CA2417356A1 - Directed differentiation of embryonic cells - Google Patents

Directed differentiation of embryonic cells Download PDF

Info

Publication number
CA2417356A1
CA2417356A1 CA002417356A CA2417356A CA2417356A1 CA 2417356 A1 CA2417356 A1 CA 2417356A1 CA 002417356 A CA002417356 A CA 002417356A CA 2417356 A CA2417356 A CA 2417356A CA 2417356 A1 CA2417356 A1 CA 2417356A1
Authority
CA
Canada
Prior art keywords
cells
differentiation
condition
cell
exogenous
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
CA002417356A
Other languages
French (fr)
Inventor
Nissim Benvenisty
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.)
Yissum Research Development Co of Hebrew University of Jerusalem
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
Application filed by Individual filed Critical Individual
Publication of CA2417356A1 publication Critical patent/CA2417356A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/502Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects
    • G01N33/5023Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing non-proliferative effects on expression patterns
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • 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
    • C12N5/0619Neurons
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5073Stem 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
    • C12N2501/10Growth 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
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • 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
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • 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
    • 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
    • C12Q2600/136Screening for pharmacological compounds
    • 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
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells

Abstract

Methods are described for mapping a pathway of differentiation of a populati on of embryonic cells which includes exposing the cells to an exogenous factor and measuring gene expression products that are characteristic of a particul ar cell type or lineage. Directing differentiation of human embryonic cells relies on dissociated embryoid bodies which are then exposed to one or more exogenous factors to enrich a culture for a particular cell type. The differentiated cells may be used for treating a medical condition in a human . Kits for determining differentiation pathways and screening exogenous factor s for their utility in differentiation are provided.

Description

Directed Differentiation of Embryonic Cells Technical Field and Background Art The present invention relates to methods and kits for directed differentiation of embryonic stem cells and their use in vivo and ih vitro.
Embryonic stem (ES) cells have been described in the prior art as undifferentiated pluripotent cells derived from the inner cell mass of blastocyst stage embryos which can grow indefinitely in culture while retaining a normal karyotype. Blastocyst derived stem cells form teratomas when injected into muscles or testis of severe combined immunodeficiency mice.
Murine embryonic stem cells have been reported to form aggregates or embryoid bodies in vitro which are capable of spontaneously differentiating into various cell types. In contrast to blastocyst derived stem cells, embryoid body derived cells do not appear to form teratomas when implanted in vivo. (Robertson, (1987) IRL Press, Oxford, pp. 71-112; Thomson et al. (1998) Science Vol 282,pp. 1145-1147; Reubinoff et al. (2000) Nat.Biotechnol. Vol 18, pp. 399-404).
While human embryonic stem cells have been recovered from human embryos produced by irz vitro fertilization, the formation of embryoid bodies from non-human primates and from humans has been problematic. In contrast to mice, the formation of embryoid bodies from primates, more particularly, the common marmoset (Thomson, (1996) Biology of Reproduction, Vol. 55, pp. 254-259, USP 5,843,780) and the rhesus monkey (Thomson, (1995) Proc.Natl.Acad.Sci, USA, Vol. 92, pp. 7844-7848) were found to be inconsistent and asynchronous. Moreover, differentiation of rhesus embryonic stem cells was disorganized and vesicular structures did not form. In addition, human embryonic stem cells have been shown to differentiate in a spontaneous and uncontrolled manner, so that the experimenter cannot determine which cell types will form in vivo or in vitro (Thomson, et al., (1998) Science, Vol.
282, pp. 1145-1147; Reubinoff, et aL, (2000) Nat. Biotechnol, Vol. 19, pp. 399-404; Itskovitz Eldor, et al., (2000) Mol. Med., Vol. 6, pp. 88-95).
Development of techniques for manipulating differentiation of human embryonic stem cells to provide a uniform population of precursors and differentiated cells of a desired lineage are desirable for irz vivo medical uses and for in vitro assays.
Although many useful lessons rnay be learned about development in mammals from studying mice, not all of development is the same in all animals.
Consequently, it is desirable to have tools to analyze and compare pathways in different mammals and to combine these tools with a methodology that permits the isolation, preservation and cultivation of embryonic stem cells from mammals including humans for correcting a disease condition resulting from cell degeneration.
In particular, human embryonic stem cells may be uniquely useful as a source of cells for transplantation in numerous human pathologies, and as a component in biomedical engineering as well as providing clues on early stages of human development.
Summary of the Invention In a first embodiment of the invention there is provided a method of mapping a pathway to of differentiation of a population of embryonic cells, that includes: (i) selecting: (a) a set of gene expression products, wherein each gene expression product in the set is characteristic of a cell type that has undergone differentiation, such that a plurality of differentiated cell types are represented in the set; and (b) an exogenous factor from a library of exogenous factors; (ii) applying the exogenous factor to a population of cells; (iii) characterizing the effect of the 15 exogenous factor on the differentiation pathway of the undifferentiated cells by determining gene expression products in the set; and (iv) mapping the pathway of differentiation.
According to the above, the gene expression products may include at least one gene expression product that is expressed in the mesoderm, ectoderm and/or endoderm, where the expression product expressed in the ectoderm may be for example, neurofilament heavy chain 20 (NF-H), keratin or adrenal dopamine (3 hydroxylase (D J3H), the expression product expressed in the mesoderm may include any of enolase, renin, cartilage matrix protein (CMP), kalikrein, Wilms Tumor 1 (WTI), cardiac actin (cACT), b-globulin or ~i-globulin, the gene expression product expressed in the endoderm may include albumin, a 1-antitrypsin (a 1 AT), amylase, pancreatic and duodenal homeobox gene 1 (PDX-1), insulin and a-fetoprotein (aFP).
25 According to the above, examples of exogenous factor include interleukins, basic fibroblast growth factor (bFGF), transforming growth factor (TGF(31), activin-A, bone morphogenic protein-4 (BMP-4), hepatocyte growth factor (HGF), epidermal growth factor (EGF), (3 nerve growth factor (NGF) and retinoic acid (RA).
In another embodiment of the invention, a method is provided for directing differentiation 3o of human embryonic cells to a specific cell type, including the steps of (a) permitting a population of embryonic stem cells to form embryoid bodies; (b) dissociating the embryoid bodies to provide embryonic cells for differentiating in the presence of at least one exogenous factor for an effective period of time; and (c) causing directed differentiation of human embryonic cells to form the specific cell type.
According to the above method, the embryoid bodies may be formed in a suspension culture. The embryonic cells may form a monolayer on a substrate. The exogenous factor may be a growth factor, for example an interleukin, nerve growth factor, or retinoic acid. The differentiated cells may be a neuronal cell type and further may have neuronal processes. In all the present embodiments, the embryonic cells may be human.
In another embodiment of the invention, a method is provided of treating a subject to suffering from a condition associated with degeneration of cells or malfunction of cells, that includes: (a) accessing dissociated embryoid cells; (b) treating the cells with an exogenous factor;
(c) causing the cells to differentiate; and (d) placing an effective amount of differentiated cells into the subject to treat the condition.
According to the above, the condition may be a heart condition in which heart muscle is is degenerated. The cells may be treated with an exogenous factor selected from the group consisting of TGF-(3, FGF, R.A, HGF and EGF and an effective amount of differentiated cells targeted to the heart. The condition may be a kidney condition in which kidney tissue is degenerated, the cells being treated with NGF and an effective amount of differentiated cells targeted to the kidney. The condition may be a shin condition in which skin tissue is .
20 degenerated, the cells treated with BMP-4 and an effective amount of differentiated cells targeted to skin. The condition may be a liver condition in which the liver is degenerated, the cells treated with NGF and an effective amount of differentiated cells targeted to the liver. The condition may be a brain condition in which brain tissue is degenerated, the cells treated with at least one of NGF or RA and an effective amount of differentiated cells targeted to the brain. The condition 25 may be a spinal cord injury in which the neurons are degenerated, the cells treated with at least one of NGF or RA and an effective amount of differentiated cells are targeted to the liver. The condition may be anemia or immunodeficiency and the cells treated with at least one of NGF or interleukin and the differentiated cells targeted to the blood. The condition may be an adrenal condition in which the adrenal tissue is degenerated, the cells treated with RA and an .effective 3o amount of differentiated cells targeted to the adrenal tissue.
According to the above, the subject may be a mammal including a human subject.
The differentiated cell type may be any of brain cells, liver cells, pancreatic cells, muscle cells, chondrocytes, kidney cells, Mullerian duct cells, heart cells, blood cells, skin cells and adrenal cells.
In another embodiment of the invention, a kit for determining differentiation pathways, is provided that includes: a plurality of sets of cell specific markers forming a panel in an assay format, the assay format including reagents for detecting the cell specific markers, the cell specific markers including a first set of markers that are characteristic of each of the ectoderm, mesoderm and endoderm of the embryo, and a second set of markers that are characteristic of a body tissue, the second set containing more than one marker, and means for detecting reagents bound to cell specific markers.
In another embodiment of the invention, a method for screening an exogenous factor is provided to determine whether the factor is capable of causing directed differentiation in a population of human embryonic cells, including (a) subjecting the population of human embryonic cells to the exogenous growth factor; (b) measuring the expression of receptors, the receptors being of a type that characterizes a particular differentiated cell population; and (c) determining whether the exogenous factor enhances differentiation, maintains differentiation at a normal level or depresses differentiation of the particular cell population.
In another embodiment of the invention, a panel of cell type differentiation determining markers is provided that includes a set of reagents for specifically detecting gene expression of a plurality of ectodermal specific proteins, a plurality of mesodermal specific proteins and a plurality of endodermal specific proteins.
According to the above, the ectodermal specific proteins may include neurofilament protein, keratin and adrenal dopamine (3 hydroxylase, the mesodermal specific proteins may include enolase, CMP, renin, kallikrein, WTI, cACT, (3 globin, 8 globin and cActin and the endodermal specific proteins may include amylase, a-FP, PDX-1 and insulin. The reagents may include DNA primers or antibodies.
Brief Description of the Drawings The foregoing features of the invention will be more readily understood by reference to the following detailed description, taken with reference to the accompanying drawings, in which:
Fig. 1 shows induced differentiation of human embryonic stem cells in culture.
(A) A
schematic representation of the differentiation protocol. (B) Various morphologies of embryonic stem cells before and after induced differentiation: I: A colony of embryonic stem cells on feeders. II-IV: Differentiated cells cultured in the presence of HGF, Activin-A, RA, bFGF, or BMP-4, respectively also showing the small size of embryonic cells in the colony in comparison to the size of the differentiated cells. Scale bar, 100 ~,m.
Figure 2 shows expression of receptors for various growth factors in human embryonic stem cells. RNA samples from ES cells, 5 day old embryoid bodies (EBs) and differentiated embryonic (DE) cells were analyzed by RT-PCR for expression of specific receptors. As a control, RNA of 5 day old EBs was analyzed without the prior generation of cDNA (-RT).
Figure 3 shows (A) analysis of expression of cell specific genes in human ES
cells treated with various growth factors. RNA from ES, 20 day old EBs, and DE cells treated with the different growth factors was analyzed by RT-PCR for expression of seventeen cell specific genes and two house-keeping genes. The genes were categorized by their embryonic germ layer. (B) Schematic representation of the growth factor effects on gene expression.
Results are shown from three experiments on the effects of NGF, HGF, EGF, RA, bFGF, BMP4, TGF(31 or Activin-A on the expression of 13 genes. The results are color-coded: orange represents similar expression to the control (no growth factor), red represents induction of at least two fold, yellow represents inhibition of two fold or more, and green represent no detectable expression.
Figure 4 shows the effects of various growth factors on in vitro differentiation of human embryonic cells. Thick arrows and large fonts indicate an induction compared to the control (no growth factor) for genes specific to the tissue indicated. Dashed arrows represent a decrease in .
expression. Disappearance of the cell specific transcripts is symbolized by elimination of the 2o corresponding tissue symbol.
Figure 5(a) and (b) show examples of genes encoding gene expression products for use in panels to measure directed differentiation of cells. Figure 5 (c) and 5(d) provide the corresponding SEQ 1D number for each sequence.
Figure 6 shows that retinoic acid and [3NGF induce expression of neurofilament light chain (NF-L) mRNA. 1Z situ hybridization analysis of cells derived from dissociated EBs demonstrating expression of NF-L. Shown are RA- (low (10-7M) or high (10-6M) concentrations), TGF(31, and ~NGF-treated cultures. Scale Bar=100p,M.
Histograms show the percentage of cells expressing the NF-L RNA following each treatment. Assay of each condition was repeated three times (between 50 to 150 cells) and standard error bars are shown in the graph.
Figure 7 shows that retinoic acid enhances formation of neuronal processes from human EBs. (A) neuronal cell bodies (double arrows) and a complex proximal process (arrow) are seen in a section through an EB stained with anti-NF-H antibody.
Hoecsht staining (blue) shows both intact and fragmented nuclei. (B) A plexus of neuronal-like processes formed from intact EBs plated on collagen. (C) Immunostaining for NF-H of 20 day old EBs plated on collagen shows that plexuses of neuronal processes with varying degrees of complexity develop. (D) A histogram summarizing the effect of RA on the proportion of EBs displaying different degrees of network complexity. Each condition was assayed by counting all EBs in culture (between 20 to 40) and the experiment was repeated twice resulting in similar results. Scale Bar=100p,M.
Figure 8 shows that retinoic acid induces expression of neuron-specific genes.
RT-PCR analysis of expression of dopamine receptor D1 (DRD2), serotonin receptor 2A and 1o 5A (5HT'2A and 5HT5A), dopa decarboxylase (DDC) and the house-keeping gene glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in human embryonic stem (ES) cells, and in 21 day old EBs in the presence of RA (EB+RA). RNA samples that were not reverse transcribed were used as a negative control to ensure the absence of DNA
contamination (-RT).
Detailed Description of Specific Embodiments Definitions. As used in this description and the accompanying claims, the following terms shall have the meanings indicated, unless the context otherwise requires:
The term "differentiation" is defined here and in the claims as a change that occurs in cells to cause those cells to assume certain specialized functions and to lose the ability to change 2o into certain other specialized functional units. Cells capable of differentiation may be any of totipotent, pluripotent or multipotent cells. Differentiation may be partial or complete with respect to mature adult cells.
The term "embryonic stem cells" as used here and in the claims refers to a pluripotent cell type derived from the inner cell mass of blastocyst stage embryos. The term "embryonic cells" as used here and in the claims includes embryonic stem cells, embryoid bodies (EBs) and differentiated embryonic cells (DE) which are generally pluripotent but may include partially differentiated cells that are multipotent.
The term "exogenous factor" is defined here and in the claims as any factor which when presented in the medium of a population of cells, has an effect on the biological properties of the cell. The term exogenous factor includes a secreted factor or a synthetic agent or a receptor on the surface of a neighboring cell or an inhibitor of any of these. These factors may be proteins, carbohydrates or lipids or mixtures of the same and further may include peptides, non-peptide hormones, small organic molecules and novel therapeutic agents or candidate therapeutic molecules that may be a product of a chemical library or a naturally occurring source. The exogenous factor may be a single type of molecule or a mixture of more than one molecule. The exogenous factor may be purified before being added into the cell medium or it may be a crude preparation. Examples of exogenous factors include secreted biological molecules such as growth factors, hormones, antibodies, as well as salts or ions.
The term "subject" is defined here and in the claims as any living organism, more particularly a mammal, more particularly a human.
to The term "gene expression product" is defined here and in the claims as any of a transcribed RNA or a protein including a post translationally modified protein.
The term "a" or "an" or "the" is intended to mean "one or more"
Embodiments of the invention provide a method for screening exogenous factors to determine which are capable of causing directed differentiation of embryonic stem cells into progenitors of mature cells or mature cells themselves. The screening method provides intelligence on how to enrich a population of cells for a particular cell type using a single exogenous factor or combinations of exogenous factors. The method has shown that some exogenous factors have an inhibitory activity with respect to a particular pathway while other factors actively stimulate differentiation down a particular pathway. This latter category may 2o include inhibitors of factors with inhibitory activity. Other factors have no effect on differentiation. The method is exemplified in Figures 1 through 4. Further embodiments of the invention utilize gene expression of cell-specific markers as an indicator of cell type that results from differentiation of the embryonic cells in the presence of exogenous factors.
Other embodiments of the invention include selecting exogenous factors that have been found to direct differentiation to produce significantly homogenous cell populations for ifz vitro or ifa vivo use. For example, we have found that compared to control differentiated embryonic cells, cultures that are treated with growth factors are more homogeneous and up to half of the culture contained one or two cell types. As exemplified in Fig. 3, panels of gene expression products have been constructed for determining directed differentiation of embryonic cells in the presence of exogenous factors. Using this approach, we can characterize a growing number of partially differentiated and differentiated cell types and can provide a comprehensive profile of exogenous factors suited for inducing a predetermined selection of differentiated cells. A directed differentiation pathway for obtaining neurons from embryonic cells is described in Example 3.

The above described methods are applicable to mammalian embryonic stem cells including human embryonic cells. Prior art references have reported heterogeneous population of murine cells which are enriched for a specific cell either by addition of growth factors or by introduction of transcription factors. (Gordon et al. USP, 5,914,268; Wiles, et al., (1991) Development, Vol. 111, pp. 259-267; Biesecker, et al., (1993) Exp. Hem.atol., Vol. 21, pp. 774-778; Stager, et al., (1993) Dev. Genet., Vol. 14, pp. 212-224; Bain et al., (1995) Dev. Biol., Vol.
168, pp. 342-357; Brustle, et al., (1999) Science, Vol. 285, pp. 754-756;
Gutierrez-Ramos, et al., (1992) Proc. Natl. Acad. Sci. USA, Vol. 89, pp. 9171-9175; Levinson-Dushnik, et al., (1997) Mol. Cell. Biol., Vol. 17, pp. 3817-3822). Subsequent sorting and selection of cell types from the to heterogeneous population has been undertaken to further enrich for selected cells. (Klug, et al., (1996) J. Clizz. Invest., Vol. 98, pp. 216-224; Kolossov, et al., (1998) J.
Cell. Biol., Vol. 143, pp.
2045-2056; Li, et al., (1998) Curr. Biol. 8, Vol. 143, pp. 2045-2056; Lee et al., (2000) Nature Biotechyzology, Vol. 18, pp. 675-678). Mouse ES cells that are allowed to aggregate to form EBs can differentiate into mature neurons capable of innervating adult mouse brain and spinal chord (Bain et al., (1995) Dev. Biol., Vol. 168, pp. 342-357; Wobus et al., (1988) Biomed. Biochinz.
Acta., Vol. 47, pp. 965-973; Deacon et al., (1988) Exp. Neurol., Vol. 149, pp.
28-41; Lee et al., (2000) Nat. Biotechnol., Vol. 18, pp. 675-679 and McDonald et al., (1999) Nat.
Med., Vol. 5, pp.
1410-1412). None of these studies sought to undertake a broad analysis of the effects of a range of exogenous factors on differentiation into multiple cell types.
2o In a preferred embodiment, we have provided a novel approach for obtaining human differentiated cells from embryonic cells and have established methods to enrich mixtures of multiple cell types (for example, human cell types) ifs vitro using specific exogenous factors.
Utilizing the property of embryonic stem cells for indefinite growth in culture, we describe a means to manipulate differentiation of embryonic stem cells, in particular human embryonic cells, to provide a source of cells for transplantation into a subject. We have found that treatment of dissociated embryoid bodies with exogenous factors can give rise to populations of cells with discrete morphologies such as small cells with pronounced, muscle like syncitiums, neuronal like cells, fibroblast like cells, large round cells or other mesenchymal or epithelial cells. These differing morphologies that suggest that specific programs are initiated as a result of treatment with the factors.
The protocol described in Example 1 for inducing human embryonic stem cell differentiation consisted of an aggregation step which allows complex signaling to occur between the cells (somewhat resembling the gastrulation process) and a dissociation step, after 5 days as EBs, thereby allowing exposure to exogenous growth factors. At the stage of 5 day old EBs, the cells are not terminally differentiated as is evident from their expression of Oct-4, a marker for undifferentiated ES cells and express a wide range of receptors for growth factors.
In the examples, we describe the use of eight growth factors (bFGF, TGF(31, Activin A, BMP-4, HGF, EGF, (3NGF and retinoic acid) for directed differentiation of human embryonic stem cells in vitro. Further examples of exogenous factors include interleukins, hedgehog proteins, vascular endothelial growth factor (VEGF), granulocyte macrophage colony stimulating factox (GMCSF), macrophage colony stimulating factor (MCSF), wnt and int and other members of the FGF family not previously mentioned. Where differentiated human cells express a cell l0 specific receptor for an exogenous factor, the effects of the factor on embryonic stem cells can be monitored by measuring the increased amounts of the cell specific receptors expressed as a result of activation of a differentiation pathway. Differentiation may further be correlated with various epithelial or mesenchymal morphologies. The development of panels for analyzing embryonic cells at various stages of differentiation as described here provide a useful tool for directing differentiation in a routine manner.
Accordingly, in Examples 1 and 2, differentiation of the cells was assayed by expression of 24 cell specific molecular markers that cover all embryonic germ layers and 11 different tissues. The described growth factors are not intended to be limiting. The lineage specific diffexentiation induced by the various growth factors is summarized in Figure 4. While each growth factor has a unique effect, we can divide the factors into three categories (Figure 4) based on their effects on human embryonic stem cell differentiation. The first group (TGFj31 and Activin-A) appear to inhibit differentiation of cells into endodermal and ectodermal cells, but allow differentiation into selected mesodermal cells. Among mesodermal cells, these factors inhibit differentiation into chondrocytes in the doxsal mesoderm, kidney and Mullerian duct cells in the intermediate mesoderm and blood formation in the lateral mesoderm.
The second group includes factors that allow or induce differentiation into ectoderm as well as mesodermal cells (RA, bFGF, BMP-4 and EGF). Nonetheless, these factors vary according to Which cells are inhibited, produced at normal levels or enhanced.
For example, as shown in Figure 4, RA inhibits the differentiation of chondrocyte cells, cells produced by the intermediate mesoderm, and blood formation in human embryonic stem cells. In contrast, BMP-4 causes chondrocytes and blood formation differentiation to be reduced and inhibits differentiation of all other mesodermal cells examined while apparently permitting skin cells to form. EGF appears to stimulate differentiation of heart, and skin, permits normal levels of muscle but inhibits the formation of cells forming kidney, blood and brain.
FGF stimulates only formation of cells for the heart, permits formation of normal levels of muscle skin and brain and inhibits differentiation of cells for the kidney.
The third group of growth factors (NGF and HGF) allows differentiation into all three embryonic lineages including endoderm. For example, NGF has been shown to direct the differentiation of embryonic cells toward forming liver tissue (hepatocytes), pancreas (islet cells), brain (neurons), kidney and mullerian duct cells. To a lesser extent, NGF
appears to stimulate production of heart and blood and to down-regulate differentiation of muscle and bone (chondrocytes). However, differentiation into skin cells is completely inhibited in the presence to of NGF. This pattern is not the same for HGF. When HGF is added to human embryonic cells, skin cells are induced at their normal level. However, differentiation of kidney cells is completely inhibited in the presence of HGF, with significant inhibition of formation of liver, pancreas, brain, chondrocytes and blood.
The results provided in Figure 4 illustrate a powerful new approach to directed differentiation. Any exogenous factor may be analyzed using this approach whereby embryonic stem cells and/or differentiated embryonic cells are exposed to the factor and specific informative gene expression products are analyzed in the differentiated embryonic cells for each of the ectoderm, mesoderm and endodermal tissues and their differentiated counterparts. An advantage of the novel approach to evaluating differentiation of tissues described above is that the complex 2o relationship of a single exogenous factor with respect to differentiation can be established and then manipulated.
For example, specific differentiation in differentiated embryonic (DE) cell cultures can be detected first as a morphological change. After 10 days incubation with an exogenous factor, DE
cells were more homogenous and displayed a larger proportion of specific cell types as compared to controls not treated with an exogenous factor. We have analyzed the differentiation of these cells by measuring gene expression using a selected set of markers for different cell types. We have taken advantage of substantial public records of cell mapped proteins.
The number of protein markers that are characteristic for mature diseased and healthy cells of different types is very large and provide a convenient starting point for analyzing cells resulting from directed differentiation. Although gene expression products that characterize mature cells are well established, this is not the case for intermediate cell types that occur during differentiation of an undifferentiated embryonic cell to a mature cell. The markers described in Figure 5 provide examples of gene expression products from which a set of markers may be selected for to identifying the effect of an exogenous factor on an embryonic cell population.
This list is not intended to be comprehensive. Other markers such as T-cell receptor, oc-globin, somatostatin, phosphoenolpyruvate carboxykinase (PEPCK), lactate dehydrogenase (LDH) and G-protein coupled receptors such as glucagon, not previously mentioned can also be used.
The set of markers for intermediate cell lineages may be selected from a group that increases in size in an iterative process. For example, a particular exogenous factor is found to stimulate the differentiation of embryonic cells into a particular type of immature cell recognized by particular markers according to embodiments of the invention. These cells may then be analyzed for protein content by using standard protocols in the art such as 2D
protein blots to to identify however gene expression varies with the fully mature cells. Those gene expression products that are found in intermediate or mature cells or both may serve as markers for measuring the effect of subsequent exogenous factors on embryonic cells. This approach permits the isolation and characterization of novel precursor cells not previously recognized by prior art methodology.
15 We have exemplified the above approach using a panel of 24 tissue specific markers from all three germ layers and sub-layers. Eight different growth factors were analyzed on embryonic stem cells in culture using these 24 tissue specific markers. These cells were human although the embodiments of the method may be applied to embryonic stem cells from any source. We utilized the panel of markers in an assay format wherein in one embodiment, the assay format 20 utilized standard quantitative gel based techniques that relied on RT-PCR
and selected primers (see Example l.-2 and Figures 1-3). Other assay formats known in the art may be used to map pathways of differentiation including sensitive antibody based techniques.
Sensitive antibody techniques include radioimmune assays, enzyme linked immune absorption (ELISA) and fluorescent antibody techniques. Other defections means include ifz situ hybridization methods, 25 and solid chemistry analysis in which probes or primers may be attached to a solid substrate in arrays. DNA arrays permit large numbers of samples to be rapidly analyzed.
Accordingly, the effects of the particular growth factors on the differentiation status of multiple tissues in differentiated embryonic cells can be determined.
In addition to identifying the expression of genes associated with particular tissues, 3o differentiation can be followed by examining the changing morphology of differentiating cells.
For example, the ability to induce specific differentiation was initially evident as morphological changes in the differentiated embryonic cell cultures that had been cultured for predetermined times in the presence of exogenous factors (Figure 1-A). After incubation with an exogenous factor, differentiated embryonic cells were more homogenous and displayed a larger proportion of specific cell types as compared to controls not treated with factors. The differentiation of these cells was also followed by gene expression studies in which we analyzed the transcription of tissue specific markers from all three germ layers and sub-layers. As shown in Example 2, 16 of the 24 genes examined were expressed in the human DE cells representing 11 different tissues (Fig. 3).
The analysis of a large set of markers permits us to specify factors that can direct differentiation into specific cell types. Figure 4 shows that none of the eight growth factors tested in the examples directed a completely uniform and singular differentiation of human l0 embryonic stem cells although the number of differentiated cell types varied substantially for different factors. In the above example, differentiation observed from individual growth factors indicates that most of the growth factors tested inhibit differentiation of specific cell types and that this inhibitory effect is more pronounced than an induction effect.
Accordingly, specific differentiation may also be achieved by using growth factor inhibitors, such as follistatin or noggin, to counteract endogenous growth factors that might be produced during human embryonic stem cell differentiation.
In summary, certain exogenous factors may primarily inhibit differentiation of cells along certain pathways resulting in differentiation of cells in secondary pathways.
Specific tissues may result from withholding or blocking the action of such exogenous factors.
Other exogenous factors may directly stimulate differentiation of embryonic stem cells while still other factors may have little or no effect on the growth and development of embryonic stem cells at certain times during development. Multiple factors may be used in a certain order to obtain an optimum differentiation outcome and combined with selection methods provide a means for achieving fully directed cell differentiation.
Embodiments of the invention provide methods for obtaining cells that are differentiated partly or entirely along a particular cell lineage. These methods have both in vivo and in vitro uses. An in vivo use for cells that are a product of an in vitro differentiation pathway could for example be directed to repair of bone damage in vivo using chondrocytes derived from stem cells treated according to methods described herein. Moreover, in vitro differentiated cells may be used to repair damaged organs for example, neuronal cells for spinal cord repair.
We have shown that neurons can be formed from human embryonic cells in the presence of certain growth factors. These neuronal cultures mature to form neuronal processes with varying degrees of complexity. In addition to demonstrating that both RA and (3NGF enhance the number of neuronal cells that develop from human embryonic cells as described above, we demonstrate that RA promotes the production of mature neurons and that these neurons express dopamine or serotonin receptors, and form complex plexuses of neuronal processes. Only RA
but not NGF induce expression of adrenal markers whereas NGF and not RA induce expression of endodermal markers.
The enrichment of neurons in culture by growth factors may occur through a process of cell selection (either by expansion or improved survival) or as a consequence of induced differentiation. Our production of neuronally-differentiated embryonic cells provides a method for obtaining large amounts of neurons in culture for use for example, in the replacement of to neurons lost to degeneration or trauma in the central nervous system in humans. This example serves to underscore the utility of our approach in generating an unlimited source of cells for transplantation therapy and other uses. This can reduce the need for difficult to obtain fetal tissues, which to date are the primary sources of human cells available for transplantation therapy (Peschanski, (1995) Neuroscience, Vol. 68, pp. 273-285).
For cell therapy, the cells may be added directly to the subject to reverse an adverse medical condition. For example, differentiated embryonic cells that are neuronal in type may be administered directly to a patient for treating Parkinson's disease, multiple sclerosis or spinal cord injury. Differentiated embryonic cells may be used directly to treat Lou Gehrig's disease, diabetis or other autoimmune condition. Indeed, differentiated embryonic cells may be used 2o directly to treat disease or damage to any organ or tissue in the human body. Alternatively, the cells may be built into an artificial tissue in vitro by seeding a biocompatible three-dimensional or two-dimensional inorganic or organic matrix known in the art with cells and permitting the cells to grow and differentiate within the matrix before transplantation. An example of a biocompatible matrix seeded with adult murine thymic stroma is provided in Poznansky et al., (2000) Nature Biotechnology, Vol. 18, pp. 729-734.
Additional uses for methods for providing cells that are differentiated partly or entirely along a particular cell lineage include in vitro uses such as creating reagents for drug toxicity assays.
By adding drugs to cultures of differentiated cells such as kidney cells, liver cells, brain cells, heart smooth muscle, chondrocytes, pancreatic cells, neuronal cells, blood cells etc, it is possible 3o to avoid the use of animals in preclinical testing. Furthermore, the addition of drugs to partially differentiated cells may provide useful information on the impact of the drugs on the developing embryo. Isolation and characterization of cells that are intermediate in differentiation provides a useful resource for understanding the genotype and phenotype of cells in development in the context of health and disease.
Examples Example 1. Measuring gene expression products in differentiated human embryonic stem cells Cell Culture: Human embryonic stem cells were grown on a feeder layer of mouse embryonic fibroblasts (MEF) and then transferred to gelatin coated plates and cultured further to reduce the number of murine cells in the culture. Differentiation into embryoid bodies (EBs) was initiated by transfer to petridishes, where the embryoid bodies remained in suspension. After 5 days, the EBs become dissociated and cells are cultured as a monolayer forming differentiated embryonic (DE) cells. This protocol allows for initial differentiation of the human embryonic stem cells as aggregates and further differentiation as a monolayer wherein cells can be exposed to exogenous growth factors. The DE cells were cultured for 10 days in the presence of 8 different growth factors: NGF, bFGF, Activin-A, TGF(31, HGF, EGF, BMP4 and retinoic acid (RA) (Figure 1-A).
Human ES cells were obtained as described in Thomson et al., (1998) Science, Vol. 286, pp. 1145-1147. Cleavage stage human embryos produced by in vitro fertilization (IVF) were obtained after the requisite approval process. The embryos were cultured to the blastocyst stage and inner cell masses were isolated. ES cell lines were isolated from the embryos and a cell line was selected which had maintained a normal karyotype over a 6 months period of culture and could be passaged continuously over several months without undergoing a period of replicative crisis. The cells had a uniform undifferentiated morphology when grown on mouse embryo fibroblasts in 80% KnockOutTM DMEM - an optimized Dulbeco's modified Eagle's medium for ES cells (Gibco-BRL), 20% KnockOutTM SR - a serum-free formulation (Gibco-BRL), 1 mM
glutamine (Gibco-BRL), 0.1 mM (3-mercaptoethanol (Sigma), 1% non-essential amino acids stocle (Gibco-BRL), 103 units/ml leukemia inhibitor factor (LIF) (Gibco-BRL) and 4 ng/ml basic fibroblast growth factor (bFGF) (Gibco-BRL). Gelatin (0.1%, Merck) was used to cover the tissue culture plates.
To induce formation of EBs, ES cells were transferred using trypsin/EDTA
(0.1%/1mM) to plastic Petri dishes to allow their aggregation and prevent adherence to the plate. Human EBs were grown in the same culture medium, except that it lacked leukemia inhibitory factor (LIF) and bFGF. The EBs were cultured for 5 days and then dissociated and plated on tissue culture plates coated with 50p,g/ml Fibronectin (Boehringer Mannheim). The cells were grown in the presence of the following human recombinant growth factors used singly or in groups: Basic Fibroblast Growth Factor (bFGF) ( Swoboda, et.al., (1999) Neurology, Vol. 53, pp. 1205-1211.) - lOng/ml (Gibco BRL). Transforming Growth Factor Beta 1 (TGF(31) (Swoboda et al.) - 2ng/ml (R&D Systems). Activin-A (Stager et al., (1993) Dev. Gehet., Vol. 14, pp. 212-224)- 20ng/ml (R&D Systems). Bone Morphogenic Protein 4 (BMP-4) Stager et al., and Bain et al., (1996) Biochef~2. Bioplays. Res.
Commufa., Vol. 223, pp.
691-694 14)- lOng/ml (R&D Systems). Hepatocyte Growth Factor (HGF)/ Scatter Factor (Doetsch et al., (1999) Cell Vol. 97, pp. 703-716) - 20ng/ml (R&D Systems).
Epidermal to Growth Factor (EGF) (Doetsch et al.) - 100ng/ml (R&D Systems). (3Nerve Growth Factor (NGF) (Johansson et al., (1999) Cell, Vol., 96, pp. 25-34.) -100ng/ml (R&D
Systems).
Retinoic acid (RA) (Deacon, et al., (1998) Exp. Neurol. Vol. 149, pp. 28-41;
Peschanski, et al., (1995) Neuroscience, Vol. 68, pp. 273-285) - 1~.M (Sigma). Under these conditions the differentiated embryonic (DE) cells were grown for another 10 days. All of the examined growth factors are absent from the KnockOutTM serum replacement in which the embryonic cells were cultured.
In this example, initial differentiation of the human ES cells aggregates occurred which continued after the cells formed monolayers. In a preferred embodiment, the monolayers were exposed to exogenous factors. Figure 1-A depicts one protocol that utilizes growth factors.
As described in more detail in Example 2, the presence of receptors which correlate with the addition of exogenous factors could be determined in DE cells at the stage when the factors are added to the culture (5 day old EBs). For example, RNA from human embryonic stem cells, 5 day old EBs and 10 day old DE cells were isolated and analyzed by RT-PCR
using primers specific to retinoic acid receptor type alpha (RAR), nerve growth factor receptor (NGFR), fibroblast growth factor receptor type I (FGFRI), activin receptor type IIB
(ACTRIIB), transforming growth factor receptor type II (TGFRII), hepatocyte growth factor receptor (c-Met), epidermal growth factor receptor (EGFR), and bone morphogenic protein 4 receptor type II
(BMP4RII). As a control, the expression of octamer binding protein 4 (Oct-4), a marker of the undifferentiated cells, and of G3PD (glyceraldehyde 3-phosphate dehydrogenase), a housekeeping gene was also monitored.
Ira-situ hybridization and immunohistochemistry: EBs or their plated derivatives were fixed in 4% paraformaldehyde. In i~a-situ experiments, hybridization to a 50-mer 2'-O-methyl 5'-biotinylated cDNA of NF-L (SEQ ID NO: 1) (CCTGCGTGCGGATGGACTTGAGGTCGTTGCTGATGGCGGCTACCTGGCTC) followed. The probes were then detected with streptavidin-conjugated alkaline phosphatase using a fluorogenic substrate (Grifman, et al., (1998) Proc. Natl. Acad. Sci.
U S A, Vol. 95, pp. 13935-13940). For immunohistochemistry, EBs were either embedded in paraffin and sectioned at l Op,M, or stained directly on the plates. Mouse monoclonal anti-NF-H was used as a primary antibody, and was detected with Cy'3-conjugated goat anti-mouse IgG.
RT-PCR Analysis: Total RNA was extracted using Atlases Pure Total RNA Kit (Clontech). cDNA was synthesized from 1 ~g total RNA, using AdvantageTM RT-for-PCR Kit (Clontech). cDNA samples were subjected to PCR amplification with DNA primers selective for the human genes. For each gene, the DNA primers were derived from different exons to ensure that the PCR product represents the specific mRNA species and not genomic DNA.
PCR was performed using the Clontech Advantaq+TM RT-PCR kit and using a 2 step cycle at 68°C.
Primers were synthesized for the following genes: Neurofilament heavy Chain (NF-H), Phosphoprotein enriched in astrocytes (PEA-15), Keratin, Dopamine [3 Hydroxylase (D(3H), Follicular Stimulating Hormone (FSH), Enolase, Myosin light polypeptide2 (myosin), Cartilage Matrix Protein (CMP), Renin, Kallikrein, Wilms Tumor 1 (WT1), (3-Globin, ((3-Glob), ~-Globin (8-Glob), Cardiac Actin (cACT), Albumin, a1 anti-trypsin (cclAT), Lipase, Amylase, PDX-l, Insulin, Glucagon, Surfactant, Parathyroid Hormone (PTH), a-feto protein (ocFP), Retinoic Acid Receptor type alpha (RAR), Hepatocyte Growth Factor Receptor (c-Met), Nerve Growth Factor 2o Receptor (NGFR), Fibroblast Growth Factor Receptor type I (FGFRI), Transforming Growth Factor Receptor type II (TGFRII), Epidermal Growth Factor Receptor type (EGFR), Activin Receptor type lIB (ACTRIIB), Bone Morphogenic Protein 4 Receptor type IT
(BMP4RII), Octamer Binding Protein 4 (Oct 4), Glyceraldehyde 3-phosphate dehydrogenase (G3PD), (3-Actin. A full description of the primers and their expected products are provided in Figure 5.
Also incorporated by reference, see http:llwww.ls.huii.ac.ilhnissimb/factors eslprimers.html-ente Example 2: Mapping_pathwa~ of differentiation by measuring the effect of exogenous growth factors on undifferentiated cells All 8 target receptors were expressed in 5 day old EBs and most were expressed in ES
3o and DE cells (Figure 2). Notably, four of the receptors (TGFRII, c-Met, EGFR and BMP4RII) were expressed at very low levels in human ES cells and their expression increases following culture and further differentiation. Without wishing to be limited by theory, we propose that low levels of expression may result from partial differentiation in embryonic cell cultures.
Since growth factor receptors were expressed in 5 day old EBs, we could examine the effects of the corresponding ligands on differentiation. After several days of culture on plastic and continuous exposure to growth factors, the cells acquired different morphologies (Figure 1-B). Without growth factors, cells spontaneously differentiated into many different types of colonies, whereas the addition of growth factors produced more mature cell morphologies such as syncitial myocytes and neuronal cells. Compared to control DE cells, the growth factor treated cultures were more homogenous and up to half of the culture contained one or two cell types. For example, large populations of small cells with pronounced nuclei were found in to cultures treated with HGF (Figure 1-B-I), muscle like syncitiums in the Activin-A treated cells (Figure 1-B-III), neuronal like cells in the RA treated culture (Figure 1-B-IV), fibroblast like cells in the cultures treated with bFGF (Figure 1-B-V), and large round cells in cultures treated with BMP-4 (Figure 1-B-VI). These varied cell morphologies demonstrate that specific programs are initiated as a result of growth factor treatment.
The differentiation induced by growth factors was further examined by determining the expression of a panel of 24. cell specific genes using RT-PCR. RNA was isolated from ES cells, day old EBs, 10 day old DE cells, and DE cells that had been treated with either RA, NGF, bFGF, Activin-A, TGF(31, HGF, EGF, and BMP4. Complementary DNA was generated from the various samples and transcripts were amplified by specific primers to these genes. For each 2o gene, the two DNA primers were derived from different exons to ensure that the PCR product represented the specific mRNA species and not genomic DNA. In addition, the correct product of amplification from the primers was ascertained by amplifying cDNAs from a variety of human adult or embryonic tissues (Clontech, MTC~ Panels) (results not shown). Out of the 24 cell specific markers, 16 were expressed in the treated DE cells. Most transcripts were also expressed in the control DE cells (see Fig 3-A), indicating that many different cell types develop from ES
cells without a requirement of exogenous growth factors. Moreover, human embryonic cell cultures expressed some cell specific genes, indicating that there is partial spontaneous differentiation in the cultures.
Several conclusions can be drawn comparing the expression pattern of cell specific 3o markers in the different treatments to that of the control DE cells. Some markers are expressed only in a presence of a specific growth factor, e.g. the adrenal marker dopamine (3 hydroxylase (D(3H) is expressed only when the cells are treated with retinoic acid and the uro-genital marker WT-1 is expressed when either NGF or HGF are added (Figure 3-A). In contrast, muscle specific enolase is expressed under all conditions except following BMP-4 exposure (Figure 3-A). Some growth factors, e.g. NGF, induce expression of a variety of transcripts indicative of cells from all three germ layers whereas other factors, i.e. TGF(31, lead to the production of a relatively reduced set of specific transcripts.
Each of the growth factor induction experiments was repeated at least three times. RT-PCR'analysis was performed and a schematic representation of the results is shown in Figure 3-B. Induction or repression of gene expression is color-coded and the growth factors were ordered based on their overall induction or repression of cell specific expression.
Thus, on the left is the factor (NGF) that induces the greatest variety of cell specific gene expression of the factors tested, and on the right are the factors (BMP-4, Activin-A and TGF(31) that mostly repress or fail to direct differentiation of as many cell types.
Example 3: Characterizing derivative cells from EBs for neuronal differentiation.
Cell culture. Human ES cells were grown and EBs were generated as described in Example 1. Four days following initiation of aggregation growth factors were administered: retinoic acid (Sigma): 10'7M (Bain et al (1995) or 10-GM (Bain et al., 1996); TGF~1 (Sigma): 2ng/ml (Stager et al., (1993) Dev. Gefaet., Vo1.14, pp. 212-224.); (3NGF (New Biotechnology, Israel):
100ng/ml (Wobus et al., 1988). After 21 days, EBs were plated on 5p.g/cm2 collagen treated plates, either as whole EB's, or as single cells dissociated with trypsinBDTA.
The cultures were maintained for an additional week or 2 days respectively.
In-situ hybridization and immunohistochemistry was carried out according to Example 1. RT-PCR analysis. cDNA was synthesized from total RNA and PCR
performed according to Example 1. Primers for GAPDH, serotonin receptors 2A and 5A
(5HT2A, 5HT5A respectively) and dopamine receptor D1 (DRDl) were purchased from Clontech. The primers for dopa decarboxylase (DDC) were: (SEQ ID N0:2) TCTGTGCCTCTTAACTGTCACTGTGG and (SEQ ID NO: 3) ATCATCACAGTCTCCAGCTCTGTGC. The results are shown in Figures 6-8.
Differentiation was initially assayed by ih-situ hybridization to neurofilament light chain (NF-L) RNA, which is expressed by both immature and fully differentiated neurons (M.J.
Carden, et al., (1987) J. Neurosci., Vol. 7, pp. 3489-3504). We found that (3NGF and high concentration of RA increased the proportion of neuronal cells in the cultures from 21% in controls to 39% and 52% respectively. A lower concentration of RA (10-7M
instead of 10-6M) produced an effect similar to NGF, whereas no effect on neuronal differentiation was observed with TGF(31 treatment (Fig. 6).

Immunostaining revealed neurofilament heavy chain (NF-H) protein, found only in mature neurons (Garden et al.), in sections of mature EBs (Fig. 7a). When the EBs were plated, these neurons formed filamentous "networks" (Fig. 7b) of various complexities which were stained positive for NF-H (Fig. 7c). Treatment with RA and consequent plating caused an increase in the number of EBs containing NF-H positive neurons to 76%, compared to 54% in untreated EB's. Moreover, the RA-treated EBs showed dramatic shift towards more complex network morphology (Fig. 7d) suggesting a positive effect of the growth factor on the differentiation status of the cells.
To further characterize the nerve cells obtained from human ES cells, we assayed for to expression of a dopamine receptor (DRDl) and two serotonin receptors (5HT2A
and 5HT5A) in EBs treated with RA and in ES cells as a negative control (Fig. 8). These receptors were detected in EBs following RA treatment, but not in naive ES cells. Since dopamine and serotonin receptors were expressed, it was interesting to determine whether neurons in the cultures also synthesized their neurotransmitter ligands. Consistent with this possibility, we observed the expression of dope decarboxylase (DDC) a key enzyme in the synthesis of both serotonin and dopamine (Swoboda et aL, (1999) Neurology, Vol. 53, pp. 1205-1211) (Fig. 8).
All references recited above are herein incorporated by reference. The particular embodiments of the invention described above are not intended to limit the scope of the present invention which will be limited only by the appended claims.

SEQUENCE LISTING
<110> Benvenisty, Nissim <120> Directed Differention of Embryonic Stem Cells <130> 1822/113 <160> 73 <170> FastSEQ for Windows Version 4.0 <210> 1 <211> 50 <212> DNA
<213> Homo sapien <220>
<223> 50-mer 2'-O-methyl 5-biotinylated cDNA probe of NF-L
<400> 1 cctgcgtgcg gatggacttg aggtcgttgc tgatggcggc tacctggctc 50 <210> 2 <211> 26 <212> DNA
<213> Homo sapien <220>
<223> DNA primer for human dopa decarboxylase <400> 2 tctgtgcctc ttaactgtca ctgtgg 26 <220> 3 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> DNA primer for human dopa decarboxylase <400> 3 atcatcacag tctccagctc tgtgc 25 <210> 4 <211> 20 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of alpha-feto protein <400> 4 agaacctgtc acaagctgtg 20 <210> 5 <211> 20 <212> DNA
<213> Homo sapien <220>

<223>
3' primer of alpha-feto protein <400>

gacagcaagctgaggatgtc 20 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of alpha 1 anti-trypsin <400>

agaccctttgaagtcaagga caccg 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of alpha 1 anti-trypsin <400>

ccattgctgaagaccttagt gatgc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Activin Receptor type llB

<400>

acacgggagtgcatctacta caacg 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Activin Receptor type llB

<400>

ttcatgagctgggccttcca gacac 25 <210> 10 <211> 26 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Albumin <400> 10 cctttggcac aatgaagtgg gtaacc 26 <210> 11 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Albumin <400> 11 cagcagtcag ccatttcacc atagg 25 <210> 12 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Amylase <400> 12 gctgggctca gtattcccca aatac 25 <210> 13 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Amylase <400> 13 gacgacaatc tctgacctga gtagc 25 <210> 14 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Beta-Actin <400> 14 tggcaccaca ccttctacaa tgagc 25 <210> 15 <211> 25 <212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Beta-Actin <400>

gcacagcttctccttaatgt cacgc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Beta-Globin <400>

acctgactcctgaggagaag tctgc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Beta-Globin <400>

tagccacaccagccaccact ttctg 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Bone Morphogenic Protein 4 Receptor type 11 <400>

tctgcagctaggtcctctca tcagc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Bone Morphogenic Protein 4 Receptor type 11 <400>

tatactgctccatatcgacc tcggc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Cardiac Actin <400>

tctatgagggctacgctttg <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Cardiac Actin <400>

cctgactggaaggtagatgg 20 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Cartilage Matrix Protein <400>

atgactgtgagcaggtgtgc atcag 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Cartilage Matrix Protein <400>

ctggttgatggtcttgaagt cagcc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Delta-Globin <400>

accatggtgcatctgactcc tgagg 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223> 3' primer of Delta-Globin <400> 25 acttgtgagc caaggcatta gccac 25 <210> 26 <211> 26 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Dopamine Beta Hydroxylase <400> 26 cacgtactgg tgctacatta aggagc 26 <210> 27 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Dopamine Beta Hydroxylase <400> 27 aatggccatc actggcgtgt acacc 25 <210> 28 <211> 24 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Enolase <400> 28 tgacttcaag tcgcctgatg atcc 24 <210> 29 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Enolase <400> 29 tgcgtccagc aaagattgcc ttgtc 25 <210> 30 <211> 26 <212> DNA

<213> Homo sapien <220>
<223> 5' primer of Epidermal Growth Factor Receptor type <400> 30 cagtcgtcag cctgaacata 26 acatcc <210> 31 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of EpidermalGrowth Factor Receptor type <400> 31 aggttgcact tgtccacgca 25 ttccc <210> 32 <211> 26 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of FibroblastGrowth Factor Receptor type I

<400> 32 agcatcataa tggactctgt 26 ggtgcc <210> 33 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of FibroblastGrowth Factor Receptor type I

<400> 33 agtccgatag agttacccgc 25 caagc <210> 34 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of FollicularStimulating Hormone <400> 34 gtgagctgac caacatcacc 25 attgc <210> 35 <211> 25 <212> DNA

<213> Homo sapien <220>
<223> 3' primer of Follicular Stimulating Hormone <400> 35 tttcaccaaa ggagcagtag ctggg 25 <210> 36 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of Glucagen <400> 36 ctcagtgatc ctgatcagat gaacg 25 <210> 37 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 3' primer of Glucagen <400> 37 agtccctggc ggcaagatta tcaag 25 <210> 38 <211> 26 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of Glyceraldehyde 3-phosphate dehydrogenase <400> 38 tgaaggtcgg agtcaacgga tttggt 26 <210> 39 <211> 24 <212> DNA
<213> Homo sapien <220>
<223> 3' primer of Glyceraldehyde 3-phosphate dehydrogenase <400> 39 catgtgggcc atgaggtcca ccac 24 <210> 40 <211> 26 <212> DNA
<213> Homo sapien <220>

<223> 5' primer of Hepatocyte Growth Factor Receptor <400> 40 agaaattcat caggctgtga agcgcg 26 <210> 41 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Hepatocyte Growth Factor Receptor <400> 41 ttcctccgat cgcacacatt tgtcg 25 <210> 42 <211> 24 <212> DNA

<2~3> Homo sapien <220>

<223> 5' primer of Insulin <400> 42 ctgcatcaga agaggccatc aagc 24 <210> 43 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Insulin <400> 43 ggctttattc catctctctc ggtgc 25 <210> 44 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Kallikrein <400> 44 gttcatgtca gtgagagctt cccac 25 <210> 45 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Kallikrein <400> 45 tcacataaga cagcactctg acggc 25 <210> 46 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of Keratin <400> 46 aggaaatcat ctcaggagga agggc 25 <210> 47 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 3' primer of Keratin <400> 47 aaagcacaga tcttcgggag ctacc 25 <210> 48 <211> 24 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of Lipase <400> 48 gattcatcaa gcatcagtgg ctcc 24 <210> 49 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 3' primer of Lipase <400> 49 ccaatcggac taattcaggt gtgcc 25 <210> 50 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of Myosin light polypeptide2 <400> 50 tccaacgtgt tctccatgtt cgaac 25 <210> 51 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Myosin polypeptide2 light <400> 51 cttgtagtcc aagttgccag tcacg 25 <210> 52 <211> 26 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Nerve Factor Receptor Growth <400> 52 tgttctcctg ccaggacaag cagaac 26 <210> 53 <211> 26 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Nerve Factor Receptor Growth <400> 53 tcttgaaggc tatgtaggcc acaagg 26 <210> 54 <211> 24 <212> DNA

<213> Homo sapien <220>

<223> 5' primer of Neurofilamentheavy Chain <400> 54 tgaacacaga cgctatgcgc tcag 24 <210> 55 <211> 25 <212> DNA

<213> Homo sapien <220>

<223> 3' primer of Neurofilament heavy Chain <400> 55 cacctttatg tgagtggaca cagag 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Octamer Binding Protein <400>

cgagaagctggagaaggaga agctg 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Octamer Binding Protein <400>

caagggccgcagcttacaca tgttc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Parathyroid Hormone <400>

ggctaaagttatgattgtca tgttggc 27 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Parathyroid Hormone <400>

tcagctttgtctgcctctcc aagac 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of PDX-1 <400>

ggatgaagtctaccaaagct cacgc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of PDX-1 <400>

ccagatcttgatgtgtctct cggtc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Phosphoprotein enriched in astrocytes <400>

agagtgaggagatcactact ggcag 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Phosphoprotein enriched in astrocytes <400>

acctgctggtactcaggaaa cagtc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Renin <400>

agtcgtctttgacactggtt cgtcc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Renin <400>

ggtagaacctgagatgtagg atgc <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Retinoic Acid Receptor type alpha <400>

agcagcagttctgaagagat agtgcc 26 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Retinoic Acid Receptor type alpha <400>

gtggagagttcactgaactt gtccc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Surfactant <400>

tccagctcatctagatgagg agctc 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
3' primer of Surfactant <400>

gtcccatggcctaaatgcct ctcag 25 <210>

<211>

<212>
DNA

<213> sapien Homo <220>

<223>
5' primer of Transforming Growth Factor Receptor type <400>

tagtcactgacaacaacggt gcagtc 26 <210>

<211>

<212>
DNA

<213> sapien Homo <220>
<223> 3' ' primer of Surfactant <400> 71 acagtgctcg ctgaactcca tgagc 25 <210> 72 <211> 24 <212> DNA
<213> Homo sapien <220>
<223> 5' primer of WT 1 <400> 72 tccttcatca aacaggagcc gagc 24 <210> 73 <211> 25 <212> DNA
<213> Homo sapien <220>
<223> 3' primer of WT 1 <400> 73 ctgtagggcg tcctcagcag caaag 25

Claims (47)

What is claimed is:
1. A method of mapping a pathway of differentiation of a population of embryonic cells, comprising:
(i) selecting: (a) a set of gene expression products, wherein each gene expression product in the set is characteristic of a cell type that has undergone differentiation, such that a plurality of differentiated cell types are represented in the set; and (b) an exogenous factor from a library of exogenous factors;
(ii) applying the exogenous factor to the population of embryonic cells;
(iii) characterizing the effect of the exogenous factor on the differentiation pathway of the population of cells by determining gene expression products in the set; and (iv) mapping the pathway of differentiation of the cells.
2. A method according to claim 1, wherein the set of expression products comprises at least one gene expression product that is expressed in a tissue selected from the mesoderm, ectoderm and endoderm.
3. A method according to claim 1, wherein the set of expression products, comprises: at least one gene expression product expressed in the ectoderm, at least one gene expression product expressed in the mesoderm and at least one gene expression product expressed in the endoderm.
4. A method according to claim 2 or 3, wherein the at least one gene expression product expressed in the ectoderm includes any of the group consisting of NF-H, keratin and adrenal D~H.
5. A method according to claim 2 or 3, wherein the at least one gene expression product expressed in the mesoderm includes any of the group consisting of enolase, renin, CMP, kallikrein, WTI, cACT, .delta.-globulin and ~-globulin.
6. A method according to claim 2 or 3, wherein the at least one gene expression product expressed in the endoderm includes any of the group consisting of albumin, .alpha.1AT, amylase, PDX-1, insulin and .alpha.FP.
7. A method according to claim 1, wherein the exogenous factor is selected from a group consisting of interleukins, bFGF, TGF~1, activin-A, BMP-4, HGF, EGF, ~NGF and retinoic acid.
8. A method of directing differentiation of human embryonic cells to a specific cell type, comprising:
a. permitting a population of embryonic stem cells to form embryoid bodies;
b. dissociating the embryoid bodies to provide embryonic cells for differentiating in the presence of at least one exogenous factor for an effective period of time;
and c. causing directed differentiation of human embryonic cells to form the specific cell type
9. A method according to claim 8, wherein the embryoid bodies are formed in a suspension culture.
10. A method according to claim 8, wherein the embryonic cells are monolayer cultures.
11. A method according to claim 8, wherein the exogenous factor is a growth factor.
12. A method according to claim 8, wherein the exogenous factor is an interleukin.
13. A method according to claim 11, wherein the exogenous factor is nerve growth factor.
14. A method according to claim 8, wherein the exogenous factor is retinoic acid.
15. A method according to claim 8, wherein the differentiated cells are neuronal cell type.
16. A method according to claim 15, wherein the differentiated cells have neuronal processes.
17. A method according to claim 1, wherein the embryonic cells are human embryonic cells.
18. A method of treating a subject suffering from a condition associated with degeneration of cells or malfunction of cells, comprising:
a. accessing human embryoid body dissociated cells;
b. treating the cells with an exogenous factor;
c. causing the cells to differentiate; and d. placing an effective amount of differentiated cells into the subject to treat the condition.
19. A method according to claim 18, wherein the condition is a heart condition in which heart muscle is degenerated.
20. A method according to claim 19, wherein the cells are treated with at least one exogenous factor selected from the group consisting of TGF-~, FGF, RA, HGF and EGF.
21. A method according to claim 18, wherein the condition is a kidney condition in which kidney tissue is degenerated.
22. A method according to claim 21, wherein the cells are treated with NGF.
23. A method according to claim 18, wherein the condition is a skin condition in which skin tissue is degenerated.
24. A method according to claim 23, wherein the cells are treated with BMP-4.
25. A method according to claim 18, wherein the condition is a liver condition in which liver tissue is degenerated.
26. A method according to claim 25, wherein the cells are treated with NGF.
27. A method according to claim 18, wherein the condition is a brain condition in which brain tissue is degenerated.
28. A method according to claim 27, wherein the cells are treated with at least one of NGF or RA.
29. A method according to claim 18, wherein the condition is a spinal cord injury in which neurons are degenerated.
30. A method according to claim 29, wherein the cells are treated with at least one of NGF or RA.
31. A method according to claim 18, wherein the condition is anemia or immunodeficiency.
32. A method according to claim 18, wherein the cells are treated with at least one of NGF or interleukin.
33. A method according to claim 18, wherein the condition is an adrenal condition in which adrenal tissue is degenerated.
34. A method according to claim 33, wherein the cells are treated with RA.
35. A method according to claim 18, wherein the subject is a human subject.
36. A method according to claim 18, wherein the subject is a mammal.
37. A method according to claim 18, wherein the selected cell type may be any of brain cells, liver cells, pancreatic cells, muscle cells, chondrocytes, kidney cells, Mullerian duct cells, heart cells, blood cells, skin cells and adrenal cells.
38. A kit for determining differentiation pathways, comprising:
(a) a plurality of sets of cell specific markers forming a panel in an assay format, the assay format including reagents for detecting the cell specific markers, the cell specific markers including a first set of markers that are characteristic of each of the ectoderm, mesoderm and endoderm of the embryo, and a second set of markers that are characteristic of a body tissue, the second set containing more than one marker; and (b) means for detecting reagents bound to cell specific markers.
39. A kit according to claim 38, wherein the reagents are selected from DNA
primers and antibodies.
40. A method according to claim 38, wherein the means for detecting reagents bound to cell specific markers is selected form a gel based system, an immune detection assay and a solid chemistry assay.
41. A method for screening an exogenous factor to determine whether the factor is capable of causing directed differentiation in a population of human embryonic cells, comprising:
(a) subjecting the population of cells to the exogenous growth factor;
(b) measuring the expression of receptors, the receptors being of a type that characterizes a particular differentiated cell population; and (c) determining whether the exogenous factor enhances differentiation, maintains differentiation at a normal level or inhibits differentiation of the cell population.
42. A panel of cell type differentiation determining markers, comprising: a set of reagents for specifically detecting gene expression of a plurality of ectodermal specific proteins, a plurality of mesodermal specific proteins and a plurality of endodermal specific proteins.
43. A panel according to claim 40, wherein the ectodermal specific proteins, further comprise: proteins selected from the group consisting of: neurofilament protein, keratin and adrenal dopamine .beta. hydroxylase.
44. A panel according to claim 40, wherein the mesodermal specific proteins, further comprise: proteins selected from the group consisting of enolase, CMP, renin, kallikrein, WTI, cACT, .beta. globin, .delta. globin and cActin.
45. A panel according to claim 40, wherein the endodermal specific proteins further comprise: proteins selected from the group consisting of amylase, .alpha.-FP, PDX-1 and insulin.
46. A panel according to claim 40, wherein the reagents are DNA primers.
47. A panel according to claim 40, wherein the reagents are antibodies.
CA002417356A 2000-08-01 2001-07-31 Directed differentiation of embryonic cells Abandoned CA2417356A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US22216000P 2000-08-01 2000-08-01
US60/222,160 2000-08-01
US26755901P 2001-02-09 2001-02-09
US60/267,559 2001-02-09
PCT/IB2001/001719 WO2002010347A2 (en) 2000-08-01 2001-07-31 Directed differentiation of embryonic cells

Publications (1)

Publication Number Publication Date
CA2417356A1 true CA2417356A1 (en) 2002-02-07

Family

ID=26916492

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002417356A Abandoned CA2417356A1 (en) 2000-08-01 2001-07-31 Directed differentiation of embryonic cells

Country Status (7)

Country Link
US (2) US7045353B2 (en)
EP (1) EP1315835A2 (en)
JP (1) JP2004504834A (en)
AU (2) AU8617301A (en)
CA (1) CA2417356A1 (en)
IL (2) IL154159A0 (en)
WO (1) WO2002010347A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9334478B2 (en) 2001-08-24 2016-05-10 Advanced Cell Technology, Inc. Differentiating ES cells using a tenascin

Families Citing this family (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5843780A (en) * 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
IL142094A0 (en) 1998-09-29 2002-03-10 Gamida Cell Ltd Methods of controlling proliferation and differentiation of stem and progenitor cells
IL129966A (en) * 1999-05-14 2009-12-24 Technion Res & Dev Foundation ISOLATED HUMAN EMBRYOID BODIES (hEB) DERIVED FROM HUMAN EMBRYONIC STEM CELLS
US7473555B2 (en) * 2000-04-27 2009-01-06 Geron Corporation Protocols for making hepatocytes from embryonic stem cells
FR2825370A1 (en) * 2001-05-31 2002-12-06 Inst Nat Sante Rech Med KERATINOCYTES OBTAINED FROM MAMMALIAN EMBRYONIC STEM CELLS
US7732199B2 (en) * 2001-07-12 2010-06-08 Geron Corporation Process for making transplantable cardiomyocytes from human embryonic stem cells
US7425448B2 (en) * 2001-07-12 2008-09-16 Geron Corporation Cardiomyocyte precursors from human embryonic stem cells
IL159895A0 (en) * 2001-07-20 2004-06-20 Technion Res & Dev Foundation Methods of generating human cardiac cells and tissues and uses thereof
IL152904A0 (en) 2002-01-24 2003-06-24 Gamida Cell Ltd Utilization of retinoid and vitamin d receptor antagonists for expansion of renewable stem cell populations
WO2003062404A1 (en) 2002-01-25 2003-07-31 Gamida-Cell Ltd. Methods of expanding stem and progenitor cells and expanded cell populations obtained thereby
EP1485464A4 (en) * 2002-03-18 2006-11-29 Gamida Cell Ltd Methods of inducing differentiation in ex vivo expanded stem cells
US20060003446A1 (en) * 2002-05-17 2006-01-05 Gordon Keller Mesoderm and definitive endoderm cell populations
CA2684022C (en) 2002-05-17 2014-09-23 Mount Sinai School Of Medicine Of New York University Mesoderm and definitive endoderm cell populations
US20060121607A1 (en) * 2002-08-08 2006-06-08 Thomas Schulz Compositions and methods for neural differentiation of embryonic stem cells
US20050054097A1 (en) * 2002-11-17 2005-03-10 Tony Peled EX-VIVO expansion of hematopoietic system cell populations in mononuclear cell cultures
US20040214324A1 (en) * 2002-12-09 2004-10-28 Ole Isacson Dopaminergic neurons differentiated from embryonic cells for treating neurodegenerative diseases
US20050031598A1 (en) * 2002-12-10 2005-02-10 Shulamit Levenberg Engineering three-dimensional tissue structures using differentiating embryonic stem cells
US20060211111A1 (en) * 2002-12-18 2006-09-21 Maisam Mitalipova Compositions and methods for neural cell production and stabilization
US20030224411A1 (en) * 2003-03-13 2003-12-04 Stanton Lawrence W. Genes that are up- or down-regulated during differentiation of 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
US20070161105A1 (en) * 2003-03-20 2007-07-12 Reasearch Foundation Itsuu Laboratory Method of forming organ
IL161903A0 (en) * 2003-07-17 2005-11-20 Gamida Cell Ltd Ex vivo progenitor and stem cell expansion for usein the treatment of disease of endodermally- deri ved organs
US8647873B2 (en) 2004-04-27 2014-02-11 Viacyte, Inc. PDX1 expressing endoderm
US7985585B2 (en) 2004-07-09 2011-07-26 Viacyte, Inc. Preprimitive streak and mesendoderm cells
AU2013200203B2 (en) * 2003-12-23 2014-11-20 Viacyte, Inc. Definitive endoderm
US20050266554A1 (en) 2004-04-27 2005-12-01 D Amour Kevin A PDX1 expressing endoderm
US7541185B2 (en) 2003-12-23 2009-06-02 Cythera, Inc. Methods for identifying factors for differentiating definitive endoderm
WO2005116073A2 (en) * 2004-04-27 2005-12-08 Cythera, Inc. Pdx1 expressing endoderm
WO2007059007A2 (en) 2005-11-14 2007-05-24 Cythera, Inc. Markers of definitive endoderm
DK2722387T3 (en) 2003-12-23 2020-01-20 Viacyte Inc Definitely endoderm
US7625753B2 (en) 2003-12-23 2009-12-01 Cythera, Inc. Expansion of definitive endoderm cells
WO2005090558A1 (en) * 2004-03-19 2005-09-29 Geron Corporation Method for making high purity cardiomyocyte preparations suitable for regenerative medicine
US7452718B2 (en) * 2004-03-26 2008-11-18 Geron Corporation Direct differentiation method for making cardiomyocytes from human embryonic stem cells
CA2560334A1 (en) 2004-04-07 2005-10-20 Axiogenesis Ag Non-invasive, in vitro functional tissue assay systems
EP2377922B1 (en) * 2004-04-27 2020-04-08 Viacyte, Inc. PDX1 expressing endoderm
CA2565858C (en) 2004-05-11 2021-06-22 Axiogenesis Ag Assay for drug discovery based on in vitro differentiated cells
US8637065B2 (en) 2004-07-09 2014-01-28 William Marsh Rice University Dermis-derived cells for tissue engineering applications
AU2012202529B2 (en) * 2004-07-09 2015-04-02 Viacyte, Inc. Preprimitive streak and mesendoderm cells
CA2573283C (en) * 2004-07-09 2023-03-14 Cythera, Inc. Methods for identifying factors for differentiating definitive endoderm
MX2007000317A (en) * 2004-07-09 2007-03-26 Cythera Inc Preprimitive streak and mesendoderm cells.
EP1791952A4 (en) 2004-08-13 2008-06-11 Univ Georgia Res Found Compositions and methods for self-renewal and differentiation in human embryonic stem cells
WO2006030442A2 (en) 2004-09-16 2006-03-23 Gamida-Cell Ltd. Methods of ex vivo progenitor and stem cell expansion by co-culture with mesenchymal cells
AU2005289088B2 (en) * 2004-09-29 2011-04-07 Cellartis Ab Methods for the generation of hepatocyte-like cells from human blastocyst-derived stem (hBS)
EP1677131B1 (en) * 2004-12-30 2011-10-05 Proximion Fiber Systems AB Optical coupler with fibre Bragg grating and Fabry Perot cavity and method of using it
AU2006210955A1 (en) * 2005-01-31 2006-08-10 Es Cell International Pte Ltd. Directed differentiation of embryonic stem cells and uses thereof
JP5560393B2 (en) * 2005-06-22 2014-07-23 アステリアス バイオセラピューティクス インコーポレイテッド Differentiation of primate pluripotent stem cells into cardiomyocyte lineage cells
WO2007051038A2 (en) 2005-10-27 2007-05-03 Cythera, Inc. Pdx1-expressing dorsal and ventral foregut endoderm
US8846393B2 (en) 2005-11-29 2014-09-30 Gamida-Cell Ltd. Methods of improving stem cell homing and engraftment
US7695965B2 (en) 2006-03-02 2010-04-13 Cythera, Inc. Methods of producing pancreatic hormones
EP1999253B1 (en) 2006-03-02 2019-05-22 Viacyte, Inc. Endocrine precursor cells, pancreatic hormone-expressing cells and methods of production
US11254916B2 (en) 2006-03-02 2022-02-22 Viacyte, Inc. Methods of making and using PDX1-positive pancreatic endoderm cells
EP2007875A4 (en) * 2006-04-05 2009-12-16 Univ Rice William M Dermis-derived cells for tissue engineering applications
KR20140146224A (en) 2006-04-14 2014-12-24 어드밴스드 셀 테크놀로지, 인코포레이티드 Hemangio-colony forming cells
WO2007127454A2 (en) 2006-04-28 2007-11-08 Cythera, Inc. Hepatocyte lineage cells
CA2650561C (en) * 2006-05-02 2014-02-25 Wisconsin Alumni Research Foundation Method of differentiating stem cells into cells of the endoderm and pancreatic lineage
US8685730B2 (en) 2006-05-02 2014-04-01 Wisconsin Alumni Research Foundation Methods and devices for differentiating pluripotent stem cells into cells of the pancreatic lineage
WO2007143117A2 (en) * 2006-06-02 2007-12-13 Geron Corporation Differentiation of primate pluripotent cells to hepatocyte-lineage cells
US8874380B2 (en) 2010-12-09 2014-10-28 Rutgers, The State University Of New Jersey Method of overcoming therapeutic limitations of nonuniform distribution of radiopharmaceuticals and chemotherapy drugs
US20080003681A1 (en) * 2006-06-28 2008-01-03 Mahalaxmi Gita Bangera Methods for altering cellular susceptibility to infection
GB0622394D0 (en) * 2006-11-09 2006-12-20 Univ Cambridge Tech Differentiation of pluripotent cells
US7695963B2 (en) 2007-09-24 2010-04-13 Cythera, Inc. Methods for increasing definitive endoderm production
KR20110005682A (en) 2008-01-30 2011-01-18 제론 코포레이션 Synthetic surfaces for culturing stem cell derived cardiomyocytes
US20110086424A1 (en) 2008-05-06 2011-04-14 Advanced Cell Technology, Inc. Methods for producing enucleated erythroid cells derived from pluripotent stem cells
EP2291513A4 (en) 2008-05-06 2012-02-29 Advanced Cell Tech Inc Hemangio colony forming cells and non-engrafting hemangio cells
CA3229301A1 (en) 2008-11-14 2010-05-20 Viacyte, Inc. Encapsulation of pancreatic cells derived from human pluripotent stem cells
US8558913B2 (en) * 2010-02-08 2013-10-15 Apple Inc. Capture condition selection from brightness and motion
CN103459611B (en) * 2010-09-17 2016-11-02 哈佛大学校长及研究员协会 The functional genomics research that effectiveness and the safety of pluripotent stem cell are characterized
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
WO2013121426A1 (en) 2012-02-13 2013-08-22 Gamida-Cell Ltd. Culturing of mesenchymal stem cells
US9175266B2 (en) 2012-07-23 2015-11-03 Gamida Cell Ltd. Enhancement of natural killer (NK) cell proliferation and activity
US9567569B2 (en) 2012-07-23 2017-02-14 Gamida Cell Ltd. Methods of culturing and expanding mesenchymal stem cells
CN114558032A (en) 2012-12-21 2022-05-31 安斯泰来再生医药协会 Method for preparing platelets from pluripotent stem cells and composition thereof
US11085067B2 (en) 2013-06-10 2021-08-10 President And Fellows Of Harvard College Early developmental genomic assay for characterizing pluripotent stem cell utility and safety
US10160946B2 (en) 2013-09-13 2018-12-25 University Of Florida Research Foundation, Inc. Pluripotent tissue harvester and methods of manufacture thereof
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
US11771068B2 (en) 2015-04-08 2023-10-03 National Federation Of Agricultural Cooperative Associations Method for producing non-human large mammal or fish each capable of producing gamete originated from different individual
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH04506153A (en) 1989-06-15 1992-10-29 ザ・リージエンツ・オブ・ザ・ユニバーシテイ・オブ・ミシガン Cell proliferation methods, compositions and devices for cell proliferation
US5763266A (en) 1989-06-15 1998-06-09 The Regents Of The University Of Michigan Methods, compositions and devices for maintaining and growing human stem and/or hematopoietics cells
ES2076378T5 (en) 1989-09-25 1999-10-16 Genetics Inst METHOD FOR THE INHIBITION OF THE DEVELOPMENT OF STEM CELLS.
US5453357A (en) 1992-10-08 1995-09-26 Vanderbilt University Pluripotential embryonic stem cells and methods of making same
FR2726003B1 (en) 1994-10-21 2002-10-18 Agronomique Inst Nat Rech CULTURE MEDIUM OF AVIAN TOTIPOTENT EMBRYONIC CELLS, METHOD FOR CULTURING THESE CELLS, AND AVIAN TOTIPOTENT EMBRYONIC CELLS
JPH10509314A (en) 1994-11-08 1998-09-14 ブラッドリー マイケル ジョン ストリンガー, Human cell line
AU4116796A (en) 1994-11-14 1996-06-06 Sandoz Ltd. Assay for the measurement of multilineage stem cells and methods of use thereof
US5874301A (en) 1994-11-21 1999-02-23 National Jewish Center For Immunology And Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5914268A (en) 1994-11-21 1999-06-22 National Jewish Center For Immunology & Respiratory Medicine Embryonic cell populations and methods to isolate such populations
EP0796110A2 (en) 1994-12-09 1997-09-24 Joseph D. Mosca System for the maintenance, growth and differentiation of human and non-human primate pluripotent stem, progenitor and mature bone marrow cells
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5736396A (en) * 1995-01-24 1998-04-07 Case Western Reserve University Lineage-directed induction of human mesenchymal stem cell differentiation
DE19530746A1 (en) * 1995-08-22 1997-02-27 Bosch Gmbh Robert Circuit arrangement for operating a high-pressure gas discharge lamp with alternating current
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
CA2277278A1 (en) 1997-01-10 1998-07-16 Life Technologies, Inc. Embryonic stem cell serum replacement
US6331406B1 (en) 1997-03-31 2001-12-18 The John Hopkins University School Of Medicine Human enbryonic germ cell and methods of use
US6090622A (en) 1997-03-31 2000-07-18 The Johns Hopkins School Of Medicine Human embryonic pluripotent germ cells
US5914266A (en) * 1997-05-06 1999-06-22 Chiron Corporation Mammallian sex comb on midleg (mammalian SCM) acts as a tumor suppressor
AU1197699A (en) 1997-10-23 1999-05-10 Geron Corporation Methods and materials for the growth of primate-derived primordial stem cells
DE19756864C5 (en) 1997-12-19 2014-07-10 Oliver Brüstle Neural precursor cells, methods for their production and their use for the therapy of neural defects
AU2333999A (en) 1998-01-23 1999-08-09 Prolifaron, Inc. Methods and compositions for the identification of growth factor mimetics, growth factors and inhibitors
AU774581B2 (en) 1998-08-31 2004-07-01 New York University Stem cells bearing an FGF receptor on the cell surface
WO2000017326A1 (en) 1998-09-21 2000-03-30 Musc Foundation For Research Development Non-hematopoietic cells, including cardiomyocytes and skeletal muscle cells, derived from hematopoietic stem cells and methods of making and using them
EP1016413A1 (en) 1998-12-30 2000-07-05 Applied Research Systems ARS Holding N.V. Human growth hormone to stimulate mobilization of pluripotent hematopoietic stem cells
IL129966A (en) 1999-05-14 2009-12-24 Technion Res & Dev Foundation ISOLATED HUMAN EMBRYOID BODIES (hEB) DERIVED FROM HUMAN EMBRYONIC STEM CELLS
AU759643B2 (en) 1999-07-20 2003-04-17 University Of Southern California Identification of pluripotent pre-mesenchymal, pre-hematopoietic progenitor cell
US6602711B1 (en) 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9334478B2 (en) 2001-08-24 2016-05-10 Advanced Cell Technology, Inc. Differentiating ES cells using a tenascin

Also Published As

Publication number Publication date
US20020146678A1 (en) 2002-10-10
US7045353B2 (en) 2006-05-16
JP2004504834A (en) 2004-02-19
AU8617301A (en) 2002-02-13
US20060128013A1 (en) 2006-06-15
US7772001B2 (en) 2010-08-10
AU2001286173B2 (en) 2007-10-25
WO2002010347A3 (en) 2003-03-13
IL154159A0 (en) 2003-07-31
IL154159A (en) 2010-06-30
WO2002010347A2 (en) 2002-02-07
EP1315835A2 (en) 2003-06-04

Similar Documents

Publication Publication Date Title
AU2001286173B2 (en) Directed differentiation of embryonic cells
AU2001286173A1 (en) Directed differentiation of embryonic cells
JP6644277B2 (en) Neural progenitor cell population
KR101247037B1 (en) Dopaminergic neurons and proliferation-competent precursor cells for treating parkinson&#39;s disease
Schuldiner et al. Induced neuronal differentiation of human embryonic stem cells
Renoncourt et al. Neurons derived in vitro from ES cells express homeoproteins characteristic of motoneurons and interneurons
AU2001263199A1 (en) Neural progenitor cell populations
JP2018521692A (en) Haploid human embryonic stem cell lines and somatic cell lines and methods for producing them
WO2023114754A1 (en) Methods, culture medias and devices for generating embryos in vitro from stem cells
Schuldiner et al. Factors controlling human embryonic stem cell differentiation
US20210332436A1 (en) Stem cell-based multiplex methods and compositions
JP2014528706A (en) Methods for identifying drugs that affect maturation, survival and myelination
WO2023118050A1 (en) Use of novel markers to detect pluripotent stem cells
AU2004214542B2 (en) Neural progenitor cell populations
Andoniadou Intrinsic factors controlling stem cell proliferation and differentiation in the central nervous system
Ruby Genetic manipulation of Fezf2 in human embryonic stem cells and its application in studying central nervous system development and repair
Wölfle Comparative analyses of the neurogenic capacity of human neuroprogenitor populations derived from neural and mesodermal tissue
Hewitt Epigenetic Regulation of Pluripotent Stem Cell Differentiation
Golan-Mashiach The Secret of Stem Cells

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued