cell populations
US14/217,699 Abandoned US20140242691A1 (en) 2000-05-17 2014-03-18 Neural Progenitor Cell Populations

Country Status (11)

Country Link
US (6) US20020009743A1 (ja)
EP (1) EP1287116A2 (ja)
JP (5) JP5943533B2 (ja)
KR (1) KR100903755B1 (ja)
CN (1) CN100580079C (ja)
AU (2) AU2001263199B2 (ja)
CA (1) CA2409698C (ja)
GB (1) GB2379447B (ja)
HK (1) HK1055765A1 (ja)
IL (2) IL152741A0 (ja)
WO (1) WO2001088104A2 (ja)

Cited By (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003000868A1 (en) * 2001-06-21 2003-01-03 Geron Corporation Dopaminergic neurons and proliferation-competent precursor cells for treating parkinson's disease
US20030087263A1 (en) * 1999-12-16 2003-05-08 Renu Wadhwa Method of screening remedy for cancer with the use of interaction domain of p53 and mortalin
WO2003089580A2 (en) * 2002-04-16 2003-10-30 Dana-Farber Cancer Institute, Inc. Cancer models
US20040224887A1 (en) * 2002-07-16 2004-11-11 Thomas Jessel Systems and methods for screening for modulators of neural differentiation
US20040253719A1 (en) * 2003-03-07 2004-12-16 Goldman Steven A. Identification and Isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain
US20050042749A1 (en) * 2001-05-16 2005-02-24 Carpenter Melissa K. Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
US20050095706A1 (en) * 2001-10-03 2005-05-05 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20050095702A1 (en) * 2003-09-03 2005-05-05 Reliance Life Sciences Pvt. Ltd. In vitro generation of GABAergic neurons from embryonic stem cells and their use in the treatment of neurological disorders
WO2005021720A3 (en) * 2003-08-29 2005-05-19 Wisconsin Alumni Res Found Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem 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
US20050181503A1 (en) * 2004-02-13 2005-08-18 Goldman Steven A. Purines are self-renewal signals for neural stem cells, and purine receptor antagonists promote neuronal and glial differentiation therefrom
US20050265983A1 (en) * 2002-11-17 2005-12-01 Eldad Melamed Methods, nucleic acid constructs and cells for treating neurodegenerative disorders
US20060078545A1 (en) * 2000-05-17 2006-04-13 Carpenter Melissa K Neural progenitor cell populations
US20060211109A1 (en) * 2003-03-12 2006-09-21 Reliance Life Sciences Pvt. Ltd. Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells
US20070224650A1 (en) * 2002-07-16 2007-09-27 Thomas Jessell Systems and methods for screening for modulators of neural differentiation
US20080096273A1 (en) * 2004-09-06 2008-04-24 Yasushi Kondo Method for Preparing Conditioned Medium of Astrocyte-Like Cells
US20080227137A1 (en) * 2001-10-03 2008-09-18 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20090010895A1 (en) * 2005-06-16 2009-01-08 Ramot At Tel Aviv University Ltd. Isolated cells and populations comprising same for the treatment of CNS diseases
US20090175835A1 (en) * 2006-02-07 2009-07-09 Korea Institute Of Radiological & Medical Sciences Composition for treating damage of central or peripheral nerve system
US7560281B2 (en) 2000-01-11 2009-07-14 Geron Corporation Use of TGF beta superfamily antagonists to make dopaminergic neurons from embryonic stem cells
US20100021434A1 (en) * 2005-12-08 2010-01-28 Ramot At Tel Aviv University Ltd. Isolated Oligodendrocyte-Like Cells and Populations Comprising Same for the Treatment of CNS Diseases
JP2010158242A (ja) * 2001-06-21 2010-07-22 Geron Corp パーキンソン病を治療するためのドーパミン作動性ニューロンおよび増殖能のある前駆細胞
US20100196332A1 (en) * 2008-12-10 2010-08-05 Hynek Wichterle Generation of brachial, thoracic and lumbar spinal motor neurons from embryonic stem cells in the absence of all-trans retinoic acid supplement
US20100267073A1 (en) * 2007-04-13 2010-10-21 Serge Przedborski Stem cell-based culture system for drug development
US20140051085A1 (en) * 2011-03-01 2014-02-20 The Scripps Research Institute Direct reprogramming of human fibroblasts to functional neurons under defined conditions
US8663987B2 (en) 2008-05-28 2014-03-04 Ramot At Tel-Aviv University Ltd. Mesenchymal stem cells for the treatment of CNS diseases
US9796959B2 (en) 2012-12-28 2017-10-24 Kyoto University Method for inducing astrocytes
US10100279B2 (en) 2013-03-14 2018-10-16 The Regents Of The University Of California In vitro production of medial ganglionic eminence precursor cells

Families Citing this family (174)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7410798B2 (en) * 2001-01-10 2008-08-12 Geron Corporation Culture system for rapid expansion of human embryonic stem cells
DE60035191T2 (de) 1999-05-03 2008-06-19 Neuro Therapeutics Ab Materialien und methoden zur entwicklung von dopaminergen neuronen
US7011828B2 (en) * 2000-03-14 2006-03-14 Es Cell International Pte. Ltd. Implanting neural progenitor cells derived for human embryonic stem cells
IL151170A0 (en) 2000-03-14 2003-04-10 Es Cell Int Pte Ltd Embryonic stem cells and neural progenitor cells derived therefrom
US6921665B2 (en) * 2000-11-27 2005-07-26 Roslin Institute (Edinburgh) Selective antibody targeting of undifferentiated stem cells
US6576464B2 (en) * 2000-11-27 2003-06-10 Geron Corporation Methods for providing differentiated stem cells
JP3763749B2 (ja) * 2001-03-28 2006-04-05 独立行政法人科学技術振興機構 脊髄におけるシナプス形成ニューロンを誘導する中枢神経系前駆細胞
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
JP3886346B2 (ja) * 2001-06-22 2007-02-28 サンバイオ,インコーポレイティド 骨髄間質細胞由来Schwann細胞を含む神経再生用医薬組成物
DE10134667A1 (de) * 2001-07-20 2003-02-06 Neuroprogen Gmbh Leipzig Verfahren zur Herstellung isolierter Zellkulturen, Kulturmedium zur Kultivierung von Zellkulturen und Zellkultur
US6887706B2 (en) 2001-10-03 2005-05-03 Wisconsin Alumni Research Foundation Method of in vitro differentiation of transplantable neural precursor cells from primate embryonic stem cells
US7129034B2 (en) * 2001-10-25 2006-10-31 Cedars-Sinai Medical Center Differentiation of whole bone marrow
CN1630716A (zh) 2001-12-07 2005-06-22 杰龙公司 人胚胎干细胞衍生的造血细胞
US20050129664A1 (en) * 2002-01-09 2005-06-16 Hideyuki Okano Remedy for dysmnesia
WO2003060082A2 (en) 2002-01-14 2003-07-24 The Board Of Trustees Of The University Of Illinois Use of modified pyrimidine compounds to promote stem cell migration and proliferation
CA2473749C (en) * 2002-01-23 2012-05-22 University Of Utah Research Foundation Pure populations of astrocyte restricted precursor cells and methods for isolation and use thereof
AU2003229132B9 (en) * 2002-06-05 2009-07-23 Es Cell International Pte Ltd Generation of neural stem cells from undifferentiated human embryonic stem cells
EP2327762A1 (en) 2002-06-05 2011-06-01 ES Cell International Pte Ltd. Stem cells
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
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
AU2003269354B2 (en) * 2002-09-24 2008-03-06 Neuro Therapeutics Ab Methods for promoting dopaminergic neuronal development by using NG4A-subfamily and Wnt-ligands
AU2003276924A1 (en) * 2002-09-25 2004-04-19 Bresagen, Inc. Compositions and methods for enrichment of neural stem cells using ceramide analogs
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
WO2004053071A2 (en) * 2002-12-09 2004-06-24 Judith Kelleher-Andersson Method for discovering neurogenic agents
EP1572984B8 (en) * 2002-12-16 2016-06-01 Technion Research & Development Foundation Ltd. Feeder-free, xeno-free culture system for human embryonic stem cells
AU2003303741A1 (en) * 2002-12-18 2004-09-17 Bresagen, Inc. Compositions and methods for neural cell production and stabilization
ZA200401646B (en) * 2003-03-12 2004-06-07 Reliance Life Sciences Pvt Ltd Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells.
US7153650B2 (en) 2003-03-13 2006-12-26 Geron Corporation Marker system for preparing and characterizing high-quality human embryonic stem cells
US20060183221A1 (en) * 2003-03-31 2006-08-17 Schulz Thomas C Method for neural differentiation of embryonic stem cells using protease passaging techniques
WO2004099395A2 (en) * 2003-05-08 2004-11-18 Cellartis Ab A method for the generation of neural progenitor cells
AU2004320466A1 (en) * 2003-06-11 2006-02-02 Jan Remmereit Nuclear reprogramming of cells for therapeutic use
WO2004111210A1 (en) * 2003-06-12 2004-12-23 Yeda Research & Development Co.Ltd Enhancement of oligodendrocyte differentiation
US20070269412A1 (en) 2003-12-02 2007-11-22 Celavie Biosciences, Llc Pluripotent cells
US20070243610A1 (en) 2003-12-30 2007-10-18 Seoul National University Industry Foundation Embryonic Stem Cell Line and Method for Preparing the Same
JP5226928B2 (ja) * 2004-04-16 2013-07-03 株式会社カネカ 細胞の分化転換方法
US20050272149A1 (en) * 2004-06-02 2005-12-08 Life & Brain Gmbh Therapeutic delivery of adenosine into a tissue
US8785187B2 (en) 2004-06-09 2014-07-22 The University Court Of The University Of Edinburgh Neural stem cells
EP2267116B1 (en) 2004-07-13 2017-05-31 Asterias Biotherapeutics, Inc. Growth medium for primate embryonic stem cells
ES2626234T3 (es) * 2004-07-30 2017-07-24 Mayo Foundation For Medical Education And Research Tratamiento de tejido cardiovascular
WO2006020766A2 (en) 2004-08-12 2006-02-23 Neuro Probe Incorporated Point source diffusion cell activity assay apparatuses and methods
US20060040389A1 (en) * 2004-08-17 2006-02-23 Murry Charles E Purified compositions of stem cell derived differentiating cells
AU2005296063A1 (en) * 2004-10-05 2006-04-27 University Of Georgia Research Foundation, Inc. Neuronal progenitors from feeder-free human embryonic stem cell culture
US8192988B2 (en) 2004-10-22 2012-06-05 University Of Central Florida Research Foundation, Inc. Methods for increasing potency of adult mesenchymal stem cells
WO2006058427A1 (en) * 2004-12-01 2006-06-08 Stem Cell Therapeutics Corporation Platelet-derived growth factor-responsive neural precursor cells and progeny thereof
EP1844139A1 (en) * 2004-12-30 2007-10-17 Stemlifeline, Inc. Methods and systems relating to embryonic stem cell lines
WO2006072005A2 (en) * 2004-12-30 2006-07-06 The Johns Hopkins University Methods for high efficiency survival/proliferation of human embyonic stem cells and human embyro survival in culture
EP1841857A1 (en) * 2004-12-30 2007-10-10 Stemlifeline, Inc. Methods and compositions relating to embryonic stem cell lines
US8178349B2 (en) * 2005-02-09 2012-05-15 Burnham Institute For Medical Research Homogeneous neural precursor cells
PT1855700E (pt) 2005-03-07 2013-12-10 Sanbio Inc Utilização de materiais para tratamento de lesões do sistema nervoso central
EP1893749A1 (en) * 2005-06-22 2008-03-05 Geron Corporation Reporter hepatocytes and other cells for drug screening and toxicity testing
CN101233226B (zh) 2005-06-22 2017-08-11 阿斯特利亚斯生物治疗股份公司 人胚胎干细胞的悬浮培养物
US20060292695A1 (en) * 2005-06-22 2006-12-28 Roslin Institute Methods and kits for drug screening and toxicity testing using promoter-reporter cells derived from embryonic stem cells
US20060292694A1 (en) * 2005-06-22 2006-12-28 Roslin Institute Reporter hepatocytes and other cells for drug screening and toxicity testing
NZ565683A (en) * 2005-07-08 2011-04-29 Braincells Inc Methods for identifying agents and conditions that modulate neurogenesis in human neural cells
US20080226609A1 (en) * 2005-08-26 2008-09-18 University Of Rochester Transplantation of Glial Restricted Precursor-Derived Astrocytes for Promotion of Axon Growth
AU2006286149B2 (en) 2005-08-29 2012-09-13 Technion Research And Development Foundation Ltd. Media for culturing stem cells
EP1943334A1 (en) * 2005-09-02 2008-07-16 Agency for Science, Technology and Research Method of deriving progenitor cell line
JP5270362B2 (ja) 2005-11-10 2013-08-21 ジェナボン バイオファーマシューティカルズ エルエルシー 幹細胞のmntf分化および成長
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
EP4223769A3 (en) 2005-12-13 2023-11-01 Kyoto University Nuclear reprogramming factor
KR100683198B1 (ko) 2006-01-09 2007-02-15 주식회사 휴림바이오셀 신경전구세포를 도파민성 신경세포로 분화시키는 방법 및그에 사용되는 배지
KR100683199B1 (ko) 2006-01-09 2007-02-15 주식회사 휴림바이오셀 신경전구세포를 콜린성 신경세포로 분화시키는 방법 및그에 사용되는 배지
KR100838013B1 (ko) * 2006-06-07 2008-06-12 제일약품주식회사 인간배아줄기세포로부터 단계적 선별법을 통해 고수율로신경전구세포, 신경세포 및 기능성 도파민신경세포를생성하는 방법
US20080003676A1 (en) * 2006-06-26 2008-01-03 Millipore Corporation Growth of embryonic stem cells
US9765298B2 (en) * 2006-07-24 2017-09-19 Mayo Foundation For Medical Education And Research Methods and materials for providing cardiac cells
DK2733203T3 (en) 2006-08-02 2019-02-04 Technion Res & Dev Foundation PROCEDURES FOR EXPANSION OF EMBRYONAL STEM CELLS IN A SUSPENSION CULTURE
PT2008100168W (pt) * 2007-02-16 2010-06-28 Univ De Coimbra Método para a identificação funcional de novas células neuronais, células mãe neuronais, astrócitos e células imaturas de culturas de células estaminais e seus usos.
JP2008307007A (ja) * 2007-06-15 2008-12-25 Bayer Schering Pharma Ag 出生後のヒト組織由来未分化幹細胞から誘導したヒト多能性幹細胞
WO2008156708A2 (en) * 2007-06-15 2008-12-24 Massachusetts Institute Of Technology Methods and compositions for enhanced differentiation from embryonic stem cells
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
US8211698B2 (en) 2007-08-02 2012-07-03 California Stem Cells, Inc. Methods of derivation of neuronal progenitor cells from embryonic stem cells
KR100941036B1 (ko) 2007-10-05 2010-02-05 고려대학교 산학협력단 인간 배아줄기세포로부터 척수신경계 희소돌기 아교세포생산을 위한 삼단계 분화기법
WO2009066817A1 (en) * 2007-11-23 2009-05-28 Seoul National University Industry Foundation Method for inducing differentiation of human mesenchymal stem cells into motor neurons
US8513009B2 (en) 2008-01-30 2013-08-20 Geron Corporation Synthetic surfaces for culturing stem cell derived oligodendrocyte progenitor cells
WO2009133971A1 (en) 2008-05-02 2009-11-05 Kyoto University Method of nuclear reprogramming
WO2009145761A1 (en) * 2008-05-27 2009-12-03 Mayo Foundation For Medical Education And Research Methods and materials for using cells to treat heart tissue
CN102325878A (zh) 2008-12-23 2012-01-18 斯特姆塞尔思加利福尼亚有限公司 少突胶质前体细胞的靶向细胞群及制备与使用方法
US20100209399A1 (en) * 2009-02-13 2010-08-19 Celavie Biosciences, Llc Brain-derived stem cells for repair of musculoskeletal system in vertebrate subjects
US8642334B2 (en) 2009-02-17 2014-02-04 Memorial Sloan Kettering Cancer Center Methods of neural conversion of human embryonic stem cells
KR101812817B1 (ko) 2009-06-25 2017-12-27 제론 코포레이션 무관계 표현형이 고갈된 분화된 다능성 줄기 세포 자손
WO2011005326A1 (en) 2009-07-09 2011-01-13 Massachusetts Institute Of Technology Methods and compositions for increased safety of stem cell-derived populations
CA2770753C (en) * 2009-08-12 2019-01-15 Kyoto University Method for inducing differentiation of pluripotent stem cells into neural precursor cells
US11261425B2 (en) 2009-08-12 2022-03-01 Kyoto University Method for inducing differentiation of pluripotent stem cells into neural precursor cells
WO2011029053A1 (en) * 2009-09-04 2011-03-10 Buck Institute For Age Research Human embryonic stem cells for high throughout drug screening
AU2010315712B2 (en) 2009-10-19 2014-04-17 FUJIFILM Cellular Dynamics, Inc. Cardiomyocyte production
EP2499241B8 (en) 2009-11-10 2018-02-21 The J. David Gladstone Institutes Methods of generating neural stem cells
ES2779048T3 (es) 2009-11-12 2020-08-13 Technion Res & Dev Foundation Medios de cultivo, cultivos celulares y métodos de cultivo de células madre pluripotentes en un estado indiferenciado
IL301479A (en) 2009-11-17 2023-05-01 Astellas Inst For Regenerative Medicine Methods for preparing human RPE cells and pharmaceutical preparations of human RPE cells
AU2010326106B2 (en) * 2009-12-02 2015-08-20 Research Development Foundation Selection of stem cell clones with defined differentiation capabilities
CA2796251C (en) 2010-04-13 2019-05-14 Cellular Dynamics International, Inc. Hepatocyte production by forward programming
CA2796616A1 (en) 2010-04-21 2011-10-27 Research Development Foundation Methods and compositions related to dopaminergic neuronal cells
US20110312001A1 (en) 2010-06-15 2011-12-22 Emile Nuwaysir Compendium of ready-built stem cell models for interrogation of biological response
KR101861171B1 (ko) 2010-06-18 2018-05-31 후지필름 셀룰러 다이내믹스, 인코포레이티드 투석된 혈청이 있는 심근세포 배지
EP2591093B1 (en) 2010-07-07 2018-11-28 FUJIFILM Cellular Dynamics, Inc. Endothelial cell production by programming
US8808687B2 (en) 2010-07-12 2014-08-19 Mark Humayun Biocompatible substrate for facilitating interconnections between stem cells and target tissues and methods for implanting same
SG177238A1 (en) * 2010-07-22 2012-03-29 Affiliated Hospital Of Ningxia Medical University A method of producing neurons from stem cells, the neurons and uses thereof
EP2598635B1 (en) * 2010-07-30 2014-10-22 Cambridge Enterprise Limited Corticogenesis of human pluripotent cells
WO2012018933A2 (en) 2010-08-04 2012-02-09 Cellular Dynamics International, Inc. Reprogramming immortalized b cells
WO2012048276A2 (en) 2010-10-08 2012-04-12 Caridianbct, Inc. Customizable methods and systems of growing and harvesting cells in a hollow fiber bioreactor system
AU2011323197B2 (en) * 2010-11-05 2015-11-19 The Board Of Trustees Of The Leland Stanford Junior University Control and characterization of psychotic states
TWI586805B (zh) * 2011-01-12 2017-06-11 城戶常雄 獲得及維持在體外易於分化為寡樹突細胞-族系細胞之哺乳類神經幹細胞及/或神經祖源細胞的純或增富族群之培養方法
WO2012109208A2 (en) 2011-02-08 2012-08-16 Cellular Dynamics International, Inc. Hematopoietic precursor cell production by programming
AU2012236707B2 (en) 2011-03-29 2017-07-20 Asterias Biotherapeutics, Inc. Enriched populations of cardiomyocyte lineage cells from pluripotent stem cells
US9487752B2 (en) 2011-03-30 2016-11-08 Cellular Dynamics International, Inc. Priming of pluripotent stem cells for neural differentiation
WO2012149468A2 (en) 2011-04-29 2012-11-01 University Of Southern California Instruments and methods for the implantation of cell-seeded substrates
US8877489B2 (en) 2011-12-05 2014-11-04 California Institute Of Technology Ultrathin parylene-C semipermeable membranes for biomedical applications
CA2841165A1 (en) 2011-07-11 2013-01-17 Cellular Dynamics International, Inc. Methods for cell reprogramming and genome engineering
EP2737056B1 (en) 2011-07-29 2018-03-07 Cellular Dynamics International, Inc. Metabolic maturation in stem cell-derived tissue cells
CN102899285A (zh) * 2011-07-29 2013-01-30 复旦大学 一种体外诱导胚胎干细胞分化成为神经细胞方法
AU2012332269B2 (en) 2011-11-04 2018-05-31 Memorial Sloan-Kettering Cancer Center Midbrain dopamine (DA) neurons for engraftment
US9248013B2 (en) 2011-12-05 2016-02-02 California Institute Of Technology 3-Dimensional parylene scaffold cage
US9447378B2 (en) 2012-04-27 2016-09-20 Massachusetts Institute Of Technology Method for differentiating human embryonic stem cells into β-cells for the treatment of type I diabetes
JP6541577B2 (ja) * 2013-02-06 2019-07-10 ユニバーシティー オブ ロチェスター ミエリン障害の治療のための誘導多能性細胞由来オリゴデンドロサイト前駆細胞
CN105229144A (zh) 2013-02-22 2016-01-06 细胞动力学国际有限公司 通过组合的遗传工程和化学工程经由正向编程产生肝细胞
EP2981605B1 (en) 2013-04-03 2019-06-19 FUJIFILM Cellular Dynamics, Inc. Methods and compositions for culturing endoderm progenitor cells in suspension
WO2014176606A1 (en) 2013-04-26 2014-10-30 Memorial Sloan-Kettering Center Center Cortical interneurons and other neuronal cells produced by the directed differentiation of pluripotent and multipotent cells
EP3008169B1 (en) 2013-06-10 2021-11-10 Academisch Ziekenhuis Leiden Differentiation and expansion of endothelial cells from pluripotent stem cells and the in vitro formation of vasculature like structures
KR102205234B1 (ko) 2013-06-11 2021-01-20 프레지던트 앤드 펠로우즈 오브 하바드 칼리지 SC-β 세포 및 조성물 그리고 그 생성 방법
US9816070B2 (en) 2013-06-14 2017-11-14 Massachusetts Institute Of Technology Articles and methods for stem cell differentiation
ES2721440T3 (es) * 2013-09-05 2019-07-31 Univ Kyoto Nuevo método para inducir células precursoras neurales productoras de dopamina
US10336985B2 (en) * 2013-10-14 2019-07-02 Hadasit Medical Research Services & Development Limited Method of obtaining terminally differentiated neuronal lineages and uses thereof
CN105793411B (zh) 2013-11-16 2018-04-17 泰尔茂比司特公司 生物反应器中的细胞扩增
CA2931334A1 (en) 2013-11-21 2015-05-28 Memorial Sloan-Kettering Cancer Center Specification of functional cranial placode derivatives from human pluripotent stem cells
EP3119881B1 (en) 2014-03-21 2023-03-01 FUJIFILM Cellular Dynamics, Inc. Production of midbrain dopaminergic neurons and methods for the use thereof
JP6783143B2 (ja) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド 培地の受動的補充
US20170107486A1 (en) 2014-04-21 2017-04-20 Cellular Dynamics International, Inc. Hepatocyte production via forward programming by combined genetic and chemical engineering
US20170044500A1 (en) 2014-04-24 2017-02-16 Board Of Regents, The University Of Texas System Application of induced pluripotent stem cells to generate adoptive cell therapy products
CA2956787A1 (en) * 2014-07-31 2016-02-04 Universite D'aix-Marseille Method for generating neuronal and muscular cells
US20160090569A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled Feed
WO2016061071A1 (en) 2014-10-14 2016-04-21 Cellular Dynamics International, Inc. Generation of keratinocytes from pluripotent stem cells and mantenance of keratinocyte cultures
CN105624116B (zh) * 2014-11-07 2020-04-07 中国科学院上海生命科学研究院 利用多能干细胞制备神经前体细胞的方法
CN107614678B (zh) 2014-12-18 2021-04-30 哈佛学院校长同事会 干细胞来源的β细胞的产生方法及其使用方法
WO2016100909A1 (en) 2014-12-18 2016-06-23 President And Fellows Of Harvard College METHODS FOR GENERATING STEM CELL-DERIVED β CELLS AND USES THEREOF
WO2016100898A1 (en) 2014-12-18 2016-06-23 President And Fellows Of Harvard College Serum-free in vitro directed differentiation protocol for generating stem cell-derived b cells and uses thereof
US9724432B2 (en) 2015-04-30 2017-08-08 University Of Rochester Non-human mammal model of human degenerative disorder, uses thereof, and method of treating human degenerative disorder
HRP20220482T1 (hr) * 2015-06-01 2022-07-08 Memorial Sloan Kettering Cancer Center Postupci in vitro diferencijacije neurona dopamina srednjeg mozga (mda)
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US10384207B2 (en) 2015-07-21 2019-08-20 Neuro Probe Incorporated Assay apparatus and methods
ES2970537T3 (es) 2015-09-08 2024-05-29 The Us Secretary Department Of Health And Human Services Método para la diferenciación reproducible de células epiteliales del pigmento retiniano de calidad clínica
CA2997763A1 (en) 2015-09-08 2017-03-16 Cellular Dynamics International, Inc. Macs-based purification of stem cell-derived retinal pigment epithelium
WO2017062971A1 (en) 2015-10-08 2017-04-13 Neurona Therapeutics Inc. Neural precursor cell populations and uses thereof
BR112018008648A2 (pt) 2015-10-30 2018-11-27 Univ California métodos de geração de células t a partir de células tronco e métodos imunoterapêuticos ao usar células t
WO2017173064A1 (en) 2016-03-30 2017-10-05 Asterias Biotherapeutics, Inc. Oligodendrocyte progenitor cell compositions
CN109415696A (zh) 2016-05-25 2019-03-01 泰尔茂比司特公司 细胞扩增
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
ES2901379T3 (es) 2016-08-16 2022-03-22 Fujifilm Cellular Dynamics Inc Métodos para diferenciar células pluripotentes
CA3057104A1 (en) * 2017-03-21 2018-09-27 Memorial Sloan-Kettering Cancer Center Stem cell-derived astrocytes, methods of making and methods of use
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP3621434A4 (en) 2017-05-10 2021-03-31 University of Rochester METHOD OF TREATMENT OF NEUROPSYCHIATRIC DISORDERS
MX2020004939A (es) 2017-11-15 2020-11-11 Semma Therapeutics Inc Fabricacion de composiciones de celulas islote y metodos de uso de las mismas.
US20210238546A1 (en) 2018-04-20 2021-08-05 FUJIFILM Cellular Dynamics, Inc. Method for differentiation of ocular cells and use thereof
RU2690498C1 (ru) * 2018-05-17 2019-06-04 Федеральное государственное бюджетное научное учреждение "Медико-генетический научный центр" Способ получения ноотропной композиции на основе полипептидных комплексов, выделенных из нейронов и глиальных клеток, полученных методом направленной дифференцировки индуцированных плюрипотентных стволовых клеток человека
RU2690846C1 (ru) * 2018-05-17 2019-06-06 Федеральное государственное бюджетное научное учреждение "Медико-генетический научный центр" Ноотропная композиция на основе полипептидных комплексов, выделенных из нейронов и глиальных клеток, полученных методом направленной дифференцировки индуцированных плюрипотентных стволовых клеток человека
CA3108275A1 (en) 2018-08-10 2020-02-13 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
US20220016318A1 (en) 2018-11-19 2022-01-20 The United State Of America, As Represented By The Secretary, Deparment Of Health And Human Services Biodegradable tissue replacement implant and its use
KR20200077222A (ko) 2018-12-20 2020-06-30 (주)영인바이오텍 통증완화부를 가지는 회복 운동 장치
KR20200077218A (ko) 2018-12-20 2020-06-30 (주)영인바이오텍 교체가능한 부품을 가지는 회복 운동 장치
US20220119765A1 (en) * 2019-01-22 2022-04-21 Korea University Research And Business Foundation Differentiation method of neural stem cells manufactured by direct cell conversion into astrocytes
EP3945827A4 (en) * 2019-04-03 2022-12-14 The Johns Hopkins University METHODS, COMPOSITIONS AND KITS FOR MAKING SKELETAL MUSCLE STEM CELLS AND TREATMENT OF DISORDERS
ES2960527T3 (es) 2019-09-13 2024-03-05 Miltenyi Biotec Bv & Co Kg Método para la generación de una composición celular de células progenitoras dopaminérgicas del mesencéfalo ventral
AU2020381537A1 (en) 2019-11-14 2022-06-09 Amgen Inc. Hematopoietic precursor cell production
EP4065091A4 (en) * 2019-11-25 2023-11-29 The Penn State Research Foundation COMPOSITION AND METHOD FOR CONVERTING HUMAN GLIA CELLS INTO NEURONS
RU2732599C1 (ru) * 2019-12-26 2020-09-21 Федеральное государственное бюджетное научное учреждение "Медико-генетический научный центр имени академика Н.П. Бочкова" Ноотропная композиция на основе полипептидных комплексов, выделенных из глиальных прогениторных клеток в условиях теплового шока, и способ её получения
RU2732600C1 (ru) * 2019-12-26 2020-09-21 Федеральное государственное бюджетное научное учреждение "Медико-генетический научный центр имени академика Н.П. Бочкова" Ноотропная композиция на основе полипептидных комплексов, выделенных из нейрональных прогениторных клеток в условиях гипоксии, и способ ее получения
RU2752906C2 (ru) * 2019-12-26 2021-08-11 Федеральное государственное бюджетное научное учреждение "Медико-генетический научный центр имени академика Н.П. Бочкова" Ноотропная композиция на основе полипептидных комплексов, выделенных из нейрональных прогениторных клеток в условиях теплового шока, и способ ее получения
CA3180561A1 (en) 2020-05-29 2021-12-02 Andrew DIAS Bilayer of retinal pigmented epithelium and photoreceptors and use thereof
US20230201267A1 (en) 2020-05-29 2023-06-29 FUJIFILM Cellular Dynamics, Inc. Retinal pigmented epithelium and photoreceptor dual cell aggregates and methods of use thereof
KR20230165846A (ko) 2021-04-07 2023-12-05 후지필름 셀룰러 다이내믹스, 인코포레이티드 도파민성 전구세포 및 사용 방법
AU2022280062A1 (en) 2021-05-28 2023-11-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Methods to generate macular, central and peripheral retinal pigment epithelial cells
EP4346928A1 (en) 2021-05-28 2024-04-10 The United States of America, as represented by The Secretary, Department of Health and Human Services Biodegradable tissue scaffold with secondary matrix to host weakly adherent cells
CN116478923B (zh) * 2022-04-26 2024-01-02 浙江霍德生物工程有限公司 一种星形胶质细胞的制备方法

Family Cites Families (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2111845C (en) 1991-06-18 2001-08-28 Arnold I. Caplan Monoclonal antibodies specific for marrow-derived mesenchymal cells
EP0605428B9 (en) 1991-06-24 2003-01-02 hCell Technology, Inc. Hormone-secreting pancreatic cells maintained in long-term culture
US5851832A (en) 1991-07-08 1998-12-22 Neurospheres, Ltd. In vitro growth and proliferation of multipotent neural stem cells and their progeny
US5981165A (en) 1991-07-08 1999-11-09 Neurospheres Holdings Ltd. In vitro induction of dopaminergic cells
WO1993001275A1 (en) 1991-07-08 1993-01-21 Neurospheres Ltd. NOVEL GROWTH FACTOR-RESPONSIVE PROGENITOR CELLS WHICH CAN BE PROLIFERATED $i(IN VITRO)
EP0597964A4 (en) 1991-08-07 1994-11-30 Einstein Coll Med PROLIFERATION OF HEPATOCYTE PRECURSORS.
US5672499A (en) 1992-07-27 1997-09-30 California Institute Of Technology Immoralized neural crest stem cells and methods of making
US5849553A (en) 1992-07-27 1998-12-15 California Institute Of Technology Mammalian multipotent neural stem cells
WO1994003199A1 (en) 1992-08-04 1994-02-17 Regeneron Pharmaceuticals, Inc. Method of enhancing differentiation and survival of neuronal precursor cells
US5766948A (en) 1993-01-06 1998-06-16 The Regents Of The University Of California Method for production of neuroblasts
US5534404A (en) 1993-12-10 1996-07-09 Cytotherapeutics, Inc. Glucose responsive insulin secreting β-cell lines and method for producing same
US5639618A (en) 1994-05-13 1997-06-17 Plurion, Inc. Method of isolating a lineage specific stem cell in vitro
US5476975A (en) 1994-07-08 1995-12-19 Ruddick; John N. R. Extraction of toxic organic contaminants from wood and photodegradation of toxic organic contaminants
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
GB9807935D0 (en) * 1998-04-14 1998-06-10 Univ Edinburgh Lineage specific cells and progenitor cells
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
US6090622A (en) 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
US6361996B1 (en) 1997-05-07 2002-03-26 University Of Utah Research Foundation Neuroepithelial stem cells and glial-restricted intermediate precursors
US6235527B1 (en) * 1997-11-29 2001-05-22 University Of Utah Research Foundation Lineage restricted glial precursors from the central nervous system
WO1999001159A1 (en) * 1997-07-04 1999-01-14 University Of Utah Research Foundation Lineage-restricted neuronal precursors
US7144997B2 (en) 1997-07-24 2006-12-05 Curis, Inc. Vertebrate embryonic patterning-inducing proteins, compositions and uses related therto
US5968829A (en) 1997-09-05 1999-10-19 Cytotherapeutics, Inc. Human CNS neural stem cells
CA2307807C (en) 1997-10-23 2008-09-02 Andrea G. Bodnar Methods and materials for the growth of primate-derived primordial stem cells in feeder-free culture
DE19756864C5 (de) * 1997-12-19 2014-07-10 Oliver Brüstle Neurale Vorläuferzellen, Verfahren zu ihrer Herstellung und ihre Verwendung zur Therapie von neuralen Defekten
EP1056835A4 (en) 1998-02-27 2003-01-15 Hampton Roads Medical College DERIVATION OF CELLS AND TISSUES FROM THE PRE-STRAINED CELL STAGE FOR TRANSPLANTATION THERAPIES
CA2324591A1 (en) 1998-04-09 1999-10-21 Bresagen Limited Cell differentiation/proliferation and maintenance factor and uses thereof
US5958767A (en) 1998-08-14 1999-09-28 The Children's Medical Center Corp. Engraftable human neural stem cells
DE19843234A1 (de) * 1998-09-09 2000-03-23 Inst Pflanzengenetik & Kultur Differenzierte Herzzellen mit pathologischen Merkmalen als in vitro-Modell für Herzerkrankungen
AU755849B2 (en) 1998-09-22 2002-12-19 Neuralstem Biopharmaceuticals, Ltd. Stable neural stem cell lines
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
US6087168A (en) 1999-01-20 2000-07-11 Cedars Sinai Medical Center Conversion of non-neuronal cells into neurons: transdifferentiation of epidermal cells
AU780256B2 (en) 1999-02-12 2005-03-10 Boco Silicon Valley, Inc. Enriched central nervous system stem cell and progenitor cell populations, and methods for identifying, isolating and enriching for such populations
US6238922B1 (en) 1999-02-26 2001-05-29 Stemcells, Inc. Use of collagenase in the preparation of neural stem cell cultures
EP1165771A2 (en) 1999-03-17 2002-01-02 The University Of Tennessee Research Corporation Rt-pcr of clonal neurospheres
EP1196426A4 (en) 1999-06-30 2003-09-03 Advanced Cell Tech Inc CYTOPLASMIC TRANSFER TO REDUCE DIFFERENTIATION OF RECEIVED CELLS
AU2913801A (en) 1999-12-23 2001-07-03 Cornell Research Foundation Inc. A method for isolating and purifying multipotential neural progenitor cells and multipotential neural progenitor cells
US20050042749A1 (en) * 2001-05-16 2005-02-24 Carpenter Melissa K. Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
US6602711B1 (en) 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells
IL151170A0 (en) * 2000-03-14 2003-04-10 Es Cell Int Pte Ltd Embryonic stem cells and neural progenitor cells derived therefrom
US6458589B1 (en) 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
WO2001083715A2 (en) 2000-05-01 2001-11-08 THE GOVERNMENT OF THE UNITED STATES OF AMERICA as represented by the Secretary, Derivation of midbrain dopaminergic neurons from embryonic stem cells
US8273570B2 (en) 2000-05-16 2012-09-25 Riken Process of inducing differentiation of embryonic cell to cell expressing neural surface marker using OP9 or PA6 cells
EP1287116A2 (en) 2000-05-17 2003-03-05 Geron Corporation 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
WO2001098463A1 (en) 2000-06-20 2001-12-27 Es Cell International Pte Ltd Method of controlling differentiation of embryonic stem (es) cells by culturing es cells in the presence of bmp-2 pathway antagonists
JP3660601B2 (ja) 2001-03-30 2005-06-15 独立行政法人科学技術振興機構 胚性幹細胞からの神経幹細胞、運動ニューロン及びgaba作動性ニューロンの製造法
WO2002086106A1 (en) 2001-04-23 2002-10-31 Nsgene A/S Method and culture medium for producing neural cells expressing tyrosine hydroxylase
US20030113910A1 (en) 2001-12-18 2003-06-19 Mike Levanduski Pluripotent stem cells derived without the use of embryos or fetal tissue
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
WO2006058427A1 (en) * 2004-12-01 2006-06-08 Stem Cell Therapeutics Corporation Platelet-derived growth factor-responsive neural precursor cells and progeny thereof

Cited By (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030087263A1 (en) * 1999-12-16 2003-05-08 Renu Wadhwa Method of screening remedy for cancer with the use of interaction domain of p53 and mortalin
US10351821B2 (en) 2000-01-11 2019-07-16 Asterias Biotherapeutics Inc. Neural cell populations from primate pluripotent stem cells
US9790466B2 (en) 2000-01-11 2017-10-17 Asterias Biotherapeutics, Inc. Neural cell populations from primate pluripotent stem cells
US20090291495A1 (en) * 2000-01-11 2009-11-26 Geron Corporation Neural Cell Populations from Primate Pluripotent Stem Cells
US7560281B2 (en) 2000-01-11 2009-07-14 Geron Corporation Use of TGF beta superfamily antagonists to make dopaminergic neurons from embryonic stem cells
US20090305405A1 (en) * 2000-01-11 2009-12-10 Geron Corporation Use of tgf beta superfamily antagonists and neurotrophins to make neurons from embryonic stem cells
US8252586B2 (en) 2000-01-11 2012-08-28 Geron Corporation Neural cell populations from primate pluripotent stem cells
US8153428B2 (en) 2000-01-11 2012-04-10 Geron Corporation Use of TGF beta superfamily antagonists and neurotrophins to make neurons from embryonic stem cells
US8148148B2 (en) 2000-05-17 2012-04-03 Geron Corporation Neural progenitor cell populations
US20090117639A1 (en) * 2000-05-17 2009-05-07 Carpenter Melissa K 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
US9803174B2 (en) 2000-05-17 2017-10-31 Asterias Biotherapeutics, Inc. Neural progenitor cell populations
US20050095707A1 (en) * 2000-05-17 2005-05-05 Carpenter Malissa K. Use of cyclic AMP and ascorbic acid to produce dopaminergic neurons from embryonic stem cells
US20060078545A1 (en) * 2000-05-17 2006-04-13 Carpenter Melissa K Neural progenitor cell populations
US20030103949A1 (en) * 2000-05-17 2003-06-05 Carpenter Melissa K. Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
US7763463B2 (en) 2000-05-17 2010-07-27 Geron Corporation Use of cyclic AMP and ascorbic acid to produce dopaminergic neurons from embryonic stem cells
US8252585B2 (en) 2000-05-17 2012-08-28 Geron Corporation Neural progenitor cell populations
US20050042749A1 (en) * 2001-05-16 2005-02-24 Carpenter Melissa K. Dopaminergic neurons and proliferation-competent precursor cells for treating Parkinson's disease
WO2003000868A1 (en) * 2001-06-21 2003-01-03 Geron Corporation Dopaminergic neurons and proliferation-competent precursor cells for treating parkinson's disease
GB2393733B (en) * 2001-06-21 2005-09-14 Geron Corp Dopaminergic neurons and proliferation-competent precursor cells for treating parkinson's disease
JP2010158242A (ja) * 2001-06-21 2010-07-22 Geron Corp パーキンソン病を治療するためのドーパミン作動性ニューロンおよび増殖能のある前駆細胞
GB2393733A (en) * 2001-06-21 2004-04-07 Geron Corp Dopaminergic neurons and proliferation-competent precursor cells for treating parkinson's disease
US20050095706A1 (en) * 2001-10-03 2005-05-05 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US9080151B2 (en) 2001-10-03 2015-07-14 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
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
US7972850B2 (en) 2001-10-03 2011-07-05 Wisonsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20080206865A1 (en) * 2001-10-03 2008-08-28 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20080227137A1 (en) * 2001-10-03 2008-09-18 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
WO2003089580A3 (en) * 2002-04-16 2004-06-03 Dana Farber Cancer Inst Inc Cancer models
US20030226159A1 (en) * 2002-04-16 2003-12-04 Bachoo Robert M. Cancer models
WO2003089580A2 (en) * 2002-04-16 2003-10-30 Dana-Farber Cancer Institute, Inc. Cancer models
US20070185024A1 (en) * 2002-07-16 2007-08-09 Thomas Jessell Systems and methods for screening for modulators of neural differentiation
US20040224887A1 (en) * 2002-07-16 2004-11-11 Thomas Jessel Systems and methods for screening for modulators of neural differentiation
US7632679B2 (en) 2002-07-16 2009-12-15 The Trustees Of Columbia University In The City Of New York Systems and methods for screening for modulators of neural differentiation
US20070224650A1 (en) * 2002-07-16 2007-09-27 Thomas Jessell Systems and methods for screening for modulators of neural differentiation
US20050265983A1 (en) * 2002-11-17 2005-12-01 Eldad Melamed Methods, nucleic acid constructs and cells for treating neurodegenerative disorders
US20040253719A1 (en) * 2003-03-07 2004-12-16 Goldman Steven A. Identification and Isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain
US8642332B2 (en) * 2003-03-07 2014-02-04 Cornell Research Foundation, Inc. Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain
US7674620B2 (en) 2003-03-12 2010-03-09 Reliance Life Sciences Pvt. Ltd. Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells
US20060211109A1 (en) * 2003-03-12 2006-09-21 Reliance Life Sciences Pvt. Ltd. Derivation of terminally differentiated dopaminergic neurons from human embryonic stem cells
GB2421029B (en) * 2003-08-29 2008-04-09 Wisconsin Alumni Res Found Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
AU2004269361B2 (en) * 2003-08-29 2010-05-20 Wisconsin Alumni Research Foundation Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
WO2005021720A3 (en) * 2003-08-29 2005-05-19 Wisconsin Alumni Res Found Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
GB2421029A (en) * 2003-08-29 2006-06-14 Wisconsin Alumni Res Found Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20050095702A1 (en) * 2003-09-03 2005-05-05 Reliance Life Sciences Pvt. Ltd. In vitro generation of GABAergic neurons from embryonic stem cells and their use in the treatment of neurological disorders
US7820439B2 (en) 2003-09-03 2010-10-26 Reliance Life Sciences Pvt Ltd. In vitro generation of GABAergic neurons from pluripotent stem 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
US7829332B2 (en) 2004-02-13 2010-11-09 Cornell Research Foundation, Inc. Purines are self-renewal signals for neural stem cells, and purine receptor antagonists promote neuronal and glial differentiation therefrom
US20050181503A1 (en) * 2004-02-13 2005-08-18 Goldman Steven A. Purines are self-renewal signals for neural stem cells, and purine receptor antagonists promote neuronal and glial differentiation therefrom
EP1773983A4 (en) * 2004-02-26 2009-07-22 Univ Columbia SYSTEMS AND METHOD FOR STUDYING MODULATORS OF NEURAL DIFFERENTIATION
EP1773983A2 (en) * 2004-02-26 2007-04-18 The Trustees of Columbia University in the City of New York Systems and methods for screening for modulators of neural differentiation
US20080096273A1 (en) * 2004-09-06 2008-04-24 Yasushi Kondo Method for Preparing Conditioned Medium of Astrocyte-Like Cells
US10869899B2 (en) 2005-06-16 2020-12-22 Ramot At Tel-Aviv University Ltd. Isolated cells and populations comprising same for the treatment of CNS diseases
US8647874B2 (en) * 2005-06-16 2014-02-11 Ramot At Tel-Aviv University Ltd. Isolated cells and populations comprising same for the treatment of CNS diseases
US9879225B2 (en) 2005-06-16 2018-01-30 Ramot At Tel-Aviv University Ltd. Isolated cells and populations comprising same for the treatment of CNS diseases
US20090010895A1 (en) * 2005-06-16 2009-01-08 Ramot At Tel Aviv University Ltd. Isolated cells and populations comprising same for the treatment of CNS diseases
US20100021434A1 (en) * 2005-12-08 2010-01-28 Ramot At Tel Aviv University Ltd. Isolated Oligodendrocyte-Like Cells and Populations Comprising Same for the Treatment of CNS Diseases
US20090175835A1 (en) * 2006-02-07 2009-07-09 Korea Institute Of Radiological & Medical Sciences Composition for treating damage of central or peripheral nerve system
US9765297B2 (en) 2007-04-13 2017-09-19 The Trustees Of Columbia University In The City Of New York Stem cell-based culture system for drug development
US20100267073A1 (en) * 2007-04-13 2010-10-21 Serge Przedborski Stem cell-based culture system for drug development
US9474787B2 (en) 2008-05-28 2016-10-25 Ramot At Tel-Aviv University Ltd. Mesenchymal stem cells for the treatment of CNS diseases
US11185572B2 (en) 2008-05-28 2021-11-30 Ramot At Tel-Aviv University Ltd. Mesenchymal stem cells for the treatment of CNS diseases
US8900574B2 (en) 2008-05-28 2014-12-02 Ramot At Tel-Aviv University Ltd. Mesenchymal stem cells for the treatment of CNS diseases
US8663987B2 (en) 2008-05-28 2014-03-04 Ramot At Tel-Aviv University Ltd. Mesenchymal stem cells for the treatment of CNS diseases
US10052363B2 (en) 2008-05-28 2018-08-21 Ramot At Tel-Aviv University Ltd. Mesenchymal stem cells for the treatment of CNS diseases
US20100196332A1 (en) * 2008-12-10 2010-08-05 Hynek Wichterle Generation of brachial, thoracic and lumbar spinal motor neurons from embryonic stem cells in the absence of all-trans retinoic acid supplement
US8969081B2 (en) 2008-12-10 2015-03-03 The Trustees Of Columbia University In The City Of New York Caudal motor neuron derived from embryonic stem cells under conditions essentially free of any retinoid
US20140051085A1 (en) * 2011-03-01 2014-02-20 The Scripps Research Institute Direct reprogramming of human fibroblasts to functional neurons under defined conditions
US9796959B2 (en) 2012-12-28 2017-10-24 Kyoto University Method for inducing astrocytes
US10760048B2 (en) 2013-03-14 2020-09-01 The Regents Of The University Of California In vitro production of medial ganglionic eminence precursor cells
US10100279B2 (en) 2013-03-14 2018-10-16 The Regents Of The University Of California In vitro production of medial ganglionic eminence precursor cells
US11884932B2 (en) 2013-03-14 2024-01-30 The Regents Of The University Of California In vitro production of medial ganglionic eminence precursor cells

Also Published As

Publication number Publication date
US20060078545A1 (en) 2006-04-13
JP6644277B2 (ja) 2020-02-12
JP2013063080A (ja) 2013-04-11
CN100580079C (zh) 2010-01-13
US9803174B2 (en) 2017-10-31
CA2409698C (en) 2010-10-26
US8252585B2 (en) 2012-08-28
US20090117639A1 (en) 2009-05-07
WO2001088104A3 (en) 2002-06-20
IL152741A (en) 2011-01-31
AU6319901A (en) 2001-11-26
EP1287116A2 (en) 2003-03-05
HK1055765A1 (en) 2004-01-21
WO2001088104A2 (en) 2001-11-22
JP2018029585A (ja) 2018-03-01
US8148148B2 (en) 2012-04-03
US20140242691A1 (en) 2014-08-28
GB2379447B (en) 2004-12-29
KR100903755B1 (ko) 2009-06-18
GB2379447A (en) 2003-03-12
JP5685242B2 (ja) 2015-03-18
JP2014110790A (ja) 2014-06-19
KR20030032953A (ko) 2003-04-26
JP5943533B2 (ja) 2016-07-06
JP2016146839A (ja) 2016-08-18
CN1429267A (zh) 2003-07-09
US6833269B2 (en) 2004-12-21
US20120308534A1 (en) 2012-12-06
IL152741A0 (en) 2003-06-24
US20020039724A1 (en) 2002-04-04
AU2001263199B2 (en) 2004-09-16
JP2003533224A (ja) 2003-11-11
CA2409698A1 (en) 2001-11-22
GB0229369D0 (en) 2003-01-22

Similar Documents

Publication Publication Date Title
US9803174B2 (en) Neural progenitor cell populations
US7250294B2 (en) Screening small molecule drugs using neural cells differentiated from human embryonic stem cells
AU2001263199A1 (en) Neural progenitor cell populations
AU2002322270A1 (en) Dopaminergic neurons and proliferation-competent precursor cells for treating parkinson's disease
JP2010158242A (ja) パーキンソン病を治療するためのドーパミン作動性ニューロンおよび増殖能のある前駆細胞
US7763463B2 (en) Use of cyclic AMP and ascorbic acid to produce dopaminergic neurons from embryonic stem cells
AU2004214542B2 (en) Neural progenitor cell populations

Legal Events

Date Code Title Description
AS Assignment

Owner name: GERON CORPORATION, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CARPENTER, MELISSA K.;REEL/FRAME:011822/0567

Effective date: 20010516

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: ASTERIAS BIOTHERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GERON CORPORATION;REEL/FRAME:031578/0248

Effective date: 20130926