US20100240090A1 - Methods and platforms for drug discovery - Google Patents

Methods and platforms for drug discovery Download PDF

Info

Publication number
US20100240090A1
US20100240090A1 US12/709,379 US70937910A US2010240090A1 US 20100240090 A1 US20100240090 A1 US 20100240090A1 US 70937910 A US70937910 A US 70937910A US 2010240090 A1 US2010240090 A1 US 2010240090A1
Authority
US
United States
Prior art keywords
cells
cell
ipsc
cases
days
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
US12/709,379
Inventor
Kazuhiro Sakurada
Hideki Masaki
Tetsuya Ishikawa
Shunichi Takahashi
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.)
Kyoto University
iZumi Bio Inc
Original Assignee
iZumi Bio Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by iZumi Bio Inc filed Critical iZumi Bio Inc
Priority to US12/709,379 priority Critical patent/US20100240090A1/en
Publication of US20100240090A1 publication Critical patent/US20100240090A1/en
Assigned to KYOTO UNIVERSITY reassignment KYOTO UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: IPIERIAN, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal 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
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • 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
    • C12N5/0696Artificially induced pluripotent stem cells, e.g. iPS
    • 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
    • C12N5/10Cells modified by introduction of foreign genetic material
    • 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
    • C12N2501/60Transcription factors
    • C12N2501/602Sox-2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/603Oct-3/4
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/604Klf-4
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/60Transcription factors
    • C12N2501/606Transcription factors c-Myc
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2510/00Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/027Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a retrovirus

Definitions

  • the present invention involves methods for identifying an agent that corrects a phenotype associated with a health condition or a predisposition for a health condition comprising contacting a first population of cells from a human induced pluripotent stem cell line, or cells differentiated from the human induced pluripotent stem cell line, with a candidate agent; contacting a second population of cells from a human induced pluripotent stem cell line, or cells differentiated from the human induced pluripotent stem cell line, with a control agent; wherein the cells in both populations comprise at least one endogenous allele associated with the health condition or predisposition for the health condition; assaying the two populations and identifying candidate agents as correcting the phenotype if the first population is closer to a normal phenotype following treatment than the second population.
  • the condition may be selected from health conditions such as a neurodegenerative disorder, a neurological disorder, a mood disorder, a cardiovascular disease, a metabolic disorder, a respiratory disease, a drug sensitivity condition, an eye disease, an immunological disorder, or a hematological disease.
  • the cells may be differentiated from induced stem cells to neural stem cells, neurons, cardiomyocytes, hepatic stem cells, or hepatocytes.
  • the phenotype described may be apoptosis, intracellular calcium level, calcium flux, protein kinase activity, enzyme activity, cell morphology, receptor activation, protein trafficking, intracellular protein aggregation, organellar composition, motility, intercellular communication, protein expression, or gene expression.
  • the invention also involves methods for identifying a diagnostic cellular phenotype comprising comparing a set of cells from a subject to cells from a subject free of the health condition wherein both sets of cells were induced pluripotent stem cells, or were cells differentiated from induced pluripotent stem cells, and wherein the comparison is performed on a computer.
  • the cells may be differentiated from induced stem cells to neural stem cells, neurons, cardiomyocytes, hepatic stem cells, or hepatocytes.
  • the invention also involves methods for determining the risk of a health condition in a subject comprising comparing at least one phenotype determined in a first set of cells derived from the subject to the at least one phenotype determined in a second set of cells derived from subjects free of the health condition and to the at least one phenotype determined in a third set of cells derived from subjects suffering from the health condition; and indicating that the subject is at high risk for the health condition if the at least one phenotype determined in the first set of cells is more similar to the at least one phenotype determined in the third set of cells than the at least one phenotype determined in the second set of cells, wherein the first, second, and third sets of cells were induced pluripotent stem cells, or were cells differentiated from induced pluripotent stem cells, and wherein the comparison is performed on a computer.
  • the invention also involves methods for reducing the risk of drug toxicity in a human subject, comprising contacting one or more cells differentiated from an induced pluripotent stem cell line generated from the subject with a dose of a pharmacological agent, assaying the contacted one or more differentiated cells for toxicity, and prescribing or administering the pharmacological agent to the subject if, and only if, the assay is negative for toxicity in the contacted cells.
  • the cells differentiated from the induced pluripotent stem cell line may be hepatocytes, cardiomyocytes, or neurons.
  • the invention also involves methods for identifying a candidate gene that contributes to a human disease, comprising comparing a global gene expression profile of cultured human cells of a differentiated cell type from a plurality of healthy individuals to a global gene expression profile of cultured human cells of the differentiated cell type from a plurality of individuals suffering from the human disease and identifying one or more genes that have different expression levels as candidate genes that contribute to the human disease, wherein the comparison is performed on a computer.
  • the invention also discloses a human induced pluripotent stem cell line generated from a subject diagnosed as suffering from a health condition, or comprising at least one endogenous allele associated with a health condition or a predisposition for the health condition.
  • the invention also discloses an isolated population of human cells comprising neural stem cells or neurons from a subject having at least one endogenous allele associated with a neurodegenerative disorder, a neurological disorder, or a mood disorder, or from a subject diagnosed with the neurodegenerative disorder, neurological disorder, or mood disorder.
  • the invention also discloses an isolated population of human cells comprising human cardiac progenitor cells or cardiomyocytes from a subject having at least one endogenous allele associated with a cardiovascular disease, or from a subject diagnosed with the cardiovascular disease.
  • the invention also discloses an isolated population of human cells comprising hepatic stem cells or hepatocytes from a subject having at least one endogenous allele associated with a drug sensitivity condition, or from a subject diagnosed with the drug sensitivity condition.
  • the invention further discloses a panel of genetically diverse human induced pluripotent stem cell lines, comprising human induced pluripotent stem cell lines generated from a plurality of individuals each of which carry at least one polymorphic allele that is unique among the plurality of individuals.
  • FIG. 1 is a schematic comparison of a traditional drug discovery scheme (left) in which lead compounds are tested against a disease target in heterologous systems (e.g., animal models) prior to testing compound efficacy and safety in patients versus a new drug discovery paradigm (right) in which lead compounds are first identified based on their efficacy in correcting a disease-relevant cellular phenotype in patient-derived, disease-relevant cell types.
  • heterologous systems e.g., animal models
  • FIG. 2 is an overview of an exemplary, non-limiting, scheme for patient iPSC-based disease modeling and drug discovery.
  • FIG. 3 is an overview of an exemplary, non-limiting, scheme for patient iPSC-based testing of lead drug candidate efficacy and safety in cells from a genetically diverse cohort of patient iPSC lines.
  • FIG. 4 is an overview of an exemplary, non-limiting, scheme for patient iPSC-based identification of predictive biomarkers for drug efficacy and toxicity.
  • biomarkers are used in, e.g., patient stratification for clinical trials of drug candidates, and also for optimal dosing and safety of approved therapeutics in specific patients or patient populations, which is sometimes referred to as “personalized medicine.”
  • FIG. 5 shows photomicrographs of fibroblasts from three SMN1 ⁇ / ⁇ SMA patients and two SMN1 ⁇ /+ healthy control subjects; (Bottom Panel) shows photomicrographs of iPSC colonies derived from the corresponding SMA case and control subject fibroblasts illustrated in the top panel.
  • FIG. 6 shows photomicrographs of embryoid bodies obtained from the SMA case and control iPSC lines shown in FIG. 5 .
  • FIG. 7 shows immunofluorescence photomicrographs of staining for ectodermal (TuJ1), mesodermal (Desmin), and endodermal (AFP) lineage markers in cells differentiated from SM10d iPSCs.
  • Genetic variations within and among human patient populations underlie, to a large extent, differences in individual disposition to diseases, disease manifestation, disease severity, and response to treatment (e.g., to drug treatment).
  • the prevalent animal and cellular models for human disease and drug discovery provide a poor representation of the genotypic/phenotypic spectrum extant in the patient populations to be treated. For example, strains of mice and rats commonly used in drug discovery are highly inbred, and thus only represent a very narrow range of possible genotype/phenotype combinations in mice or rats, let alone humans.
  • the relatively small number of human cell lines used for drug screening may reflect the genotypic/phenotypic scope of the individuals from which they were derived, but not that of a genetically diverse population.
  • most human cell lines are quite limited in their capacity to generate or phenocopy specific differentiated cell types (e.g., neurons, cardiomyocytes, and hepatocytes) affected by a particular health condition.
  • the cell lines are not representative of cell populations in a subject, since cell lines have been altered to indefinitely replicate.
  • animal models or genetically modified cell models of disease simply fail to adequately recapitulate the cellular disease phenotypes as they actually occur in a human patient's cells.
  • typical preclinical drug discovery strategies miss many genotype/phenotypes that are present in the human population and will have a direct impact on the therapeutic efficacy and toxicity of a candidate drug compound.
  • drug screening and drug target discovery would be performed in biological models that recapitulate the genetic and phenotypic diversity present in a human patient population and the appropriate disease state at the cellular level, well before the clinical trial stage.
  • drug discovery paradigms are illustrated schematically in FIG. 1 .
  • candidate therapeutic agents are selected for clinical trials in patients based on their action on specific drug targets and their efficacy/lack of toxicity in animal models.
  • drug discovery model the disease-relevant cells derived from patient iPSC lines, as described herein, are the starting point for identification of lead compounds based on their ability to ameliorate a disease-relevant cellular phenotype in patient derived cells.
  • the present disclosure describes human induced pluripotent stem cell lines from selected individuals (e.g., patients), genetically diverse panels of such cell lines, differentiated cells derived from such cell lines, and methods for their use in disease modeling, drug discovery, diagnostics, and individualized therapy.
  • Test drug compound refers to any test compound to be assayed for its ability to affect a functional endpoint.
  • Some examples of such functional endpoints are ligand binding to a receptor, receptor antagonism, receptor agonism, protein-protein interactions, enzymatic activities, transcriptional responses, etc.
  • “Correcting” a phenotype refers to altering a phenotype such that it more closely approximates a normal phenotype.
  • iPSC donor refers to a subject, e.g., a human patient from which one or more induced stem cell lines have been generated. Generally, the genome of an iPSC line corresponds to that of its iPSC donor.
  • Phenotypes e.g., resting calcium level, gene expression profiles, apoptotic index, electrophysiological properties, sensitivity to free radicals, compound uptake and extrusion, kinase activity, second messenger pathway responses
  • a particular type of cell e.g., cardiomyocytes
  • Phenome refers to the set of phenotypes that is subject and cell-type specific. For example, the phenome of hepatocytes and cardiomyocytes from the same individual will be quite distinct even though they share the same genome.
  • an “endogenous allele,” as used herein, refers to a naturally occurring allele that is native to the genome of a cell, i.e., an allele that is not introduced by recombinant methodologies.
  • iPSC-derived cell refers to a cell that is generated from an iPSC either by proliferation of the iPSC to generate more iPSCs, or by differentiation of the iPSC into a different cell type.
  • iPSC-derived cells include cells not differentiated directly from an iPSC, but from an intermediary cell type, e.g., a glial progenitor cell, a neural stem cell, or a cardiac progenitor cell.
  • a “normal” phenotype refers to a phenotype (e.g., apoptotic rate, resting calcium level, kinase activity, gene expression level) that falls within a range of phenotypes found in healthy individuals or that are not associated with (e.g., predictive of) a health condition.
  • a phenotype e.g., apoptotic rate, resting calcium level, kinase activity, gene expression level
  • the present disclosure provides human induced pluripotent stem cell (iPSC) lines, panels of stem cell lines, and methods for their use in drug discovery, diagnostic, and therapeutic methods as described in detail below.
  • the induced pluripotent stem cell lines disclosed herein are characterized by long term self renewal, a normal karyotype, and the developmental potential to differentiate into a wide variety of cell types (e.g., neurons, cardiomyocytes, and hepatocytes).
  • Induced pluripotent stem cell lines can be differentiated into cell lineages of all three germ layers, i.e., ectoderm, mesoderm, and endoderm.
  • a subject e.g., a patient
  • iPSC lines generated from that subject.
  • all of the genotypes of iPSC lines and those of the corresponding subject are identical.
  • genotype-phenotype correlations, uncovered in one are informative for the other, and vice versa.
  • differentiated cells e.g., neurons
  • a phenome a complete set of cellular phenotypes
  • iPSC lines provide a renewable source of differentiated cells (e.g., inaccessible differentiated cells) in which pathological cellular phenotypes that are associated with a disease, cell type, and individual may be examined and screened against test compounds.
  • iPSC lines and iPSC-derived cells are also useful for predicting the efficacy and/or adverse side effects of a candidate drug compound in specific individuals or groups of individuals, as schematically illustrated in FIG. 3 .
  • test compounds can be tested for toxicity in hepatocytes differentiated from a genetically diverse panel of induced pluripotent stem cells. Toxicity testing in iPSC-derived hepatocytes can reveal both the overall likelihood of toxicity of a test compound in a target patient population, and the likelihood of toxicity in specific patients within that population.
  • iPSC lines and iPSC-derived cells can serve as “cellular avatars,” that reveal cellular phenotypes that are disease, cell-type, and subject-specific to the extent the phenotypes are determined or predisposed by the genome.
  • panels of patient induced stem cell lines will represent a wide range of genotype/phenotype combinations in a patient population. Thus, they are useful for developing therapeutics that are effective and safe across a wide range of the relevant target population, or for determining which individuals can be treated effectively and safely with a given therapeutic agent.
  • Some of the methods described herein utilize induced stem cell lines or panels of induced stem cell lines derived from subjects that meet one or more pre-determined criteria. In some cases subjects and cellular samples from such subjects may be selected for the generation of induced stem cell lines and panels of induced stem cell lines based on one or more of such pre-determined criteria.
  • a health condition in a subject (e.g., spinal muscular atrophy, Parkinson's disease, or amyotrophic lateral sclerosis), one or more positive diagnostic criteria for a health condition, a family medical history indicating a predisposition or recurrence of a health condition, the presence or absence of a genotype associated with a health condition, or the presence of at least one polymorphic allele that is not already represented in a panel of induced stem cell lines.
  • a health condition in a subject e.g., spinal muscular atrophy, Parkinson's disease, or amyotrophic lateral sclerosis
  • a family medical history indicating a predisposition or recurrence of a health condition
  • the presence or absence of a genotype associated with a health condition e.g., the presence or absence of a genotype associated with a health condition
  • the presence of at least one polymorphic allele that is not already represented in a panel of induced stem cell lines.
  • a panel of induced stem cell lines is generated specifically from individuals diagnosed with a health condition, and from subjects that are free of the health condition.
  • health conditions include, without limitation, neurodegenerative disorders; neurological disorders such as cognitive impairment, and mood disorders; auditory disease such as deafness; osteoporosis; cardiovascular diseases; diabetes; metabolic disorders; respiratory diseases; drug sensitivity conditions; eye diseases such as macular degeneration; immunological disorders; hematological diseases; kidney diseases; proliferative disorders; genetic disorders, traumatic injury, stroke, organ failure, or loss of limb.
  • neurodegenerative disorders include, but are not limited to, Alexander's disease, Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis, ataxia telangiectasia, Batten disease, bovine spongiform encephalopathy, Canavan disease, Cockayne syndrome, corticobasal degeneration, Creutzfeldt-Jakob disease, Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, lewy body dementia, Machado-Joseph disease, multiple sclerosis, multiple system atrophy, narcolepsy, neuroborreliosis, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, primary lateral sclerosis, prion diseases, Refsum's disease, Sandhoffs disease, Schilder's disease, subacute combined degeneration of spinal cord secondary to pernicious anaemia, schizophrenia, spinocerebellar ataxia, spinal muscular atrophy, Steele-R
  • neurological disorders include, stroke, cognitive impairment, and mood disorders.
  • immunological disorders include but are not limited to acquired immune deficiency, leukemia, lymphoma, hypersensitivities (allergy), autoimmune diseases, and severe combined immune deficiency.
  • autoimmune diseases include but are not limited to acute disseminated encephalomyelitis, addison's disease, ankylosing spondylitis, antiphospholipid antibody syndrome, autoimmune hemolytic anemia, autoimmune hepatitis, bullous pemphigoid, coeliac disease, dermatomyositis, diabetes mellitus type 1, Goodpasture's syndrome, Graves' disease, Guillain-Barré syndrome, Hashimoto's disease, idiopathic thrombocytopenic purpura, lupus erythematosus, multiple sclerosis, myasthenia gravis, pemphigus, pernicious anaemia, polymyositis, primary biliary cirrhosis, rheumatoid arthritis, Sjögren's syndrome, temporal arthritis (also known as “giant cell arthritis”), vasculitis, Wegener's granulomatosis.
  • acute disseminated encephalomyelitis
  • cardiovascular diseases include but are not limited to aneurysm, angina, arrhythmia, atherosclerosis, cardiomyopathy, cerebrovascular accident (stroke), cerebrovascular disease, congenital heart disease, congestive heart failure, myocarditis, valve disease coronary, artery disease dilated, cardiomyopathy, diastolic dysfunction, endocarditis, high blood pressure (hypertension), hypertrophic cardiomyopathy, mitral valve prolapse, myocardial infarction (heart attack), and venous thromboembolism.
  • stroke cerebrovascular accident
  • congenital heart disease congestive heart failure
  • myocarditis valve disease coronary, artery disease dilated
  • cardiomyopathy diastolic dysfunction
  • endocarditis high blood pressure (hypertension)
  • hypertrophic cardiomyopathy mitral valve prolapse
  • myocardial infarction heart attack
  • venous thromboembolism examples include but are not limited to aneurysm, angina, arrhythmia
  • metabolic disorders include but are not limited to acid lipase disease, amyloidosis, Barth Syndrome, biotinidase deficiency, carnitine palmitoyl transferase deficiency type II, central pontine myelinolysis, metabolic diseases of muscle including muscular dystrophy, Farber's Disease, glucose-6-phosphate dehydrogenase deficiency, gangliosidoses, trimethylaminuria, Lesch-Nyhan syndrome, lipid storage diseases, metabolic myopathies, methylmalonic aciduria, mitochondrial myopathies, mucopolysaccharidoses, mucolipidoses, mucolipidoses, mucopolysaccharidoses, multiple CoA carboxylase deficiency, nonketotic hyperglycinemia, Pompe disease, propionic acidemia, type I glycogen storage disease, urea cycle disorders, hyperoxaluria, and oxalosis.
  • proliferative disorders include but are not limited to one or more of the following: carcinomas, sarcomas, lymphomas, leukemias, germ cell tumors, blastic tumors, prostate cancer, lung cancer, colorectal cancer, bladder cancer, cutaneous melanoma, breast cancer, endometrial cancer, and ovarian cancer.
  • Such subjects may be identified in, e.g., gene association studies, clinical studies, and hospitals, preferably after a final diagnosis of a health condition has been made.
  • subjects are identified in gene association studies that include non-affected control individuals.
  • iPSC lines are generated from subjects screened for the presence or absence of at least one allele associated with a health condition or a predisposition for a health condition.
  • Such alleles indicate that an individual, though not exhibiting overt symptoms of a health condition, has a high risk of developing the health condition.
  • BRCA1 have been used to indicate a high likelihood of developing breast cancer. Genotyping of subjects may be performed on samples from a number of sources, e.g., blood banks, sperm banks, gene-association studies, hospitals, clinical trials, or any other source as long as a living cellular sample can be obtained from the individual that is genotyped.
  • cellular phenotypes from individuals carrying alleles associated with health conditions will exhibit abnormalities that can serve as more reliable prognostic indicators of a health condition in combination with a genotype than a genotype alone. Further, identification of specific abnormal cellular phenotypes associated with a health condition may indicate a target pathway for screening of prophylactic and therapeutic agents for the health condition.
  • polymorphic alleles present in the human population, e.g., single polymorphisms (SNPs) and the occurrence of common health conditions, e.g., neurodegenerative diseases, psychiatric disorders, metabolic disorders, and cardiovascular diseases.
  • SNPs single polymorphisms
  • common health conditions e.g., neurodegenerative diseases, psychiatric disorders, metabolic disorders, and cardiovascular diseases.
  • Various types of polymorphic alleles can be found in the human genome as summarized in Table 1.
  • VNTR Microsatellites also termed short tandem repeat >500,000 tandem recaps (STR) polymorphisms are typically tandem repeats of two, three or four nucleotides, but repeats up to ten nucleotides in length may also classified in this group.
  • Minisatellites are VNTR polymorphisms in which 10-100 nucleotides are repeated in variable numbers. Repeated segments often do not have exactly identical sequences.
  • VNTRs with larger repeat units (100-1000 bp) are termed satellites. Copy number CNV Inheritable deletion of multiplication of DNA >1500 loci variation segments larger than 1 kb.
  • HuGE Navigator hugenavigator.net HuGE Navigator provides access to a continuously updated knowledge base in human genome epidemiology, including information on population prevalence of genetic variants, gene-disease associations, gene-gene and gene-environment interactions, and evaluation of genetic tests.
  • GenAtlas www.genatlas.org Regularly updated database of genes, phenotypes and references. Among numerous databases are brief sections on disorders associated with genes, with lists of citations. May be biased towards statistically significant results.
  • OMIM www.ncbi.nlm.nih.gov/omim/ Database of human genes and genetic disorders, containing textual information with links to Medline and sequence records in the Entrez system, and links to additional related resources at NCBI and elsewhere.
  • PharmGKB www.pharmgkb.org Database of genomic data and clinical information from participants in pharmacogenetics research studies. Welcomes submission of primary data. T1DBase t1dbase.org/ Database of type 1 diabetes data, including information from collaborating laboratories. Some indication given of unpublished data.
  • Subjects may be screened for alleles in genes that affect response to a therapeutic agent or to a class of therapeutic agents.
  • alleles include, but are not limited to, alleles of drug metabolizing enzymes, such as Glucose 6 phosphate dehydrogenase (G6PDH), Butyrlcholine esterase, N-acetyltransferase, Cytochrome P450 isoforms (e.g., 2B6, 2D6, C19, 2C9), Thiopurine S-methyltransferase, Dihydropyrimidine dehydrogenase, and Uridin diphospho-glucuronic acid transferase type 1A1.
  • G6PDH Glucose 6 phosphate dehydrogenase
  • Butyrlcholine esterase N-acetyltransferase
  • Cytochrome P450 isoforms e.g., 2B6, 2D6, C19, 2C9
  • Alleles of Cytochrome P450 enzyme isoforms can be found, e.g., in a database provided under the “Home Page of the Human Cytochrome P450 (CYP) Allele Nomenclature Committee,” at “www.cypalleles.ki.se/.”
  • Some alleles occur in genes that affect drug transport, including, e.g., multiple drug resistance conferring transporters (MDRs), breast cancer resistance protein (BRCP), multidrug resistance-associated-associated proteins (MRPs), and organic anion-transporting polypeptide (OATP1B1).
  • MDRs multiple drug resistance conferring transporters
  • BRCP breast cancer resistance protein
  • MRPs multidrug resistance-associated-associated proteins
  • Other alleles occur in genes that encode drug targets, including, but not limited to, Vitamin K epoxide reductase, Factor V, G-protein coupled receptors (GPCRs).
  • GPCRs are one of the most common drug targets.
  • polymorphic alleles in GPCRs can be found in, e.g., the GPCR Natural Variants (“NaVa”) Database, which is accessible on the internet at “nava.liacs.nl/”
  • GPCR NaVa GPCR Natural Variants
  • the GPCR NaVa database describes sequence variants within the family of human G Protein-Coupled Receptors (GPCRs). It includes: rare mutations (frequency ⁇ 1%); polymorphisms (frequency >1%), including Single Nucleotide Polymorphisms (SNPs); variants without estimates of allele frequency.
  • Polymorphic alleles of interest may be detected and scored in a nucleic acid sample from a subject by any of a number of methods known in the art. For example, detection of multiple alleles may be performed by conducting a nucleic acid array-based assay on a nucleic acid sample from a subject, where the nucleic acid array comprises allele-specific probes (e.g., SNP-specific probes), which, under high stringency hybridization conditions, selectively hybridize with and discriminate between the nucleic acid sequences of two or more polymorphic alleles of interest, e.g., alleles of G-protein coupled receptors.
  • allele-specific probes e.g., SNP-specific probes
  • the nucleic acid arrays used to detect polymorphisms may be commercially available nucleic acid arrays.
  • the Affymetrix® Genome-Wide SNP Array 6.0 includes probes for more than 906,000 SNPs and more than 946,000 probes for the detection of copy number variation.
  • the nucleic acid arrays may be custom-made to include to a limited subset of alleles of interest.
  • the design of suitable probe arrays for analysis of predetermined polymorphisms and interpretation of the hybridization patterns is described in detail in WO 95/11995; EP 717,113; and WO 97/29212.
  • Such arrays typically contain first and second groups of probes which are designed to be complementary to different allelic forms of the polymorphism.
  • Each group contains a first set of probes, which is subdivided into subsets, one subset for each polymorphism.
  • Each subset contains probes that span a polymorphism and proximate bases and are complementary to one allelic form of the polymorphism.
  • the hybridization patterns of these probes to target samples can be analyzed by footprinting or cluster analysis, as described above.
  • first and second probes groups contain subsets of probes respectively complementarity to first and second allelic forms of a polymorphic site spanned by the probes
  • first and second probes groups contain subsets of probes respectively complementarity to first and second allelic forms of a polymorphic site spanned by the probes
  • on hybridization of the array to a sample that is homozygous for the first allelic form all probes in the subset from the first group show specific hybridization, whereas probes in the subset from the second group that span the polymorphism show only mismatch hybridization.
  • the mismatch hybridization is manifested as a footprint of probe intensities in a plot of normalized probe intensity (i.e., target/reference intensity ratio) for the subset of probes in the second group.
  • a footprint is observed in the normalized hybridization intensities of probes in the subset from the first probe group. If the target sample is heterozygous for both allelic forms then a footprint is seen in normalized probe intensities from subsets in both probe groups although the depression of intensity ratio within the footprint is less marked than in footprints observed with homozygous alleles.
  • Analysis of the hybridization pattern of a nucleic acid array to a nucleic acid sample indicates which allelic form is present at some or all of the SNP sequences represented on the array.
  • an individual or an iPSC line generated from an individual can be characterized with a polymorphic profile representing allelic variants of interest, e.g., alleles associated with a health condition.
  • an allele is detected using a primer extension reaction or amplification reaction.
  • a nucleic acid sample containing (or suspected of containing) a target nucleic acid molecule can be contacted with an oligonucleotide primer that, upon further contact with a polymerase, can be extended up to and, if desired, beyond the position of the SNP.
  • the nucleic acid sample can be contacted with an amplification primer pair, comprising a first primer and a second primer, which selectively hybridize to complementary strands of a target nucleic acid molecule and, in the presence of polymerase, allow for generation of an amplification product.
  • primers of an amplification primer pair are referred to as a “first primer” and a “second primer”; however, reference herein to a “first primer” or a “second primer” is not intended to indicate any importance, order of addition, or the like. It will be further recognized that an amplification primer pair requires that the first and second primer comprise what are commonly referred to as a forward primer and a reverse primer.
  • a primer extension or PCR amplification reaction can be designed such that the presence of a particular nucleotide at an SNP position can be determined by the presence or size of the extension and/or amplification product, in which case the SNP can be determined using a method such as gel electrophoresis, capillary gel electrophoresis, or mass spectrometry; or the amplification product can be sequenced to determine the nucleotide at the SNP position.
  • the SNP can be detected indirectly, for example, by further contacting the sample with a detector oligonucleotide, which can selectively hybridize to a nucleotide sequence of the first amplification product comprising the SNP position; and detecting selective hybridization of the detector oligonucleotide, as above.
  • a detector oligonucleotide which can selectively hybridize to a nucleotide sequence of the first amplification product comprising the SNP position; and detecting selective hybridization of the detector oligonucleotide, as above.
  • PCR can be performed using TaqMan® reagents, followed by reading the plates at this endpoint.
  • Amplification products also can be detected using an ELISA format, for example, using a design in which one primer is biotinylated and the other contains digoxygenin. The amplification products are then bound to a streptavidin plate, washed, reacted with an enzyme-conjugated antibody to digoxygenin, and developed with a chromogenic, fluorogenic, or chemiluminescent substrate for the enzyme.
  • a radioactive method can be used to detect generated amplification products, for example, by including a radiolabeled deoxynucleoside triphosphate into the amplification reaction, then blotting the amplification products onto DEAE paper for detection.
  • a radioactive method can be used to detect generated amplification products, for example, by including a radiolabeled deoxynucleoside triphosphate into the amplification reaction, then blotting the amplification products onto DEAE paper for detection.
  • one primer is biotinylated
  • streptavidin-coated scintillation proximity assay plates can be used to measure the PCR products.
  • chemiluminescent label for example, a lanthanide chelate such as used in the DELFIA® assay (Pall Corp.), an electrochemiluminescent label such as ruthenium tris-bipyridy (ORI-GEN), or a fluorescent label, for example, using fluorescence correlation spectroscopy.
  • a chemiluminescent label for example, a lanthanide chelate such as used in the DELFIA® assay (Pall Corp.), an electrochemiluminescent label such as ruthenium tris-bipyridy (ORI-GEN), or a fluorescent label, for example, using fluorescence correlation spectroscopy.
  • SNP-IT® assay system An assay system that is commercially available and can be used to identify a nucleotide occurrence of one or more SNPs is the SNP-IT® assay system (Orchid BioSciences, Inc.; Princeton N.J.).
  • the SNP-IT® method is a three step primer extension reaction. In the first step a target nucleic acid molecule is isolated from a sample by hybridization to a capture primer, which provides a first level of specificity. In a second step the capture primer is extended from a terminating nucleotide triphosphate at the target SNP site, which provides a second level of specificity.
  • the extended nucleotide triphosphate can be detected using a variety of known formats, including, for example, by direct fluorescence, indirect fluorescence, an indirect colorimetric assay, mass spectrometry, or fluorescence polarization. Reactions conveniently can be processed in 384 well format in an automated format using a SNP stream® instrument (Orchid BioSciences, Inc.).
  • Various methods for genotyping SNP alleles are readily adaptable to high throughput assays.
  • an amplification reaction such as PCR can be performed using inexpensive robotic thermocyclers for a specified number of cycles, then the amplification product generated can be determined at the endpoint of the reaction.
  • the methods can be performed in a multiplex format, for example, using differentially labeled oligonucleotide probes, or performing oligonucleotide ligation assays that result in different sized ligation products, or amplification reactions that result in different sized amplification products.
  • high-throughput mass spectrometry is used to detect SNP alleles in a target nucleic acid sample. Mass spectrometric methods for SNP genotyping are described in, e.g., U.S. Pat. Nos. 7,132,519, 6,994,998; and U.S. Patent Application No 20060275789.
  • high stringency conditions are those that result in perfect matches remaining in hybridization complexes, while imperfect matches melt off.
  • low stringency conditions are those that allow the formation of hybridization complexes with both perfect and imperfect matches.
  • High stringency conditions are known in the art; see for example Maniatis et al. (1989), Molecular Cloning: A Laboratory Manual, 2d Edition; and Short Protocols in Molecular Biology, ed. Ausubel, et al. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures.
  • Tm thermal melting point
  • Stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 3° C. for short probes (e.g. 10 to 50 nucleotides) and at least about 6° C. for long probes (e.g. greater than 50 nucleotides).
  • Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide.
  • less stringent hybridization conditions are used; for example, moderate or low stringency conditions may be used, as are known in the art. See, e.g., Maniatis and Ausubel, supra, and Tijssen, supra.
  • iPSC lines may be induced from a wide variety of mammalian cells, e.g., human somatic cells, such as fibroblasts, bone marrow-derived mononuclear cells, skeletal muscle cells, adipose cells, peripheral blood mononuclear cells, macrophages, hepatocytes, keratinocytes, oral keratinocytes, hair follicle dermal cells, gastric epithelial cells, lung epithelial cells, synovial cells, kidney cells, skin epithelial cells or osteoblasts.
  • mammalian cells e.g., human somatic cells, such as fibroblasts, bone marrow-derived mononuclear cells, skeletal muscle cells, adipose cells, peripheral blood mononuclear cells, macrophages, hepatocytes, keratinocytes, oral keratinocytes, hair follicle dermal cells, gastric epithelial cells, lung epithelial cells, syn
  • the cells to be induced can originate from many different types of tissue, e.g., bone marrow, skin (e.g., dermis, epidermis), muscle, adipose tissue, peripheral blood, foreskin, skeletal muscle, or smooth muscle.
  • tissue e.g., bone marrow, skin (e.g., dermis, epidermis), muscle, adipose tissue, peripheral blood, foreskin, skeletal muscle, or smooth muscle.
  • the cells can also be derived from neonatal tissue, including, but not limited to: umbilical cord tissues (e.g., the umbilical cord, cord blood, cord blood vessels), the amnion, the placenta, or other various neonatal tissues (e.g., bone marrow fluid, muscle, adipose tissue, peripheral blood, skin, skeletal muscle etc.).
  • the cells can be derived from neonatal or post-natal tissue collected from a mammal within the period from birth, including cesarean birth, to death.
  • the tissue may be from a mammal who is >10 minutes old, >1 hour old, >1 day old, >1 month old, >2 months old, >6 months old, >1 year old, >2 years old, >5 years old, >10 years old, >15 years old, >18 years old, >25 years old, >35 years old, >45 years old, >55 years old, >65 years old, ⁇ 80 years old, ⁇ 70 years old, ⁇ 60 years old, ⁇ 50 years old, ⁇ 40 years old, ⁇ 30 years old, ⁇ 20 years old or ⁇ 10 years old.
  • the tissue is from a human age 18, 20, 21, 23, 24, 25, 28, 29, 31, 33, 34, 35, 37, 38, 40, 41, 42, 43, 44, 47, 51, 55, 61, 63, 65, 70, 77, or 85 years old.
  • the cells may be from non-embryonic tissue, e.g., at a stage of development later than the embryonic stage. In some cases, the cells may be derived from a fetus. In some cases, the cells are not from a fetus. In some cases, the cells are from an embryo. In some cases, the cells are not from an embryo.
  • the cells can be obtained from a single cell or a population of cells.
  • the population may be homogenous or heterogeneous.
  • the cells may be a population of cells found in a human cellular sample, e.g., a biopsy or blood sample.
  • the cells are a cell line.
  • the cells are somatic cells.
  • the cells are derived from cells fused to other cells.
  • the cells are not derived from cells fused to other cells.
  • the cells are not derived from cells artificially fused to other cells.
  • the cells are not: a cell that has been fused with an embryonic stem cell, or a cell that has undergone the procedure known as somatic cell nuclear transfer.
  • the cellular population may include both differentiated and undifferentiated cells. In some cases, the population primarily contains differentiated cells. In other cases, the population primarily contains undifferentiated cells, e.g., undifferentiated stem cells. The undifferentiated cells within the population may be induced to become pluripotent or multipotent. In some cases, differentiated cells within the cellular population are induced to become pluripotent or multipotent.
  • the cellular population may include undifferentiated cells such as mesenchymal stem cells (MSCs), see, e.g., Pittenger et al. (1999), Science 284 (5411): 143-7, multipotent adult progenitor cells (MAPCs), see, e.g., Jahagirdar et al. (2005), Stem Cell Rev. 1(1): 53-9, and/or marrow-isolated adult multilineage inducible (MIAMI) cells (D'Ippolioto et al., (2004), J. Cell Sci. 117 (Pt 14): 2971-81.
  • MSCs are multipotent cells that arise from the mesenchyme during development.
  • the undifferentiated stem cells are stem cells that have not undergone epigenetic inactivating modification by heterochromatin formation due to DNA methylation or histone modification of at least four genes, at least three genes, at least two genes, at least one gene, or none of the following: Nanog, Oct3/4, Sox2 and Tert.
  • Activation, or expression of such genes may occur when human pluripotent stem cells are induced from undifferentiated stem cells present in a human postnatal tissue.
  • the methods include obtaining a cellular sample, e.g., by a biopsy, blood draw, or alveolar or other pulmonary lavage.
  • Other suitable methods for obtaining various types of human somatic cells include, but are not limited to, the following exemplary methods:
  • the donor is given a general anesthetic and placed in a prone position. From the posterior border of the ilium, a collection needle is inserted directly into the skin and through the iliac surface to the bone marrow, and liquid from the bone marrow is aspirated into a syringe. A mononuclear cell fraction is then prepared from the aspirate by density gradient centrifugation. The collected crude mononuclear cell fraction is then cultured prior to use in the methods described herein for induction pluripotency. For convenience, methods for induction of pluripotency, as described herein, are collectively referred to as “induction.”
  • Skin tissue containing the dermis is harvested, for example, from the back of a knee or buttock. The skin tissue is then incubated for 30 minutes at 37° C. in 0.6% trypsin/DMEM (Dulbecco's Modified Eagle's Medium)/F-12 with 1% antibiotics/antimycotics, with the inner side of the skin facing downward.
  • trypsin/DMEM Dulbecco's Modified Eagle's Medium
  • F-12 1% antibiotics/antimycotics
  • the skin tissue is finely cut into 1 mm2 sections using scissors, which are then centrifuged at 1200 rpm and room temperature for 10 minutes. The supernatant is removed, and to the tissue precipitate is added 25 ml of 0.1% trypsin/DMEM/F-12/1% antibiotics, antimycotics, and stirred using a stirrer at 37° C. and 200-300 rpm for 40 minutes.
  • FBS fetal bovine serum
  • JRH 3 ml fetal bovine serum
  • gauze Type I manufactured by PIP
  • a 100 ⁇ m nylon filter manufactured by FALCON
  • a 40 ⁇ m nylon filter manufactured by FALCON
  • DMEM/F-12/1% antibiotics, antimycotics is added to wash the precipitate, and then centrifuged at 1200 rpm and room temperature for 10 minutes. The cell fraction thus obtained is then cultured prior to induction.
  • a connective tissue containing muscle such as the lateral head of the biceps brachii muscle or the sartorius muscle of the leg is cut and the muscle tissue is excised, it is sutured.
  • the whole muscle obtained is minced with scissors or a scalpel, and then suspended in DMEM (high glucose) containing 0.06% collagenase type IA and 10% FBS, and incubated at 37° C. for 2 hours.
  • DMEM high glucose
  • the cell fraction obtained may be cultured according to the method described in 6. below as crude purified cells containing undifferentiated stem cells, and used for the induction of human pluripotent stem cells of the present invention.
  • adipose tissue for use in the present invention may be isolated by various methods known to a person skilled in the art. For example, such a method is described in U.S. Pat. No. 6,153,432, which is incorporated herein in its entirety.
  • a preferred source of adipose tissue is omental adipose tissue. In humans, adipose cells are typically isolated by fat aspiration.
  • adipose tissue is treated with 0.01% to 0.5%, preferably 0.04% to 0.2%, and most preferably about 0.1% collagenase, 0.01% to 0.5%, preferably 0.04%, and most preferably about 0.2% trypsin and/or 0.5 ng/ml to 10 ng/ml dispase, or an effective amount of hyaluronidase or DNase (DNA digesting enzyme), and about 0.01 to about 2.0 mM, preferably about 0.1 to about 1.0 mM, most preferably 0.53 mM concentration of ethylenediaminetetraacetic acid (EDTA) at 25 to 50° C., preferably 33 to 40° C., and most preferably 37° C. for 10 minutes to 3 hours, preferably 30 minutes to 1 hour, and most preferably 45 minutes.
  • EDTA ethylenediaminetetraacetic acid
  • Cells are passed through nylon or a cheese cloth mesh filter of 20 microns to 800 microns, more preferably 40 microns to 400 microns, and most preferably 70 microns. Then the cells in the culture medium are subjected to differential centrifugation directly or using Ficoll or Percoll or another particle gradient. The cells are centrifuged at 100 to 3000 ⁇ g, more preferably 200 to 1500 ⁇ g, most preferably 500 ⁇ g for 1 minute to 1 hours, more preferably 2 to 15 minutes and most preferably 5 minutes, at 4 to 50° C., preferably 20 to 40° C. and more preferably about 25° C.
  • the adipose tissue-derived cell fraction thus obtained may be cultured according to the method described herein as crude purified cells containing undifferentiated stem cells, and used for the induction of human pluripotent or multipotent stem cells.
  • a mononuclear cell fraction is obtained by the Ficoll-Hypaque method, as described in, e.g., Kanof et al. (1993), Current Protocols in Immunology (J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevack, and W. Strober, eds.), ch. 7.1.1.-7.1.5, John Wiley & Sons, New York).
  • a RPMI 1640 medium containing 10% fetal bovine serum, 100 ⁇ g/ml streptomycin and 100 units/ml penicillin, and after washing twice, the cells are recovered.
  • the recovered cells are resuspended in RPMI 1640 medium and then plated in a 100 mm plastic petri dish at a density of about 1 ⁇ 10 7 cells/dish, and incubated in a 37° C. incubator at 8% CO 2 . After 10 minutes, cells remaining in suspension are removed and adherent cells are harvested by pipetting.
  • the resulting adherent mononuclear cell fraction is then cultured prior to the induction period as described herein.
  • the peripheral blood-derived or cord blood-derived adherent cell fraction thus obtained may be cultured according to the method described herein as crude purified cells containing undifferentiated stem cells, and used for the induction of human pluripotent stem cells of the present invention.
  • forced expression of certain polypeptides is carried out in cultured cells for a period of time, after which the iPSCs are screened for a number of morphological and gene expression properties that characterize multipotent and pluripotent stem cells. Induced cells that meet these screening criteria may then be subcloned and expanded.
  • the cells to be induced may be cultured for a period of time prior to the induction procedure. Alternatively, the cells to be induced may be used directly in the induction process without a prior culture period.
  • the type of cell culture medium used is the same or very similar before, during, and after the induction process. In other cases, different cell culture media are used at different points. For example, one type of culture medium may be used directly before the induction process, while a second type of media is used during the induction process. At times, a third type of culture medium is used during the induction process.
  • Cells may be cultured in medium supplemented with a particular serum.
  • the serum is fetal bovine serum (FBS).
  • the serum can also be fetal calf serum (FCS).
  • the serum may be Human AB serum. Mixtures of serum may also be used, e.g. mixture of FBS and Human AB, FBS and FCS, or FCS and Human AB.
  • a “low serum culture condition” refers to the use of a cell culture medium containing a concentration of serum ranging from 0% (v/v) (i.e., serum-free) to about 5% (v/v), e.g., 0% to 2%, 0% to 2.5%, 0% to 3%, 0% to 4%, 0% to 5%, 0.1% to 2%, 0.1% to 5%, 0.1%, 0.5%, 1%, 1.2%, 1.5%, 2%, 2.5%, 3%, 3.5%, or 4%.
  • the serum concentration is from about 0% to about 2%. In some cases, the serum concentration is about 2%.
  • the serum concentration is preferably 2% or less.
  • cells are cultured under a “high serum condition,” i.e., greater than 5% serum to about 20% serum, e.g., 6%, 7%, 8%, 10%, 12%, 15%, or 20%. Culturing under high serum conditions may occur prior to, during, and/or after induction.
  • Some representative media that the cells can be cultured in include: MAPC, FBM, ES, MEF-conditioned ES (MC-ES), and mTeSRTM (available, e.g., from StemCell Technologies, Vancouver, Canada), See Ludwig et al (2006), Nat Biotechnol, 24(2):185-187.
  • alternative culture conditions for growth of human ES cells are used, as described in, e.g., Skottman et al (2006), Reproduction, 132(5):691-698.
  • the cells are cultured in MAPC, FBM, MC-ES, or mTeSRTM prior to and/or during the introduction of induction factors to the cells; and the cells are cultured in MC-ES or mTeSRTM medium later in the induction process.
  • MAPC (2% FBS) Medium may comprise: 60% Dulbecco's Modified Eagle's Medium-low glucose, 40% MCDB 201, Insulin Transferrin Selenium supplement, (0.01 mg/ml insulin; 0.0055 mg/ml transferrin; 0.005 ⁇ g/ml sodium selenite), 1 ⁇ linolenic acid albumin (1 mg/mL albumin; 2 moles linoneic acid/mole albumin), 1 nM dexamethasone, 2% fetal bovine serum, 1 nM dexamethasone, 10 ⁇ 4 M ascorbic acid, and 10 ⁇ g/ml gentamycin.
  • FBM (2% FBS) Medium may comprise: MCDB202 modified medium, 2% fetal bovine serum, 5 ⁇ g/ml insulin, 50 mg/ml gentamycin, and 50 ng/ml amphotericin-B.
  • ES Medium may comprise: 40% Dulbecco's Modified Eagle's Medium (DMEM) 40% F12 medium, 2 mM L-glutamine, 1 ⁇ non-essential amino acids (Sigma, Inc., St. Louis, Mo.), 20% Knockout Serum ReplacementTM (Invitrogen, Inc., Carlsbad, Calif.), and 10 ⁇ g/ml gentamycin.
  • DMEM Dulbecco's Modified Eagle's Medium
  • F12 medium 2 mM L-glutamine
  • 1 ⁇ non-essential amino acids Sigma, Inc., St. Louis, Mo.
  • Knockout Serum ReplacementTM Invitrogen, Inc., Carlsbad, Calif.
  • 10 ⁇ g/ml gentamycin 10 ⁇ g/ml gentamycin.
  • MC-ES medium may be prepared as follows.
  • ES medium is conditioned on mitomycin C-treated murine embryonic fibroblasts (MEFs), harvested, filtered through a 0.45- ⁇ M filter, and supplemented with about 0.1 mM ⁇ mercaptoethanol, about 10 ng/ml bFGF or FGF-2, and, optionally, about 10 ng/ml activin A.
  • MEFs murine embryonic fibroblasts
  • irradiated MEFs are used in place of the mitomycin C-treated MEFs.
  • one or more growth factors such as fibroblast growth factor (FGF)-2; basic FGF (bFGF); platelet-derived growth factor (PDGF), epidermal growth factor (EGF); insulin-like growth factor (IGF); or insulin can be included in the culture medium.
  • FGF fibroblast growth factor
  • bFGF basic FGF
  • PDGF platelet-derived growth factor
  • EGF epidermal growth factor
  • IGF insulin-like growth factor
  • Other growth factors that can be used to supplement cell culture media include, but are not limited to one or more: Transforming Growth Factor ⁇ -1 (TGF ⁇ -1), Activin A, Noggin, Brain-derived Neurotrophic Factor (BDNF), Nerve Growth Factor (NGF), Neurotrophin (NT)-1, NT-2, or NT 3. In some cases, one or more of such factors is used in place of the bFGF or FGF-2 in the MC-ES medium or other cell culture medium.
  • the concentration of growth factors in the culture media described herein is from about 2 ng/ml to about 20 ng/ml, e.g., about 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 10 ng/ml, 12 ng/ml, 14 ng/ml, 15 ng/ml, 17 ng/ml, or 20 ng/ml.
  • the concentration of growth factors in the culture media described herein is from about 2 ng/ml to about 20 ng/ml, e.g., about 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 10 ng/ml, 12 ng/ml, 14 ng/ml, 15 ng/ml, 17 ng/ml, or 20
  • the concentration of bFGF or FGF2 is from about 2 ng/ml to about 5 ng/ml; from about 5 ng/ml to about 8 ng/ml; from about 9 ng/ml to about 11 ng/ml; from about 11 ng/ml to about 15 ng/ml; or from about 15 ng/ml to about 20 ng/ml.
  • the growth factors may be used alone or in combination.
  • FGF-2 may be added alone to the medium; in another example, both PDGF and EGF are added to the culture medium.
  • the “iPSCs” are maintained in MC-ES medium as described herein.
  • cells are maintained in the presence of a rho, or rho-associated, protein kinase (ROCK) inhibitor to reduce apoptosis.
  • a rho, or rho-associated, protein kinase (ROCK) inhibitor to reduce apoptosis.
  • an inhibitor of Rho associated kinase is added to the culture medium.
  • the addition of Y-27632 (Calbiochem; water soluble) or Fasudil (HA1077: Calbiochem) an inhibitor of Rho associated kinase (Rho associated coiled coil-containing protein kinase) may be used to culture the human pluripotent stem cells of the present invention.
  • the concentration of Y-27632 or Fasudil is from about 5 ⁇ M to about 20 e.g., about 5 ⁇ M, 10 ⁇ M, 15 ⁇ M, or 20 ⁇ M.
  • the cells may be cultured for about 1 to about 12 days e.g., 2 days, 3 days, 4.5 days, 5 days, 6.5 days, 7 days, 8 days, 9 days, 10 days, or any other number of days from about 1 day to about 12 days prior to undergoing the induction methods described herein.
  • the iPSCs are cultured in complete ES medium in a 37° C., 5% CO 2 incubator, with medium changes about every 1 to 2 days.
  • induced the iPSCs are cultured and observed for about 14 days to about 40 days, e.g., 15, 16, 17, 18, 19, 20, 23, 24, 27, 28, 29, 30, 31, 33, 34, 35, 36, 37, 38 days, or any other period from about 14 days to about 40 days prior to identifying and selecting clones comprising “iPSCs” based on morphological characteristics.
  • Morphological characteristics for identifying iPSC clones include, but are not limited to, a small cell size with a high nucleus-to-cytoplasm ratio; formation of small monolayer colonies within the space between parental cells (e.g., between fibroblasts).
  • the cells may be plated at a cell density of about 1 ⁇ 10 3 cells/cm 2 to about 1 ⁇ 10 4 cells/cm 2 , e.g., 2 ⁇ 10 3 cells/cm 2 , 3.5 ⁇ 10 3 cells/cm 2 , 6 ⁇ 10 3 cells/cm 2 , 7 ⁇ 10 3 cells/cm 2 , 9 ⁇ 10 3 cells/cm 2 , or any other cell density from about 1 ⁇ 10 3 cells/cm 2 to about 1 ⁇ 10 4 cells/cm 2 .
  • the cells can be plated and cultured directly on tissue culture-grade plastic.
  • cells are plated and cultured on a coated substrate, e.g., a substrate coated with fibronectin, gelatin, matrigelTM, collagen, or laminin.
  • Suitable cell culture vessels include, e.g., 35 mm, 60 mm, 100 mm, and 150 mm cell culture dishes, 6-well cell culture plates, and other size-equivalent cell culture vessels.
  • the cells are cultured with feeder cells.
  • the cells may be cultured on a layer, or carpet, of MEFs.
  • Media with low concentrations of serum may be particularly useful to enrich for undifferentiated stem cells.
  • the undifferentiated cells cultured under low serum conditions may or may not share certain properties with MSCs, MAPCs, and/or MIAMI cells. Differences in phenotype may be due, in part, to culture methods used to obtain MSCs, MAPCs and MIAMI cells.
  • MSCs are often obtained by isolating the non-hematopoeitic cells (e.g., interstitial cells) adhering to a plastic culture dish when tissue, e.g., bone marrow, fat, muscle, or skin etc., is cultured in a culture medium containing a high-concentration serum (5% or more).
  • tissue e.g., bone marrow, fat, muscle, or skin etc.
  • a high-concentration serum 5% or more
  • the cells in order to culture and grow human pluripotent stem cells induced from the undifferentiated stem cells of the present invention present in a human postnatal tissue, it is preferred that the cells are subcultured every 5 to 7 days in a culture medium containing the additives described herein on a MEF-covered plastic culture dish or a matrigel-coated plastic culture dish.
  • the cells may be cultured at a low density, which may be accomplished by splitting the cells from about 1:6 to 1:3 or by plating the cells at 10 3 cells/cm 2 to 3 ⁇ 10 4 cells/cm 2 .
  • Primary culture ordinarily occurs immediately after the cells are isolated from a donor, e.g., human.
  • the primary cells can be subjected to a second subculture, a third subculture, a fourth subculture, and greater than four subcultures.
  • a “second” subculture describes primary culture cells subcultured once
  • a “third” subculture describes primary cultures subcultured twice
  • a “fourth” subculture describes primary cells subcultured three times, etc.
  • the culture techniques described herein may generally include culturing from the period between the primary culture and the fourth subculture, but other culture periods may also be employed.
  • cells are cultured from primary culture to second subculture.
  • the methods for induction of pluripotency in one or more cells include forcing expression of a set of induction factors (Ws).
  • the set of IFs includes one or more: an Oct3/4 polypeptide, a Sox2 polypeptide, a Klf4 polypeptide, or a c-Myc polypeptide.
  • the set does not include a c-Myc polypeptide.
  • the set of IFs can include: an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide, but not a c-Myc polypeptide.
  • the set of IFs does not include polypeptides that might increase the risk of cell transformation.
  • the set may include a c-Myc polypeptide.
  • the c-Myc polypeptide is a constitutively active variant of c-Myc.
  • the set includes a c-Myc polypeptide capable of inducible activity, e.g., a c-Myc-ER polypeptide, see, e.g., Littlewood, et al. (1995) Nucleic Acid Res. 23(10):1686-90.
  • the set of IFs may include: an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide, but not a TERT polypeptide, a SV40 Large T antigen polypeptide, HPV16 E6 polypeptide, a HPV16 E7 polypeptide, or a Bmi1 polypeptide.
  • the set of IFs does not include a TERT polypeptide.
  • the set of IFs does not include a SV40 Large T antigen.
  • the set of IFS does not include a HPV16 E6 polypeptide or a HPV16 E7 polypeptide.
  • the set of Ws includes three Ws, wherein two of the three IFs are an Oct3/4 polypeptide and a Sox2 polypeptide.
  • the set of IFs includes two Ws, wherein the two polypeptides are a c-Myc polypeptide and a Sox2 polypeptide
  • the set of induction factors is limited to Oct 3/4, Sox2, and Klf4 polypeptides.
  • the set of induction factors may be limited to a set of four Ws: an Oct3/4 polypeptide, a Sox2 polypeptide, a Klf4 polypeptide, and a c-Myc polypeptide.
  • a set of Ws may include Ws in addition to an Oct 3/4, a Sox2, and a Klf4 polypeptide.
  • additional Ws include, but are not limited to Nanog, TERT, LIN28, CYP26A1, GDF3, FoxD3, Zfp42, Dnmt3b, Ecat1, and Tel1 polypeptides.
  • the set of additional Ws does not include a c Myc polypeptide.
  • the set of additional IFs does not include polypeptides that might increase the risk of cell transformation.
  • Forced expression of Ws may be maintained for a period of at least about 7 days to at least about 40 days, e.g., 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 25 days, 30 days, 33 days, or 37 days.
  • the efficiency of inducing pluripotency in cells of a human population of cells is from at least about 0.001% to at least about 0.01% of the total number of cells to be induced, e.g., 0.002%, 0.0034%, 0.004%, 0.005%, 0.0065%, 0.007%, 0.008%, or 0.0085%.
  • Induction of the cells may be accomplished by combining histone deacetylase (HDAC) inhibitor treatment with forced expression of sets of Ws.
  • HDAC histone deacetylase
  • the cells to be induced may be undifferentiated stem cells present in a human postnatal tissue. In other cases, the cells to be induced are differentiated cells or are a mixture of differentiated or undifferentiated cells.
  • the HDAC may be combined with the forced expression of a specific set of Ws, e.g., Oct 3/4, a Sox2, and a Klf4.
  • a human somatic cell is induced to become pluripotent after HDAC inhibitor treatment is combined with forced expression of Oct3/4, Sox2 and Klf4 or forced expression of Oct3/4, Sox2, Klf4, and c-Myc.
  • human pluripotent stem cells can be induced by introducing three genes of Oct3/4, Sox2 and Klf4 or three genes of Oct3/4, Sox2 and Klf4 plus the c-Myc gene or a HDAC inhibitor into undifferentiated stem cells present in a human postnatal tissue in which each gene of Tert, Nanog, Oct3/4 and Sox2 has not undergone epigenetic inactivation.
  • human pluripotent stem cells are induced by introducing three genes of Oct3/4, Sox2 and Klf4 or three genes of Oct3/4, Sox2 and Klf4 plus the c-Myc gene or a histone deacetylase inhibitor into undifferentiated stem cells after the undifferentiated stem cells were amplified by a primary culture or a second subculture, or a subculture in a low density and subculturing in a culture medium comprising a low-concentration serum.
  • Cells may be treated with one or more HDACs for about 2 hours to about 5 days, e.g., 3 hours, 6 hours, 12 hours, 14 hours, 18 hours, 1 day, 2 days, 3 days, or 4 days.
  • Treatment with HDAC inhibitor may be initiated prior to beginning forced expression of IFs in the cells. In some cases, HDAC inhibitor treatment begins during or after forced expression of IFs in the cells. In other cases, HDAC inhibitor treatment begins prior to forced expression and is maintained during forced expression.
  • Suitable concentrations of an HDAC inhibitor range from about 0.001 nM to about 10 mM, depending on the particular HDAC inhibitor to be used, but are selected so as to not significantly decrease cell survival in the treated cells.
  • the HDAC concentration may range from 0.01 nM, to 1000 nM.
  • the HDAC concentration ranges from about 0.01 nM to about 1000 nM, e.g., about 0.05 nM, 0.1 nM, 0.5 nM, 0.75 nM, 1.0 nM, 1.5 nM, 10 nM, 20 nM, 40 nM, 50 nM, 100 nM, 200 nM, 300 nM, 500 nM, 600 nM, 700 nM, 800 nM, or other concentration from about 0.01 nM to about 1000 nM.
  • Cells are exposed for 1 to 5 days or 1 to 3 days. For example, cells are exposed 1 day, 2 days, 3 days, 4 days or 5 days.
  • HDAC inhibitor MS-275 is used.
  • suitable HDAC inhibitors include, but are not limited to, any the following:
  • Trichostatin A and its analogs for example: trichostatin A (TSA); and trichostatin C (Koghe et al. 1998, Biochem. Pharmacol. 56: 1359-1364).
  • Peptides for example: oxamflatin [(2E)-5-[3-[(phenylsulfonyl)aminophenyl]-pent-2-ene-4-inohydroxamic acid (Kim et al., Oncogene 18: 2461-2470 (1999)); Trapoxin A (cylco-(L-phenylalanyl-L-phenylalanyl-D-pipecolinyl-L-2-amino-8-oxo-9,10-epoxy-decanoyl) (Kijima et al., J. Biol. Chem.
  • FR901228 depsipeptide
  • FR225497 cyclic tetrapeptide
  • apicidin cyclic tetrapeptide [cyclo-(N—O-methyl-L-tryptophanyl-L-isoleucinyl-D-pipecolinyl-L-2-amino-8-oxodecanoyl)] (Darkin-Rattray et al., Proc. Natl.
  • Hybrid polar compounds based on hydroxamic acid, for example: salicyl hydroxamic acid (SBHA) (Andrews et al., International J. Parasitology 30: 761-8 (2000)); suberoylanilide hydroxamic acid (SAHA) (Richon et al., Proc. Natl. Acad. Sci. U.S.A. 95: 3003-7 (1998)); azelaic bishydroxamic acid (ABHA) (Andrews et al., supra); azelaic-1-hydroxamate-9-anilide (AAHA) (Qiu et al., Mol. Biol.
  • SBHA salicyl hydroxamic acid
  • SAHA suberoylanilide hydroxamic acid
  • SAHA suberoylanilide hydroxamic acid
  • SAHA suberoylanilide hydroxamic acid
  • ABHA azelaic bishydroxamic acid
  • AAHA a
  • M-carboxy cinnamic acid bishydroxamide (CBHA) (Ricon et al., supra); 6-(3-chlorophenylureido) carpoic hydroxamic acid, 3-Cl-UCHA) (Richon et al., supra); MW2796 (Andrews et al., supra); and MW2996 (Andrews et al., supra).
  • SCFA Short chain fatty acid
  • valproic acid valerate (McBain et al., supra); 4-phenyl butyric acid (4-PBA) (Lea and Tulsyan, Anticancer RESearch 15: 879-3 (1995)); phenyl butyric acid (PB) (Wang et al., Cancer RESearch 59: 2766-99 (1999)); propinate (McBain et al., supra); butylamide (Lea and Tulsyan, supra); isobutylamide (Lea and Tulsyan, supra); phenyl acetate (Lea and Tulsyan, supra); 3-bromopropionate (Lea and Tulsyan, supra); tributyrin (Guan et al., Cancer RESearch 60: 749-55 (2000)); arginine butyrate; isobutyl amide; and valproate.
  • 4-PBA 4-phenyl butyric acid
  • PB phenyl butyric
  • Benzamide derivatives for example: MS-275 [N-(2-aminophenyl)-4-[N-(pyridine-3-yl-methoxycarbonyl)aminomethyl]benzamide] (Saito et al., Proc. Natl. Acad. Sci. U.S.A. 96: 4592-7 (1999)); and a 3′-amino derivative of MS-275 (Saito et al., supra); and CI-994.
  • a histone deacetylase inhibitor treatment may be carried out, for example, as follows.
  • the concentration of the HDAC inhibitor may depend on a particular inhibitor, but is preferably 0.001 nM to about 10 mM, and more preferably about 0.01 nM to about 1000 nM.
  • the effective amount or the dosage of a histone deacetylase inhibitor is defined as the amount of the histone deacetylase inhibitor that does not significantly decrease the survival rate of cells, specifically undifferentiated stem cells.
  • Cells are exposed for 1 to 5 days or 1 to 3 days. The exposure period may be less than one day. In a specific embodiment, cells are cultured for about 1 to 5 days, and then exposed to an effective amount of a histone deacetylase inhibitor.
  • the histone deacetylase inhibitor may be added at the start of culturing.
  • a gene-carrying vehicle such as a vector containing a nucleic acid encoding three genes (Oct3/4, Sox2 and Klf4) is introduced into cultured cells by a known method.
  • Forced expression of the IFs may comprise introducing one or more mammalian expression vectors encoding an Oct3/4, a Sox2, and a Klf4 polypeptide to a population of cells.
  • the IFs may be introduced into the cells as exogenous genes.
  • the exogenous genes are integrated into the genome of a host cell and its progeny. In other cases, the exogenous genes persist in an episomal state in the host cell and its progeny.
  • Exogenous genes are genes that are introduced to the cell from an external source.
  • a gene as used herein is a nucleic acid that includes an open reading frame encoding a polypeptide of interest, e.g., an IF.
  • the gene preferably includes a promoter operably linked to an open reading frame.
  • a natural version of the gene may already exist in the cell but an additional “exogenous gene” is added to the cell to induce polypeptide expression.
  • the one or more mammalian expression vectors may be introduced into greater than 20% of the total population of cells, e.g., 25%, 30%, 35%, 40%, 44%, 50%, 57%, 62%, 70%, 74%, 75%, 80%, 90%, or other percent of cells greater than 20%.
  • a single mammalian expression vector may contain two or more of the just-mentioned IFs.
  • one or more expression vectors encoding an Oct 3/4, Sox2, Klf4, and c Myc polypeptide are used.
  • each of the IFs to be expressed is encoded on a separate mammalian expression vector.
  • the IFs are genetically fused in frame with a transport protein amino acid sequence, e.g., that of a VP22 polypeptide as described in, e.g., U.S. Pat. Nos. 6,521,455, 6,251,398, and 6,017,735.
  • a transport protein amino acid sequence e.g., that of a VP22 polypeptide as described in, e.g., U.S. Pat. Nos. 6,521,455, 6,251,398, and 6,017,735.
  • Such VP22 sequences confer intercellular transport of VP22 fusion polypeptides from cells that have been transfected with a VP22 fusion polypeptide expression vector to neighboring cells that have not been transfected or transduced. See, e.g., Lemken et al (2007), Mol Ther, 15(2):310-319. Accordingly, the use of IF-VP22 fusion polypeptides can significantly increase the functional efficiency of transfected mammalian expression vector
  • suitable mammalian expression vectors include, but are not limited to: recombinant viruses, nucleic acid vectors, such as plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, human artificial chromosomes, cDNA, cRNA, and PCR product expression cassettes.
  • suitable promoters for driving expression of IFs in include retroviral LTR elements; constitutive promoters such as CMV, HSV1-TK, SV40, EF-1a, 13 actin; PGK, and inducible promoters, such as those containing Tet-operator elements.
  • one or more of the mammalian expression vectors encodes, in addition to an IF, a marker gene that facilitates identification or selection of cells that have been transfected or infected.
  • marker genes include, but are not limited to, fluorescent protein genes, e.g., for EGFP, DS-Red, YFP, and CFP; proteins conferring resistance to a selection agent, e.g., the neoR gene, and the blasticidin resistance gene.
  • Forced expression of an IF may be accomplished by introducing a recombinant virus carrying DNA or RNA encoding an IF to one or more cells. Additionally, the recombinant virus may carry DNA or RNA encoding more than 1 IF. This includes multiple copies of a single IF or multiple Ws contained within a single virus. For ease of reference, at times a virus will be referred to herein by the IF it is encoding. For example, a virus encoding an Oct3/4 polypeptide, may be described as an “Oct3/4 virus.” In certain cases, a virus may encode more than one copy of an IF or may encode more than one IF, e.g., two IFs, at a time.
  • the set of recombinant viruses may include combinations included in any set of IFs described herein.
  • the set of recombinant viruses may include at least: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus.
  • the set of recombinant viruses may be limited to a set of four recombinant viruses: an Oct3/4 virus, a Sox2 virus, a Klf4 virus, and a c-Myc virus.
  • the set of recombinant viruses is limited to a set of at least: an Oct3/4 virus, a Sox2 virus, a Klf4 virus, and a c-Myc virus.
  • the set of recombinant viruses is limited to Oct 3/4, Sox2, and Klf4 viruses.
  • the set of recombinant viruses may be limited a set of at least: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus.
  • the set of recombinant viruses includes three recombinant viruses, wherein two of the three recombinant viruses are an Oct3/4 virus and a Sox2 virus.
  • the set of recombinant viruses may be limited to a Sox2 virus and a c-Myc virus.
  • the set of recombinant viruses does not include a recombinant virus that encodes a polypeptide that might increase the risk of cell transformation, e.g., a c-Myc polypeptide.
  • the set of recombinant viruses can include: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus but not a c-Myc virus.
  • the set of recombinant viruses includes a c-Myc virus.
  • the c-Myc polypeptide encoded by the c-Myc virus may be wild-type c-Myc or a constitutively active variant of c-Myc.
  • the set includes a virus encoding c-Myc polypeptide capable of inducible activity, e.g., a c-Myc-ER polypeptide, see, e.g., Littlewood, et al. (1995) Nucleic Acid Res. 23(10):1686-90.
  • the set of recombinant viruses may include: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus, but not a TERT virus, a SV40 Large T antigen virus, HPV16 E6 virus, a HPV16 E7 virus, or a Bmi1 virus.
  • the set of recombinant viruses does not include a TERT virus.
  • the set of recombinant viruses does not include a SV40 virus.
  • the set of recombinant viruses does not include a HPV16 E6 virus or a HPV16 E7 virus.
  • a set of recombinant viruses may include viruses in addition to an Oct 3/4, a Sox2, and a Klf4 virus.
  • additional recombinant viruses include, but are not limited to Nanog, TERT, CYP26A1, GDF3, FoxD3, Zfp42, Dnmt3b, Ecat1, and Tel1 viruses.
  • the set of recombinant viruses includes any IF variant described herein.
  • Individual viruses may be added to the cells sequentially in time or simultaneously.
  • at least one virus e.g., an Oct3/4 virus, a Sox2 virus, a Klf4 virus, or a c-Myc virus, is added to the cells at a time different from the time when one or more other viruses are added.
  • the Oct3/4 virus, Sox2 virus and KlF4 virus are added to the cells simultaneously, or very close in time, and the c-Myc virus is added at a time different from the time when the other viruses are added.
  • At least two recombinant viruses may be added to the cells simultaneously or very close in time.
  • Oct3/4 virus and Sox2 virus are added simultaneously, or very close in time, and the Klf4 virus or c-Myc virus is added at a different time.
  • Oct3/4 virus and Sox2 virus; Oct3/4 virus and Klf4 virus; Oct3/4 virus and c-Myc virus; Sox2 virus and Klf4 virus; Sox2 virus and c-Myc virus; or Klf4 and c-Myc virus are added simultaneously or very close in time.
  • At least three viruses e.g., an Oct3/4 virus, a Sox2 virus, and a Klf4 virus
  • at least four viruses e.g., Oct3/4 virus, Sox2 virus, Klf4 virus, and c-Myc virus are added to the cells simultaneously or very close in time.
  • one or more Oct3/4 virus, Sox2 virus, Klf4 virus, or c-Myc virus is added to the cells at least two, at least three, or at least four separate times.
  • recombinant viruses include, but are not limited, to retroviruses (including lentiviruses); adenoviruses; and adeno-associated viruses.
  • the recombinant retrovirus is murine moloney leukemia virus (MMLV), but other recombinant retroviruses may also be used, e.g., Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus (MLV), Mink-Cell focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus, Gibbon Abe Leukemia Virus, Mason Pfizer Monkey Virus, or Rous Sarcoma Virus, see, e.g., U.S. Pat. No. 6,333,195.
  • MMLV murine moloney leukemia virus
  • retroviruses e.g., Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus (
  • the recombinant retrovirus is a lentivirus (e.g., Human Immunodeficiency Virus-1 (HIV-1); Simian Immunodeficiency Virus (SW); or Feline Immunodeficiency Virus (FIV)), See, e.g., Johnston et al (1999), Journal of Virology 73(6)” 4991-5000 (FIV); Négre D et al (2002) Current Topics in Microbiology and Immunology 261:53-74 (SIV); Naldini et al (1996) Science. 272:263-267 (HIV).
  • HIV-1 Human Immunodeficiency Virus-1
  • SW Simian Immunodeficiency Virus
  • FV Feline Immunodeficiency Virus
  • the recombinant retrovirus may comprise a viral polypeptide (e.g., retroviral env) to aid entry into the target cell.
  • a viral polypeptide e.g., retroviral env
  • the viral polypeptide may be an amphotropic viral polypeptide, e.g., amphotropic env, that aids entry into cells derived from multiple species, including cells outside of the original host species. See, e.g., id.
  • the viral polypeptide may be a xenotropic viral polypeptide that aids entry into cells outside of the original host species. See, e.g., id.
  • the viral polypeptide is an ecotropic viral polypeptide, e.g., ecotropic env, that aids entry into cells of the original host species. See, e.g., id.
  • viral polypeptides capable of aiding entry of retroviruses into cells include but are not limited to: MMLV amphotropic env, MMLV ecotropic env, MMLV xenotropic env, vesicular stomatitis virus-g protein (VSV-g), HIV-1 env, Gibbon Ape Leukemia Virus (GALV) env, RD114, FeLV-C, FeLV-B, MLV 10A1 env gene, and variants thereof, including chimeras. See e.g., Yee et al (1994), Methods Cell Biol. Pt A:99-112 (VSV-G); U.S. Pat. No. 5,449,614.
  • the viral polypeptide is genetically modified to promote expression or enhanced binding to a receptor.
  • a recombinant virus is produced by introducing a viral DNA or RNA construct into a producer cell.
  • the producer cell does not express exogenous genes.
  • the producer cell is a “packaging cell” comprising one or more exogenous genes, e.g., genes encoding one or more gag, pol, or env polypeptides and/or one or more retroviral gag, pol, or env polypeptides.
  • the retroviral packaging cell may comprise a gene encoding a viral polypeptide, e.g., VSV-g that aids entry into target cells.
  • the packaging cell comprises genes encoding one or more lentiviral proteins, e.g., gag, pol, env, vpr, vpu, vpx, vif, tat, rev, or nef.
  • the packaging cell comprises genes encoding adenovirus proteins such as E1A or E1B or other adenoviral proteins.
  • proteins supplied by packaging cells may be retrovirus-derived proteins such as gag, pol, and env, lentivirus-derived proteins such as gag, pol, env, vpr, vpu, vpx, vif, tat, rev, and nef; and adenovirus-derived proteins such as E1A and E1B.
  • the packaging cells supply proteins derived from a virus that differs from the virus from which the viral vector derives.
  • Packaging cell lines include but are not limited to any easily-transfectable cell line.
  • Packaging cell lines can be based on 293T cells, NIH3T3, COS or HeLa cell lines.
  • As packaging cells any cells may be used that can supply a lacking protein of a recombinant virus vector plasmid deficient in at least one gene encoding a protein required for virus packaging.
  • packaging cell lines include but are not limited to: Platinum-E (Plat-E); Platinum-A (Plat-A); BOSC 23 (ATCC CRL 11554); and Bing (ATCC CRL 11270), see, e.g., Morita et al (2000) Gene Therapy 7:1063-1066; Onishi et al (1996) Experimental Hematology 24:324-329; U.S. Pat. No. 6,995,009.
  • Commercial packaging lines are also useful, e.g., Ampho-Pak 293 cell line, Eco-Pak 2-293 cell line, RetroPack PT67 cell line, and Retro-X Universal Packaging System (all available from Clontech).
  • the retroviral construct may be derived from a range of retroviruses, e.g., MMLV, HIV-1, SIV, Fly, or other retrovirus described herein.
  • the retroviral construct may encode all viral polypeptides necessary for more than one cycle of replication of a specific virus. In some cases, the efficiency of viral entry is improved by the addition of other factors or other viral polypeptides. In other cases, the viral polypeptides encoded by the retroviral construct do not support more than one cycle of replication, e.g., U.S. Pat. No. 6,872,528. In such circumstances, the addition of other factors or other viral polypeptides can help facilitate viral entry.
  • the recombinant retrovirus is HIV-1 virus comprising a VSV-g polypeptide but not comprising a HIV-1 env polypeptide.
  • the retroviral construct may comprise: a promoter, a multi-cloning site, and/or a resistance gene.
  • promoters include but are not limited to CMV, SV40, EF1 ⁇ , ⁇ actin; retroviral LTR promoters, and inducible promoters.
  • the retroviral construct may also comprise a packaging signal (e.g., a packaging signal derived from the MFG vector; a psi packaging signal).
  • Examples of retroviral constructs known in the art include but are not limited to: pMX, pBabeX or derivatives thereof. See e.g., Onishi et al (1996) Experimental Hematology 24:324-329.
  • the retroviral construct is a self-inactivating lentiviral vector (SIN) vector, see, e.g., Miyoshi et al., (1998) J Virol. 72(10): 8150-8157.
  • the retroviral construct is LL-CG, LS-CG, CL-CG, CS-CG, CLG or MFG. Miyoshi et al., (1998) J Virol. 72(10): 8150-8157; Onishi et al (1996) Experimental Hematology 24:324-329; Riviere et al. (1995) PNAS 92: 6733-6737.
  • Virus vector plasmids include: pMXs, pMXs-IB, pMXs-puro, pMXs-neo (pMXs-IB is a vector carrying the blasticidin-resistant gene in stead of the puromycin-resistant gene of pMXs-puro) [Experimental Hematology, 2003, 31 (11): 1007-14], MFG [Proc. Natl. Acad. Sci. U.S.A.
  • the retroviral construct comprises blasticidin (e.g., pMXs-IB), puromycin (e.g., pMXs-puro, pBabePuro); or neomycin (e.g., pMXs-neo).
  • blasticidin e.g., pMXs-IB
  • puromycin e.g., pMXs-puro, pBabePuro
  • neomycin e.g., pMXs-neo
  • the retroviral construct may encode one or more IFs.
  • pMX vectors encoding Oct3/4, Sox2, Klf4, or c-Myc polypeptides, or variants thereof are generated or obtained.
  • Oct3/4 is inserted into pMXs-puro to create pMX-Oct3/4
  • Sox2 is inserted into pMXs-neo to create pMX-Sox2
  • Klf4 is inserted into pMXs-IB to create pMX-Klf4
  • c-Myc is inserted into pMXs-IB to create pMX-c-Myc.
  • the viral construct may be introduced by any method known in the art, including but not limited to: the calcium phosphate method [Kokai (Japanese Unexamined Patent Publication) No. 2-227075], the lipofection method [Proc. Natl. Acad. Sci. U.S.A. 84: 7413 (1987)], the electroporation method, microinjection, Fugene transfection, and the like, and any method described herein.
  • pMX-Oct3/4, pMX-Sox2, pMX-Klf4 or pMX-c-Myc is introduced into PlatE cells by Fugene HD (Roche) transfection.
  • the cell culture medium may be replaced with fresh medium comprising FBM (Lonza) supplemented with FGM-2 Single Quots (Lonza).
  • the medium is replaced from about 12 to about 60 hours following the introduction of the viral construct, e.g., from about 12 to about 18 hours; about 18 to about 24; about 24 to about 30; about 30 to about 36; about 36 to about 42; about 42 to about 48; about 48 to about 54; or about 54 to about 60 hours following introduction of the viral construct to the producer cells.
  • the medium may be replaced from about 24 to about 48 hours after introduction of the viral construct to the producer cells.
  • the supernatant can be recovered from about 4 to about 24 hours following the addition of fresh media, e.g., about 4 hours. In some cases, the supernatant may be recovered about every 4 hours following the addition of fresh media.
  • the recovered supernatant may be passed through a 0.45 uM filter (Millipore). In some cases, the recovered supernatant comprises retrovirus derived from one or more: pMX-Oct3/4, pMX-Sox2, pMX-Klf4 or pMX-c-Myc.
  • Adenoviral transduction may be used to force expression of the sets of IFs.
  • Methods for generating adenoviruses and their use are well established as described in, e.g., Straus, The Adenovirus, Plenum Press (NY 1984), 451 496; Rosenfeld, et al, Science, 252:431-434 (1991); U.S. Pat. Nos. 6,203,975, 5,707,618, and 5,637,456.
  • adenoviral-associated viral transduction is used to force expression of the sets of Ws.
  • Methods for preparing adeno-associated viruses and their use are well established as described in, e.g., U.S. Pat. Nos. 6,660,514 and 6,146,874.
  • an adenoviral construct is obtained or generated, wherein the adenoviral construct, e.g., Adeno-X, comprises DNA encoding Oct3/4, Sox2, Klf4, or c-Myc.
  • An adenoviral construct may be introduced by any method known in the art, e.g., Lipofectamine 2000 (Invitrogen) or Fugene HD (Roche), into HEK 293 cells.
  • the method further comprises (1) collecting the cells when they exhibit a cytopathic effect (CPE), such effect occurring from about 10 to about 20 days, e.g., about 11, 13, 14, 15, 18, or 20 days after transfection (2) subjecting the cells to from about 2 to about 5 freeze-thaw cycles, e.g., about 3, (3) collecting the resulting virus-containing liquid; (4) purifying the virus using an adenovirus purification kit (Clontech) and (5) storing the virus at ⁇ 80° C.
  • the titer, or plaque-forming unit (PFU), of the adenoviral stocks is determined using an Adeno-X rapid titer kit (Clontech), as described herein.
  • the cells may be infected with a recombinant retrovirus that naturally targets a different cell type or cells originating from a different host.
  • an exogenous receptor may be first introduced into the human cells.
  • an exogenous mouse receptor may be added to human cells, e.g., postnatal dermal fibroblasts, in order help entry of murine moloney leukemia virus (MMLV).
  • MMLV murine moloney leukemia virus
  • the exogenous receptor may improve infection efficiency by facilitating viral entry, especially if the receptor recognizes a viral polypeptide, e.g., MMLV env, or HIV env.
  • exogenous receptors include but are not limited to any receptor recognized by a specific retrovirus or lentivirus known in the art.
  • a murine receptor mCAT1, GenBank Accession No NM — 007513 protein is used in order to aid MMLV infection of a human target cell.
  • a CXCR4 or CCR5 receptor is used to aid HIV-1 infection of a target cell.
  • the exogenous receptor may be introduced by methods described herein. Methods of introducing the exogenous receptor include but are not limited to: calcium phosphate transfection, Lipofectamine transfection, Fugene transfection, microinjection, or electroporation.
  • a virus e.g., recombinant adenovirus or retrovirus (including lentivirus), is used to introduce the exogenous receptor to the target cell.
  • a recombinant adenovirus is used to introduce MCAT1 to human cells and then a recombinant retrovirus, e.g., MMLV, is used to introduce the IF genes, e.g., Oct 3/4, a Sox2, a Klf4, or c-Myc, to the cells.
  • a recombinant retrovirus e.g., MMLV
  • the IF genes e.g., Oct 3/4, a Sox2, a Klf4, or c-Myc
  • a solution of adenovirus comprising DNA encoding the mCAT1 protein, e.g., an adenovirus generated by using a pADEX-mCAT1 construct, is generated or obtained.
  • the adenovirus solution can comprise Hanks' balanced salt solution.
  • infection of cells is accomplished by: (1) contacting the p-ADEX-mCAT1 adenovirus solution with cells, e.g., human, non-embryonic fibroblasts, at a multiplicity of infection (m.o.i.) from about 1:5 to about 1:50, e.g., about 1:5, about 1:7; about 1:10; about 1:15, about 1:20, about 1:25; about 1:30, about 1:35; about 1:40; about 1:45, or about 1:50; (2) incubating the cells with the adenovirus solution at room temperature from about 15 minutes to about 2 hours, e.g., about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 1.25 hours, about 1.5 hours, about 1.75 hours, or about 2 hours; and (3) culturing the somatic cell population in culture medium from about 24 hours to about 60 hours, e.g., about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 54
  • the cells can be infected using a wide variety of methods.
  • the infection of cells occurs by (1) combining one or more, two or more, three or more, or all four: pMX-Oct3/4 retrovirus, pMX-Sox2 retrovirus, pMX-Klf4, or pMX-c-Myc to obtain a retrovirus solution (2) supplementing the retrovirus solution with from about 2 ug/ml to about 15 ug/ml Polybrene, e.g., about 2 ug/ml, about 3 ug/ml, about 5 ug/ml, about 7 ug/ml, about 10 ug/ml, about 12 ug/ml, or about 15 ug/ml Polybrene; (3) contacting the retroviral solution with the somatic cells, at a m.o.i.
  • step (3) of from about 1:100 to about 1:500, e.g., about 1:100, about 1:150, about 1:200, about 1:250, about 1:300, about 1:350, about 1:400, about 1:450, or about 1:500 m.o.i.; (4) allowing the contacting of step (3) to continue at 37° C.
  • step (4) changing the medium to MC-ES medium, as described herein; and (6) changing the MC-ES medium with fresh medium every 1 to 2 days.
  • infection of somatic cells occurs by following steps (1) through (6) described herein, with the added step of pre-incubating the somatic cells for a length of time, e.g., about 48 hours, prior to contacting the cells with the retroviral solution.
  • a length of time e.g., about 48 hours
  • pre-incubation may be necessary when the somatic cell expresses an exogenous receptor that was introduced by viral transduction, transfection, or other method.
  • an adenovirus or lentivirus is used to introduce an exogenous receptor, e.g., mCAT1, to the somatic cell; such cells may need to be cultured for a length of time from at least about 30 hours to at least about 60 hours, e.g., about 30, about 35, about 40, about 48, about 52, about 55, or about 60 hours.
  • the infection of cells may be accomplished by any method known in the art. e.g., Palsson, B., et al. WO95/10619. Apr. 20, 1995; Morling, F. J. et al. (1995). Gene Therapy. 2: 504-508; Gopp et al. (2006) Methods Enzymol. 420:64-81.
  • the infection may be accomplished by spin-infection or “spinoculation” methods that involve subjecting the cells to centrifugation during the period closely following the addition of virus to the cells.
  • virus may be concentrated prior to the infection, e.g., by ultracentrifugation.
  • other technologies may be used to aid or improve entry of retroviruses into the target cell.
  • the retrovirus may be contacted with a liposome or immunoliposome to aid or direct entry into a specific cell type. See, e.g., Tan et al. (2007) Mol. Med. 13(3-4): 216-226.
  • the methods of infecting cells described herein may be used to infect cells expressing an exogenous receptor, e.g., MCAT1 or other exogenous receptor described herein. Depending on how the exogenous receptor was introduced, the preincubation period of the cells prior to infection may need to be varied. In some cases, cells that do not express an exogenous receptor are used. Some recombinant retroviruses, e.g., VSV-G pseudotyped recombinant retroviruses, may not need the aid of an exogenous receptor in order to efficiently enter cells. In some examples, VSV-G pseudotyped recombinant retrovirus is introduced to cells following the method described herein, except that the timing of the preculturing of the cells may vary.
  • an exogenous receptor e.g., MCAT1 or other exogenous receptor described herein.
  • Nucleic acid vector transfection e.g., transient transfection
  • Methods for preparation of transfection-grade nucleic acid expression vectors are well established. See, e.g., Sambrook and Russell (2001), “Molecular Cloning: A Laboratory Manual,” 3rd ed, (CSHL Press).
  • Examples of high efficiency transfection efficiency methods include “nucleofection,” as described in, e.g., Trompeter (2003), J Immunol Methods, 274(1-2):245-256, and in international patent application publications WO2002086134, WO200200871, and WO2002086129, transfection with lipid-based transfection reagents such as Fugene® 6 and Fugene® HD (Roche), DOTAP, and lipofectamineTM LTX in combination with the PLUSTM (Invitrogen, Carlsbad, Calif.), DreamfectTM (OZ Biosciences, Marseille, France), GeneJuiceTM (Novagen, Madison, Wis.), polyethylenimine (see, e.g., Lungwitz et al (2005), Eur J Pharm Biopharm, 60(2):247-266), and GeneJammerTM (Stratagene, La Jolla, Calif.), and nanoparticle transfection reagents as described in, e.g., U.S
  • the induction methods may use protein transduction to introduce at least one of the IFs directly into cells.
  • protein transduction method includes contacting cells with a composition containing a carrier agent and at least one purified polypeptide comprising the amino acid sequence of one of the above-mentioned IFs.
  • suitable carrier agents and methods for their use include, but are not limited to, commercially available reagents such as ChariotTM (Active Motif, Inc., Carlsbad, Calif.) described in U.S. Pat. No.
  • the protein transduction method may comprise contacting a cells with at least one purified polypeptide comprising the amino acid sequence of one of the above-mentioned TAs fused to a protein transduction domain (PTD) sequence (IF-PTD fusion polypeptide).
  • PTD protein transduction domain
  • IF-PTD fusion polypeptide The PTD domain may be fused to the amino terminal of an IF sequence; or, the PTD domain may be fused to the carboxy terminal of an IF sequence.
  • the IF-PTD fusion polypeptide is added to cells as a denatured polypeptide, which may facilitate its transport into cells where it is then renatured. Generation of PTD fusion proteins and methods for their use are established in the art as described in, e.g., U.S. Pat. Nos.
  • Exemplary PTD domain amino acid sequences include, but are not limited to, any of the following: YGRKKRRQRRR (SEQ ID NO:1); RKKRRQRR (SEQ ID NO:2); YARAAARQARA (SEQ ID NO:3); THRLPRRRR (SEQ ID NO:4); and GGRRARRRRRR (SEQ ID NO:5).
  • individual purified IF polypeptides are added to cells sequentially at different times.
  • a set of at least three purified IF polypeptides, but not a purified c-Myc polypeptide, e.g., an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide are added to cells.
  • a set of four purified IF polypeptides, e.g., purified Oct3/4, Sox2, Klf4, and c-Myc polypeptides are added to cells.
  • the purified IF polypeptides are added to cells as one composition (i.e., a composition containing a mixture of the IF polypeptides).
  • cells are incubated in the presence of a purified IF polypeptide for about 30 minutes to about 24 hours, e.g., 1 hours, 1.5 hours, 2 hours, 2.5 hours, 3 hours, 3.5 hours 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 12 hours, 16 hours, 18 hours, 20 hours, or any other period from about 30 minutes to about 24 hours.
  • protein transduction of cells is repeated with a frequency of about every day to about every 4 days, e.g., every 1.5 days, every 2 days, every 3 days, or any other frequency from about every day to about every four days
  • Forced expression of IFs may also be achieved by using nucleic acid-free IF-containing protein transducing nanoparticles (PTN). Details of methods for generating and using PTNs are found in, e.g., Link et al (2006), Nuc Acids Res, 34(2):e16.
  • PTN nucleic acid-free IF-containing protein transducing nanoparticles
  • the methods described herein utilize protein transduction and expression vector transduction/transfection in any combination to force expression of a set of IFs as described herein.
  • retroviral expression vectors are used to force expression of Oct 3/4, a Sox2, and a Klf4 polypeptides in cells, and purified c-Myc purified polypeptide is introduced into cells by protein transduction as described herein.
  • HDAC inhibitor treatment can be used in addition to the purified IF polypeptide.
  • a set of at least three purified IF polypeptides, but not a purified c-Myc polypeptide, e.g., an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide are added to cells which are also subjected to HDAC inhibitor treatment.
  • Cell colonies may be subcloned by any method known in the art.
  • the iPSCs are cultured and observed for about 14 days to about 40 days, e.g., 15, 16, 17, 18, 19, 20, 23, 24, 27, 28, 29, 30, 31, 33, 34, 35, 36, 37, 38 days, or any other period from about 14 days to about 40 days prior to identifying and selecting clones comprising “iPSCs” based on morphological characteristics.
  • Morphological characteristics for identifying iPSC clones include, but are not limited to, a small cell size with a high nucleus-to-cytoplasm ratio; formation of small monolayer colonies within the space between parental cells (e.g., between fibroblasts).
  • the cell suspension in the ring containing the detached colonies is then added to about 2 ml of MC ES medium (or other medium described herein), and plated in one well of a MEF-coated 24-well plate or other cell culture vessel of equivalent surface area. After culturing the colony-derived cells in a 5% CO 2 cell culture incubator at 37° C. for about 14 hours, the medium is replaced. Subsequently, the medium is replaced about every two days until about 8 days later when a second subculture is carried out.
  • MC ES medium or other medium described herein
  • the medium in the first subculture, the medium is removed, the cells are washed with Hank's balanced salt solution, and Detachment Medium For Primate ES Cells (ReproCell, Tokyo, Japan) is then added to the cells and incubated at 37° C. for 10 minutes. After the incubation, MC-ES medium (2 ml) is added to the resulting cell suspension to quench the activity of the Detachment Medium. The cell suspension is then transferred to a centrifuge tube, and centrifuged at 200 ⁇ g at 4° C. for 5 minutes.
  • Detachment Medium For Primate ES Cells (ReproCell, Tokyo, Japan) is then added to the cells and incubated at 37° C. for 10 minutes. After the incubation, MC-ES medium (2 ml) is added to the resulting cell suspension to quench the activity of the Detachment Medium. The cell suspension is then transferred to a centrifuge tube, and centrifuged at 200 ⁇ g at 4° C. for 5 minutes.
  • the supernatant is removed, the cell pellet is resuspended in MC ES medium, and the resuspended cells are plated on four wells of a MEF-coated 24-well plate and cultured for about seven days until a second subculture is prepared.
  • the second subculture prepared by the method described above, cells are plated on a 60 mm cell culture dish coated with matrigel at a concentration of 20 ⁇ g/cm 2 .
  • a third subculture is prepared in which cells are plated on two matrigel-coated 60 mm cell culture dishes, one of which can subsequently be used for gene expression analysis and the other for continued passaging as described below.
  • One of the subcultures is used for gene expression analysis, as described herein, and the other is passaged as needed to maintain a cell line derived from the iPSC clone.
  • the iPSCs may be subcultured about every 5 to 7 days.
  • the cells are washed with Hank's balanced salt solution, and dispase or Detachment Medium For Primate ES Cells is added, and incubated at 37° C. for 5 to 10 minutes.
  • MC-ES medium is added to quench enzymatic activity of the detachment medium, and the resulting cell/colony suspension is transferred to a centrifuge tube. Colonies in the suspension are allowed to settle on the bottom of the tube, the supernatant is carefully removed, and MC-ES medium is then added to resuspend the colonies.
  • any extremely large ones are broken up into smaller sizes by slow up and down pipetting.
  • Appropriately sized colonies are plated on a matrigel-coated plastic culture dish with a base area of about 3 to 6 times that before subculture.
  • Examples of culture media useful for culturing human pluripotent stem cells induced from undifferentiated stem cells present in a human postnatal tissue of the present invention include, but are not limited to, the ES medium, and a culture medium suitable for culturing human ES cells such as MEF-conditioned ES medium (MC-ES) or other medium described herein, e.g., mTeSRTTM.
  • MC-ES MEF-conditioned ES medium
  • mTeSRTTM e.g., mTeSRTTM.
  • the cells are maintained in the presence of a ROCK inhibitor, as described herein.
  • Cell colonies subcultured from those initially identified on the basis of morphological characteristics may be assayed for any of a number of properties associated with pluripotent stem cells, including, but not limited to, expression of alkaline phosphatase activity, expression of ES cell marker genes, expression of protein markers, hypomethylation of Oct3/4 and Nanog promoters relative to a parental cells, long term self-renewal, normal diploid karyotype, and the ability to form a teratoma comprising ectodermal, mesodermal, and endodermal tissues.
  • properties associated with pluripotent stem cells including, but not limited to, expression of alkaline phosphatase activity, expression of ES cell marker genes, expression of protein markers, hypomethylation of Oct3/4 and Nanog promoters relative to a parental cells, long term self-renewal, normal diploid karyotype, and the ability to form a teratoma comprising ectodermal, mesodermal, and endodermal tissues.
  • a number of assays and reagents for detecting alkaline phosphatase activity in cells are known in the art.
  • colonies to be analyzed are fixed with a 10% formalin neutral buffer solution at room temperature for about 5 minutes, e.g., for 2 to 5 minutes, and then washed with PBS.
  • a chromogenic substrate of alkaline phosphatase, 1 step BCIP (5-Bromo-4-Chloro-3′-Indolylphosphate p-Toluidine Salt) and NBT (Nitro-Blue Tetrazolium Chloride) manufactured by Pierce (Rockford, Ill.) is then added and reacted at room temperature for 20 to 30 minutes. Cells having alkaline phosphatase activity are stained blue-violet.
  • Putative iPS cell colonies tested for alkaline phosphatase activity may be then assayed for expression of a series of human embryonic stem cell marker (ESCM) genes including, but not limited to, Nanog, TDGF1, Dnmt3b, Zfp42, FoxD3, GDF3, CYP26A1, TERT, Oct 3/4, Sox2, Sa114, and HPRT.
  • ECM embryonic stem cell marker
  • TDGF1, Dnmt3b, Zfp42, FoxD3, GDF3, CYP26A1, TERT, Oct 3/4, Sox2, Sa114, and HPRT See, e.g., Assou et al (2007), Stem Cells, 25:961-973.
  • Many methods for gene expression analysis are known in the art. See, e.g., Lorkowski et al (2003), Analysing Gene Expression, A Handbook of Methods: Possibilities and Pitfalls, Wiley-VCH.
  • suitable nucleic acid-based gene expression assays
  • levels of ESCM gene mRNA expression levels in putative iPS cell colonies are determined by qRT-PCR.
  • Putative iPS cell colonies are harvested, and total RNA is extracted using the “Recoverall total nucleic acid isolation kit for formaldehyde- or paraformaldehyde-fixed, paraffin-embedded (FFPE) tissues” (manufactured by Ambion, Austin, Tex.).
  • FFPE paraffin-embedded
  • the colonies used for RNA extraction are fixed colonies, e.g., colonies that have been tested for alkaline phosphatase activity. The colonies can be used directly for RNA extraction, i.e., without prior fixation.
  • the target gene is amplified using the TaqMan® PreAmp mastermix (manufactured by Applied Biosystems, Foster City, Calif.).
  • Real-time quantitative PCR is performed using an ABI Prism 7900HT using the following PCR primer sets (from Applied Biosystems) for detecting mRNA of the above-mentioned ESCM genes: Nanog, Hs02387400_g1, Dnmt3b, Hs00171876_m1, FoxD3, Hs00255287_s1, Zfp42, Hs01938187_s1, TDGF1, Hs02339499_g1, TERT, Hs00162669_m1, GDF3, Hs00220998_m1, CYP26A1, Hs00175627_m1, GAPDH, Hs99999905_ml).
  • Putative iPS cell colonies may be assayed by an immunocytochemistry method for expression of protein markers including, but not limited to, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, CD9, CD24, Thy-1, and Nanog.
  • immunocytochemistry assays e.g., fluorescence immunocytochemistry assays
  • expression of one or more of the above-mentioned protein markers in putative iPS cell colonies is assayed as follows. Cultured cells are fixed with 10% formaldehyde for 10 min and blocked with 0.1% gelatin/PBS at room temperature for about an hour. The cells are incubated overnight at 4° C. with primary antibodies against SSEA-3 (MC-631; Chemicon), SSEA-4 (MC813-70; Chemicon), TRA-1-60 (ab16288; abcam), TRA-1-81 (ab16289; abcam), CD9 (M-L13; R&D systems), CD24 (ALB9; abcam), Thy1 (5E10; BD Bioscience), or Nanog (MAB1997; R&D Systems).
  • Nanog staining cells are permeabilized with 0.1% Triton X-100/PBS before blocking. The cell colonies are washed with PBS three times, then incubated with AlexaFluor 488-conjugated secondary antibodies (Molecular Probes) and Hoechst 33258 (Nacalai) at room temperature for 1 h. After further washing, fluorescence is detected with a fluorescence microscope, e.g., Axiovert 200M microscope (Carl Zeiss).
  • ESC marker genes in iPSC colonies may be assayed in live cells, which increases the efficiency of identifying iPSC colonies following an induction method as described herein.
  • Examples of ESC marker genes useful for identifying induced stem cell colonies include, e.g., Oct3/4, Nanog, Klf4, Lin28, Sox2, c-Myc, or TERT.
  • mRNA for one or more of these genes is detected in live cells.
  • mRNAs for two or more of the ESC marker genes is detected.
  • cells are contacted with one or more molecular beacon probes that hybridize to and signal the presence of one or more stem cell marker genes.
  • Molecular beacons are single-stranded oligonucleotide hybridization probes that form a stem-and-loop structure.
  • the loop contains a probe sequence that is complementary to a target sequence, and the stem is formed by the annealing of complementary arm sequences that are located on either side of the probe sequence.
  • a fluorophore is covalently linked to the end of one arm and a quencher is covalently linked to the end of the other arm.
  • MBs do not fluoresce when they are free in solution. However, when they hybridize to a target sequence they undergo a conformational change that enables them to fluoresce brightly.
  • the probe sequence may range in length from about 15 to about 30 nucleotides depending on the GC content of the target probe sequence.
  • the GC content of the target probe sequence should be from about 40 to about 60%.
  • the flanking stem sequences may range from about 5 to about 7 nucleotides with a GC content of about 75 to about 100 percent.
  • the design of MBs and their use to detect mRNA expression in living cells is known in the art, as described in, e.g., Rhee et al (2008), Nuc Acid Res, 36(5):e30.
  • Useful algorithms for determining melting temperatures of an MB duplex and an MB/target duplex are known in the art.
  • the iPSC colonies to be evaluated may be contacted about 14 days to about 50 days after initiating induction, e.g., 14 days to 21 days, 14 days to 28 days, 20 days to 45 days, 25 days to 40 days, 30 days to 35 days, 30 days to 50 days after induction.
  • cells are contacted with as low a concentration of an MB and as short a period as compatible with reliably detecting a signal.
  • the concentration of an MB of about 0.1 ⁇ M to about 5 ⁇ M (for each MB), e.g., 0.1 ⁇ M to 0.5 ⁇ M, 0.2 ⁇ M to 1 ⁇ M, 0.5 ⁇ M to 2 ⁇ M, or 3 ⁇ M to 5 ⁇ M.
  • Incubation periods with a MB may range from about 5 minutes to about two hours, e.g., 15 minutes to 30 minutes, 20 minutes to one hour, 30 minutes to 1.5 hours, 45 minutes to 2 hours, or any other time period form about 5 minutes to two hours.
  • MBs are introduced into the cells without the use of a transfection reagent.
  • a transfection reagent optimized for oligonucleotide transfection is utilized, e.g., TransIT® oligo transfection reagent kit or any other transfection reagents known in the art.
  • streptolysin-O is used to transiently permealize cells to allow entry of the MBs into the cells. This method is described in, e.g., Rhee et al supra and Santangelo et al (2004), Nuc Acids Res, 32(6): e57.
  • MBs are added to adherent cell cultures and cell colonies found to be positive for expression of one or more ESC marker genes are picked off the substrate as described above.
  • MBs are added to iPSCs in suspension and ESC-positive cells are selected by FACS or any other fluorescence based sorting method.
  • MBs are added to adherent iPSCs, which are then dispersed prior to FACS selection. Use of FACS for selection of iPSCs is particularly useful for high throughput generation of iPSC lines and panels of iPSC lines.
  • a characteristic of the iPSCs is reduced methylation of the genomic promoters of Oct3/4 and Nanog relative to those of their parental cells.
  • Suitable Oct3/4 promoter regions to be analyzed include, but are not limited to, the Oct3/4 proximal promoter including conserved region 1 (CR1) and the Oct3/4 promoter distal enhancer including CR4.
  • Suitable Nanog promoter regions to be analyzed include, but are not limited to, the Nanog proximal promoter including the Oct3/4 and Sox2 binding sites. See, e.g., Rodda et al (2005), J Biol Chem, 280:24731-24737 and Yang et al (2005), J Cell Biochem, 96:821-830.
  • genomic DNA isolated from putative iPSCs and cells used for a comparison is isolated and treated with bisulfate.
  • Bisulfite-treated genomic DNA is then PCR-amplified with primers containing a T7 promoter sequence.
  • RNA transcripts are generated using T7 polymerase and then treated with RNAse A to generate methylation-specific cleavage products. Methylation of individual CpG sites is assessed by MALDI-TOF mass spectrometry of the cleavage products.
  • a detailed description of the method is provided in, e.g., Ehich et al (2005), Proc Natl Acad Sci USA, 102:15785-15790.
  • iPSCs are assessed for their ability to be passaged continuously in vitro.
  • the iPSCs are assayed for their ability to be passaged for at least about 30 to at least about 100 times in vitro, e.g., about 33, 35, 40, 45, 51, 56, 60, 68, 75, 80, 90, 93, 100, or any other number of passages from at least about 30 to at least about 100 passages.
  • iPSCs are assayed for their ability to proliferate for a period of about 30 days to about 500 days from initiation of forced expression of IFs in parental cells, e.g., 40 days, 50 days, 60 days, 70 days, 80 days, 100 days, 150 days, 180 days, 200 days, 250 days, 300 days, 400 days, 450 days or any other period from about 30 days to about 500 days from initiation of forced expression of IFs in the parental cells.
  • long-term self-renewal of iPSCs is determined when the cells are passaged in a defined medium (e.g., mTeSR1 medium) and in the absence of feeder cells, e.g., mTeSR1 medium as described herein.
  • cells are passaged in MC-ES medium as described herein.
  • iPSCs are assessed for diploidy and a normal, stable karyotype, e.g., stable after the cells of have been passaged for at least one year in vitro.
  • a normal, stable karyotype e.g., stable after the cells of have been passaged for at least one year in vitro.
  • a number of karotype analysis methods are known in the art.
  • the karyotype analysis method is multicolor FISH as described in, e.g., Bayani et al (2004), Curr Protoc Cell Biol, Chapter 22: Unit 22.5.
  • the karyotype analysis includes a molecular karyotype analysis as described in, e.g., Vermeesch et al (2007), Eur J Hum Genet, 15(11):1105-1114.
  • iPSCs are pretreated with 0.02 ⁇ g/ml colecemid for about 2 to about 3 hours, incubated with about 0.06 to about 0.075M KCl for about 20 minutes, and then fixed with Carnoy's fixative. Afterwards, for multicolor FISH analysis, cells are hybridized with multicolor FISH probes, e.g., those in the Star*FISH ⁇ Human Multicolour FISH (M-FISH) Kit from Cambio, Ltd (Cambridge, UK).
  • M-FISH Human Multicolour FISH
  • pluripotent stem cells have the ability to form a teratoma, comprising ectodermal, mesodermal, and endodermal tissues, when injected into an immunocompromised animal.
  • Induced cells or induced pluripotent stem cells (iPS) or ES cell-like pluripotent stem cells may refer to cells having an in vitro long-term self-renewal ability and the pluripotency of differentiating into three germ layers, and said pluripotent stem cells may form a teratoma when transplanted into a test animal such as mouse.
  • the iPSCs may be assessed for pluripotency in a teratoma formation assay in an immunocompromised animal model.
  • the immunocompromised animal may be a rodent that is administered an immunosuppressive agent, e.g., cyclosporin or FK-506.
  • the immunocompromised animal model may be a SCID mouse.
  • About 0.5 ⁇ 10 6 to about 2.0 ⁇ 10 6 e.g., 0.6 ⁇ 10 6 , 0.8 ⁇ 10 6 , 1.0 ⁇ 10 6 , 1.2 ⁇ 10 6 , 1.5 ⁇ 10 6 , 1.7 ⁇ 10 6 , or other number of iPSCs from about 0.5 ⁇ 10 6 to about 2.0 ⁇ 10 6 iPSCs/mouse may be injected into the medulla of a testis of a 7- to 8-week-old immunocompromised animal. After about 6 to about 8 weeks, the teratomas are excised after perfusing the animal with PBS followed by 10% buffered formalin. The excised teratomas are then subjected to immunohistological analysis.
  • One method of distinguishing human teratoma tissue from host (e.g., rodent) tissue includes immunostaining for the human-specific nuclear marker HuNu.
  • Immunohistological analysis includes determining the presence of ectodermal (e.g., neuroectodermal), mesodermal, and endodermal tissues.
  • Protein markers for ectodermal tissue include, but are not limited to, nestin, GFAP, and integrin ⁇ 1.
  • Protein markers for mesodermal tissue include, but are not limited to, collagen II, Brachyury, and osteocalcin.
  • Protein markers for endodermal tissue include, but are not limited to, ⁇ -fetoprotein ( ⁇ FP) and HNF3beta.
  • global gene expression analysis is performed on putative iPS cell colonies.
  • Such global gene expression analysis may include a comparison of gene expression profiles from a putative iPS cell colony with those of one or more cell types, including but not limited to, (i) parental cells, i.e., one or more cells from which the putative iPS cell colony was induced; (ii) a human ES cell line; or (iii) an established iPS cell line.
  • gene expression data for human ES cell lines are available through public sources, e.g., on the world wide web in the NCBI “Gene Expression Omnibus” database. See, e.g., Barrett et al (2007), Nuc Acids Res, D760-D765.
  • comparison of gene expression profiles from a putative iPS colony to those of an ES cell line entails comparison experimentally obtained data from a putative iPS cell colony with gene expression data available through public databases.
  • human ES cell lines for which gene expression data are publicly available include, but are not limited to, hE14 (GEO data set accession numbers GSM151739 and GSM151741), Sheff4 (GEO Accession Nos GSM194307, GSM194308, and GSM193409), h_ES 01 (GEO Accession No. GSM194390), h_ES H9 (GEO Accession No. GSM194392), and h_ES BG03 (GEO Accession No. GSM194391).
  • RNA isolated from one or more iPS cell lines by a nucleic acid microarray hybridization assay.
  • suitable microarray platforms for global gene expression analysis include, but are not limited to, the Human Genome U133 plus 2.0 microarray (Affymetrix) and the Whole Human Genome Oligo Micoarray (Agilent).
  • gene expression data from cells produced by the methods described herein are compared to those obtained from other cell types including, but not limited to, human ES cell lines, parental cells, and multipotent stem cell lines.
  • Suitable statistical analytical metrics and methods include, but are not limited to, the Pearson Correlation, Euclidean Distance, Hierarchical Clustering (See, e.g., Eisen et al (1998), Proc Natl Acad Sci USA, 95(25): 14863-14868), and Self Organizing Maps (See, e.g., Tamayo et al (1999), Proc Natl Acad Sci USA, 96(6):2907-2912.
  • iPSC lines may be differentiated into cell-types of various lineages.
  • differentiated cells include any differentiated cells from ectodermal (e.g., neurons and fibroblasts), mesodermal (e.g., cardiomyocytes), or endodermal (e.g., pancreatic cells) lineages.
  • the differentiated cells may be one or more: pancreatic beta cells, neural stem cells, neurons (e.g., dopaminergic neurons), oligodendrocytes, oligodendrocyte progenitor cells, hepatocytes, hepatic stem cells, astrocytes, myocytes, hematopoietic cells, or cardiomyocytes.
  • the differentiated cells derived from the iPSCs may be terminally differentiated cells, or they may be capable of giving rise to cells of a specific lineage.
  • iPSCs can be differentiated into a variety of multipotent cell types, e.g., neural stem cells, cardiac stem cells, or hepatic stem cells.
  • the stem cells may then be further differentiated into new cell types, e.g., neural stem cells may be differentiated into neurons; cardiac stem cells may be differentiated into cardiomyocytes; and hepatic stem cells may be differentiated into hepatocytes.
  • Methods for differentiating iPSCs are further disclosed in U.S. application Ser. Nos.
  • iPSCs There are numerous methods of differentiating the iPSCs into a more specialized cell type. Methods of differentiating iPSCs may be similar to those used to differentiate other stem cells, particularly ES cells, MSCs, MAPCs, MIAMI, hematopoietic stem cells (HSCs). In some cases, the differentiation occurs ex vivo; in some cases the differentiation occurs in vivo.
  • neural stem cells may be generated by culturing the iPSCs as floating aggregates in the presence of noggin, or other bone morphogenetic protein antagonist, see e.g., Itsykson et al. (2005) Mol Cell Neurosci. 30(1):24-36.
  • neural stem cells may be generated by culturing the iPSCs in suspension to form aggregates in the presence of growth factors, e.g., FGF-2, Zhang et al.
  • the aggregates are cultured in serum-free medium containing FGF-2.
  • the iPSCs are co-cultured with a mouse stromal cell line, e.g., PA6 in the presence of serum-free medium comprising FGF-2.
  • the iPSCs are directly transferred to serum-free medium containing FGF-2 to directly induce differentiation.
  • Neural stems derived from the iPSCs may be differentiated into neurons, oligodendrocytes, or astrocytes. Dopaminergic neurons play a central role in Parkinson's Disease and are thus of particular interest.
  • iPSCs may be co-cultured with a PA6 mouse stromal cell line under serum-free conditions, see, e.g., Kawasaki et al. (2000) Neuron 28(1):31-40. Other methods have also been described, see, e.g., Pomp et al. (2005), Stem Cells 23(7):923-30; U.S. Pat. No. 6,395,546.
  • Oligodendrocytes may also be generated from the iPSCs.
  • oligodendrocytes may be generated by co-culturing iPSCs or neural stem cells with stromal cells, e.g., Lee et al. (2000) Nature Biotechnol 18:675-679.
  • oligodendrocytes may be generated by culturing the iPSCs or neural stem cells in the presence of a fusion protein, in which the Interleukin (IL)-6 receptor, or derivative, is linked to the IL-6 cytokine, or derivative thereof.
  • IL Interleukin
  • Astrocytes may also be produced from the iPSCs. Astrocytes may be generated by culturing iPSCs or neural stem cells in the presence of neurogenic medium with bFGF and EGF, see e.g., Housele et al. (1999) Science 285:754-756.
  • Induced cells may be differentiated into pancreatic beta cells by methods known in the art, e.g., Lumelsky et al. (2001) Science 292:1389-1394; Assady et al., (2001) Diabetes 50:1691-1697; D′Amour et al (2006) Nat Biotechnol: 1392-1401′ D'Amour et al. (2005) Nat Biotechnol 23:1534-1541.
  • the method may comprise culturing the iPSCs in serum-free medium supplemented with Activin A, followed by culturing in the presence of serum-free medium supplemented with all-trans retinoic acid, followed by culturing in the presence of serum-free medium supplemented with bFGF and nicotinamide, e.g., Jiang et al. (2007) Cell Res 4:333-444.
  • the method comprises culturing the iPSCs in the presence of serum-free medium, activin A, and Wnt protein from about 0.5 to about 6 days, e.g., about 0.5, 1, 2, 3, 4, 5, 6, days; followed by culturing in the presence of from about 0.1% to about 2%, e.g., 0.2%, FBS and activin A from about 1 to about 4 days, e.g., about 1, 2, 3, 4 days; followed by culturing in the presence of 2% FBS, FGF-10, and KAAD-cyclopamine (keto-N-aminoethylaminocaproyl dihydro cinnamoylcyclopamine and retinoic acid from about 1 to about 5 days, e.g., 1, 2, 3, 4, or 5 days; followed by culturing with 1% B27, gamma secretase inhibitor and extendin-4 from about 1 to about 4 days, e.g., 1, 2, 3, or 4 days; and finally culturing the
  • Hepatic cells or hepatic stem cells may be differentiated from the iPSCs. For example, culturing the iPSCs in the presence of sodium butyrate may generate hepatocytes, see e.g., Rambhatla et al. (2003) Cell Transplant 12:1-11. In another example, hepatocytes may be produced by culturing the iPSCs in serum-free medium in the presence of Activin A, followed by culturing the cells in fibroblast growth factor-4 and bone morphogenetic protein-2, e.g., Cai et al. (2007) Hepatology 45(5):1229-39.
  • the iPSCs are differentiated into hepatic cells or hepatic stem cells by culturing the iPSCs in the presence of Activin A from about 2 to about 6 days, e.g., about 2, about 3, about 4, about 5, or about 6 days, and then culturing the iPSCs in the presence of hepatocyte growth factor (HGF) for from about 5 days to about 10 days, e.g., about 5, about 6, about 7, about 8, about 9, or about 10 days.
  • HGF hepatocyte growth factor
  • the method may also comprise differentiating iPSCs into cardiac muscle cells.
  • the method comprises culturing the iPSCs in the presence of noggin for from about two to about six days, e.g., about 2, about 3, about 4, about 5, or about 6 days, prior to allowing formation of an embryoid body, and culturing the embryoid body for from about 1 week to about 4 weeks, e.g., about 1, about 2, about 3, or about 4 weeks.
  • cardiomyocytes may be generated by culturing the iPSCs may in the presence of LIF, or by subjecting them to other methods in the art to generate cardiomyocytes from ES cells, e.g., Bader et al. (2000) Circ Res 86:787-794, Kehat et al. (2001) J Clin Invest 108:407-414; Mummery et al. (2003) Circulation 107:2733-2740.
  • Examples of methods to generate other cell-types from iPSCs include: (1) culturing iPSCs in the presence of retinoic acid, leukemia inhibitory factor (LIF), thyroid hormone (T3), and insulin in order to generate adipoctyes, e.g., Dani et al. (1997) J. Cell Sci 110:1279-1285; (2) culturing iPSCs in the presence of BMP-2 or BMP-4 to generate chondrocytes, e.g., Kramer et al. (2000) Mech Dev 92:193-205; (3) culturing the iPSCs under conditions to generate smooth muscle, e.g., Yamashita et al.
  • sub-populations of differentiated cells may be purified or isolated.
  • one or more monoclonal antibodies specific to the desired cell type are incubated with the cell population and those bound cells are isolated.
  • the desired subpopulation of cells expresses a reporter gene that is under the control of a cell type specific promoter.
  • the hygromycin B phosphotransferase-EGFP fusion protein is expressed in a cell type specific manner.
  • the method of purifying comprises sorting the cells to select green fluorescent cells and reiterating the sorting as necessary, in order to obtain a population of cells enriched for cells expressing the construct (e.g., hygromycin B phosphotransferase-EGFP) in a cell-type-dependent manner.
  • Selection of desired sub-populations of cells may also be accomplished by negative selection of proliferating cells with the herpes simplex virus thymidine kinase/ganciclovir (HSVtk/GCV) suicide gene system or by positive selection of cells expressing a bicistronic reporter, e.g., Anderson et al. (2007) Mol. Ther. (11):2027-2036.
  • HSVtk/GCV herpes simplex virus thymidine kinase/ganciclovir
  • the methods described herein utilize a panel of iPSC lines or a panel of cells differentiated from iPSC lines.
  • a panel of iPSC lines comprises multiple iPSC lines, e.g., iPSC lines, that meet certain selection criteria.
  • panels of cells differentiated from iPSC lines as described herein include, but are not limited to, panels of neural stem cells, neurons, retinal cells, glial progenitor cells, glial cells, cardiac progenitor cells, cardiomyocytes, pancreatic progenitor cells, pancreatic beta cells, hepatic stem cells, hepatocytes or lung progenitor cells.
  • the selection criteria for inclusion of an iPSC line in a panel of iPSC lines are determined prior to generating the iPSC lines that will constitute the panel. In other cases, the selection criteria are applied to iPSC lines generated before hand, e.g., a bank of iPSC lines.
  • Selection criteria include, but are not limited to, the presence or absence of a particular health condition in an iPSC donor, a positive drug response in an iPSC donor, negative, positive, or adverse drug responses in an iPSC donor, the presence or absence of a particular phenotype in an iPSC line or in cells differentiated from the iPSC line, and the presence or absence of one or more polymorphic alleles in the cell lines or their corresponding donors.
  • the panel includes genetically diverse human iPSC lines in which each iPSC line carries at least one polymorphic allele that is unique among the iPSCs to be included in the panel, e.g., 5 to 10, 20 to 50, 50 to 200, 200 to 500, 500 to 1000, 1000 to 5000, 5000 to 20000, or 20000 to 50000 polymorphic alleles that are unique within the panel of iPSC lines.
  • polymorphic alleles may include, e.g., a SNP allele, a promoter allele, or a protein-encoding allele.
  • Polymorphic alleles can be screened and scored for by genotyping using any of a number of known genotyping assays.
  • the genotyping assay is a multiplexed genotyping assay, e.g., a nucleic acid microarray assay platform such as a “SNP chip.”
  • the one or more polymorphic alleles are pre-selected.
  • the one or more preselected alleles are polymorphic alleles associated with a health condition or a predisposition to a health condition.
  • polymorphic alleles associated with a health condition or a predisposition to a health condition include, but are not limited to, polymorphic alleles associated with a neurodegenerative disorder, a neurological disorder, an eye disease, a mood disorder, a respiratory disease, a cardiovascular disease, an immunological disorder, a hematological disease, a metabolic disorder, or a drug sensitivity condition.
  • polymorphic alleles associated with a health condition are provided in Table 3 above.
  • Polymorphic alleles may include polymorphic alleles in an encoded protein or a regulatory sequence affecting the expression of the encoded protein.
  • the encoded protein is a drug target. Examples of drug target proteins include, but are not limited to, GPCRs, ion channels, kinases, enzymes, and transcription factors.
  • the one or more polymorphic alleles are pre-selected based on the presence of a high degree of surrounding linkage disequilibrium in the genome, which has been proposed as a signature of genomic loci that are likely to impact many common health conditions.
  • Methods for identifying SNPs having a high surrounding linkage disequilibrium and genes near such SNPs are described in, e.g., Wang et al (2006), Proc Natl Acad Sci USA, 103(1):135-140.
  • a panel of iPSC lines includes iPSC lines generated from subjects that are diagnosed as suffering from one or more health conditions.
  • the one or more health conditions may be one or more health conditions that are common to all of the iPSC donors, or they may be health conditions that are different between the iPSC donors.
  • a panel of iPSC lines includes iPSC lines generated from subjects that are both diagnosed as suffering from a health condition and carry a polymorphic allele associated with a health condition, e.g., a polymorphic allele associated with the diagnosed health condition.
  • a panel of iPSC lines may include iPSC lines from at least about 10 individuals to at least about 50,000 individuals, e.g., 10 to 50, 20 to 100, 50 to 250, 100 to 1000, 250, to 2000, 500 to 5000, 1000 to 10,000, 2500 to 20,000, 10,000, to 30,000, 20,000 to 40,000, or 30,000 to 50,000 individuals.
  • a panel of iPSC lines may include iPSC lines from at least two ethnic groups, e.g., 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, or 50 ethnic groups.
  • ethnic groups include, but are not limited to, Europeans, Japanese, Chinese, and the Yoruba of Nigeria, and ethnic groups listed in Table 4.
  • iPSC lines and panels of iPSC lines described herein are useful in a number of methods relating to drug discovery and development.
  • a drug candidate compound will be evaluated in a biochemical assay (e.g., a receptor binding assay) that evaluates only a single or very few sequence variants of the drug target expressed in a patient population.
  • a biochemical assay e.g., a receptor binding assay
  • assays provide little information as to how effective the drug candidate compound is likely to be in patients that express a drug target allele that differs from the particular drug target allele that was originally screened.
  • drug candidate compounds often undergo functional cellular screens in one or few cell lines engineered to express a specific allele of the drug target, again ignoring the genetic diversity of a human patient population not only with respect to the drug target itself, but also to that of the various downstream signal transduction proteins that play a role in the response endpoint of cells to a drug.
  • adverse effects of candidate drug compounds e.g., liver toxicity
  • drug screening in panels of genetically diverse iPSC lines, as described herein addresses the lack of genetic diversity in the prevailing drug screening models.
  • the panels of genetically diverse iPSC lines described herein may be used to identify test compounds that act on a drug target of interest.
  • the panels of iPSCs cell lines include a sufficient number of iPSC lines such that at least two, e.g., at least 3, 5, 10, 20, 50, 100, or 200 polymorphic alleles of a drug target (e.g., a GPCR, ion channel, or kinase) are represented in the panel.
  • panels of iPSC lines are derived from subjects diagnosed as suffering from a health condition or identified as having a predisposition to the health condition.
  • the iPSC line panels comprise iPSC lines each of which that has at least one polymorphic allele associated with a health condition or a predisposition to the health condition.
  • Drug targets for many health conditions are known.
  • Such drug targets may include, but are not limited to, receptors, GPCRs, growth factor receptors, neurotransmitter receptors, ion channels, enzymes, protein kinases, proteases, cytoskeletal proteins, and transcription factors.
  • Test compounds can be assayed for their effect on a drug target by a number of assays known in the art.
  • assays include cell-based assays including, but not limited to, assays for determining second messenger levels, e.g., intracellular calcium, cAMP, cGMP, arachidonic acid, and inositol phosphates; channel currents; apoptosis; proliferation; morphological changes; changes in adhesion.
  • Cell based assays may also include determining the cellular localization of one or more proteins (e.g., protein kinases, receptors, and transcription factors) in cells in the presence or absence of a test compound. Test compounds may also be screened for their ability to alter a gene expression profile by any gene expression profiling method known in the art. In some cases, the cells to be screened may be genetically modified to express one or more reporter proteins that can indicate activation of a signaling pathway. For example protein-protein interactions between fusion proteins introduced into cells may be detected by a number of methods known in the art, e.g., by fluorescence resonance energy transfer (FRET) or enzyme fragment complementation.
  • FRET fluorescence resonance energy transfer
  • the mechanistic basis of a sporadic form of a disease is a combination of genetically-determined cell type-specific phenotype and epigenetic factors (e.g., oxidative stress).
  • epigenetic factors e.g., oxidative stress.
  • iPSC-derived differentiated cells from a patient with a sporadic form of a disease may bear a genetic predisposition to a pathological or pre-pathological cellular phenotype (e.g., apoptosis), but the phenotype may only manifest in vitro in the presence of an appropriate “stressor” that recapitulates environmental/epigenetic factors associated with the sporadic disease or a cellular phenotypes that precede a clinical manifestation of the disease (e.g., mitochondrial dysfunction, oxdidative stress, or nitrosylative stress).
  • disease-relevant cellular phenotypes are induced by a stressor.
  • stressors include, but are not limited to cellular oxidative stress, nitrosylative stress, proteasome inhibition, inhibition of mitochondrial electron transport, translation inhibition, decreased calcium buffering, high osmolarity, heat shock, heavy metals (e.g., Zn, Mn, Fe, Cd, Al, or Pb), protein misfolding.
  • agents that induce, increase, or result from oxidative stress include, but are not limited to, H 2 O 2 , ascorbic acid/FeSO 4 , 4-hydroxynonenal, glutamate, kainate, NMDA, dopamine, okadaic acid, A ⁇ 1-42 and isocyanate.
  • Proteasome inhibitors include, but are not limited to lactacystin, ziram, MG132, and carbobenzoxy-L-isoleucyl-gamma-t-butyl-L-glutamyl-L-alanyl-L-leucinal (PSI).
  • Mitochondrial stressors include, but are not limited to, rotenone, 3-nitropropionic acid (NPA), 1-methyl-4-phenylpyridinium (MPP + ), antimycin, paraquat, methylglycoxal, and cyanide.
  • Nitrosylative stressors include, but are not limited to, (+/ ⁇ )-S-nitroso-N-acetylpenicillamine, sodium nitroprussiate, and peroxynitrite.
  • the stressor is provided by expressing or overexpressing an exogenous wild type or mutated gene and/or protein.
  • genes include, ⁇ -synuclein, amyloid beta, A ⁇ 1-42 , Parkin, Pink1, Leucine-rich repeat kinase 2 (LRRK2), superoxide dismutase (SOD).
  • Assays of drug candidate compounds in an iPSC line or a panel of iPSC lines can include determining a dose-response.
  • the dose response of an iPSC line or that of one or more types of cells differentiated from the iPSC line provides an indication that of the likely efficacy of the compound in the corresponding iPSC donor.
  • the fraction of iPSC lines in a panel of iPSC lines that exhibit an acceptable dose-response to a test compound indicates an expected probability of an acceptable dose-response relationship in the target patient population of interest.
  • cell-based assays of drug candidate include a comparison of responses obtained in a panel of iPSC lines or iPSC-derived cells to one or more reference iPSC lines or cells that serve as a positive or negative control for the effect of a drug candidate compound.
  • the reference iPSC lines or cells may be from a healthy iPSC donor, from an iPSC donor diagnosed as suffering from a health condition, or an iPSC donor carrying a polymorphic allele associated with a health condition.
  • assays of drug candidate compounds in an iPSC line or a panel of iPSC lines can include determining effective concentrations, maximum tolerated dose and minimum effective concentration. Additional methods and assays are disclosed in U.S. application No. 61/061,594, WSGR Docket Number 36588-707.101; filed Jun. 13, 2008; First Inventor Kazuhiro Sakurada, hereby incorporated by reference.
  • the drug screening may be conducted on cells differentiated from iPSCs.
  • differentiated cells are described herein (e.g., hepatic cells, neural stem cells, neurons, pancreatic beta cells, cardiomyocytes, hepatic stem cells, oligodendrocytes).
  • the drugs may be targeted to treat a specific disease or condition, e.g., a disease or condition described herein.
  • the iPSCs may be differentiated into dopaminergic neurons, which are used to screen drugs for Parkinson's disease.
  • neurons or neural stem cells differentiated from iPSCs may be used to screen drugs for treating Alzheimer's disease, multiple sclerosis, or other neurological disorders.
  • the iPSCs may be transplanted directly into an immunocompromised animal, e.g., SCID mouse, which is then used to establish in vitro or in vivo assay systems that mimic physiologic conditions in humans or other animals.
  • the in vitro or in vivo assay systems may be used to screen for drugs, e.g., drugs for Parkinson's disease, or as a means to identify biological mechanisms.
  • test compounds may also be conducted in iPSC-derived cells when an abnormal cellular phenotype (e.g., abnormal cell morphology, gene expression, or signaling), associated with a health condition or a predisposition to the health condition is known, but a drug target has not yet been identified.
  • abnormal cellular phenotype e.g., abnormal cell morphology, gene expression, or signaling
  • Such assays may include contacting a test population of iPSC-derived cells from one or more iPSC donors with a test compound and contacting with a negative control compound a negative control population of iPSC-derived cells from the same one or more iPSC donors.
  • the assayed cellular phenotype associated with the health condition of interest in the test and negative control populations can then be compared to a normal cellular phenotype.
  • the drug candidate compound is identified as normalizing the phenotype.
  • a normal cellular phenotype with respect to a particular health condition or a predisposition for a health condition may be established in iPSC-derived cells from iPSC donors that do not suffer from the health condition or a predisposition for the health condition.
  • Test compounds identified as lead compounds may be tested on a panel of iPSC-derived cells in a manner analogous to a clinical trial.
  • the efficacy of the lead compound versus a negative control compound, e.g., a placebo compound is determined in a panel of iPSC-derived cells from patients suffering from the same health condition.
  • a panel of iPSC-derived cells is from subjects that are genetically diverse.
  • such patients may be carry at least one polymorphic allele that is unique among the iPSC-derived cells to be included in the panel, e.g., 5 to 10, 20 to 50, 50 to 200, 200 to 500, 500 to 1000, 1000 to 5000, 5000 to 20000, or 20000 to 50000 polymorphic alleles that are unique within the panel of iPSC lines.
  • a number of methods for quantifying the genetic diversity of a population are known in the art, e.g., the analysis of molecular variance (AMOVA) and generalized analysis of molecular variance (GAMOVA).
  • the efficacy of the lead compound in iPSC-derived cells may be determined based on any cellular response endpoint, e.g., a response obtained in any of the cell-based assays or gene expression profiling assays mentioned herein.
  • iPSC-derived cells may include any cell type that hepatocytes, cardiomyocytes, neurons,
  • Drug candidate compounds may be individual small molecules of choice (e.g., a lead compound from a previous drug screen) or in some cases, the drug candidate compounds to be screened come from a combinatorial library, i.e., a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical “building blocks.”
  • a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks.
  • Combinatorial chemical libraries include, but are not limited to: diversomers such as hydantoins, benzodiazepines, and dipeptides, as described in, e.g., Hobbs et al. (1993), Proc. Natl. Acad. Sci. U.S.A.
  • iPSC cell lines and iPSC-derived cells generated from a subject can be used to determine the likelihood that a particular drug will have sufficient efficacy in that subject and, if so, an appropriate dose range for that subject.
  • a subject e.g., a human subject
  • iPSC-derived cells from a subject e.g., differentiated iPSC-derived cells may be exposed ex vivo to a drug to be tested, and then assayed for their phenotypic response to the drug as described herein.
  • the response of the iPSC-derived cells may be compared to a reference response obtained in iPSC-derived cells from one or more individuals in which the drug has been shown to be effective and/or a reference response in iPSC-derived cells from subjects in which the drug was found to be ineffective.
  • the subject to be tested is a subject suffering from a health condition or a predisposition to the health condition. For example, where the subject is suffering from a health condition, and multiple drugs are available to treat the health condition, the efficacies and adverse effects of the multiple drugs may be evaluated iPSC-derived cells from that individual.
  • the iPSC-derived cells used to test drug efficacy include cells that express at least one drug target (e.g., a neurotransmitter receptor).
  • the subject is not suffering from a health condition.
  • drugs for various health conditions are tested preemptively in iPSC-derived cells from a healthy subject to establish a pharmaco-phenomic profile for that subject. The pharmaco-phenomic profile may subsequently be used as needed for selecting optimal drugs and drug dosing for treatment of the particular subject.
  • Candidate therapeutic agents are those that normalize a disease-associated cellular phenotype, i.e., alter the relevant cellular phenotype in the patient-derived cells so that it is closer to the corresponding cellular phenotype in cells derived from normal subjects under the same conditions.
  • the therapeutic agent may alter a cellular phenotype of ⁇ patient-derived iPSCs so as to protect them from a stressor, as described herein.
  • cellular phenotypes e.g., mitochondrial ROS production, expression profiles, protein aggregation
  • cellular phenotypes distinguishing patient versus normal subject can be identified, for example, by means of univariate statistical methods, such as t-test, ANOVA, regression, as well as their non-parametric analogs.
  • cellular phenotype data are further filtered using various statistical criteria, e.g., p-value of significance (Type1 error), effect size, etc.
  • disease-relevant cellular phenotypes that are found to differ significantly in patient versus normal subjects (i.e., disease-relevant cellular phenotypes) need to be validated by means of orthogonal assays.
  • the identified disease-relevant cellular phenotypes are confirmed by performing validation/cross-validation analysis on the independent data sets from the same type of cellular phenotype assays.
  • disease-relevant cellular phenotypes are determined by first assaying and analyzing only a portion of the available patient and normal iPSC lines, and then validating disease-relevant cellular phenotypes in the remaining iPSC lines.
  • one or more validated cellular phenotypes is then used to assess test agents for their ability to convert one or more cellular phenotypes reflecting a disease condition to cellular phenotypes reflecting a normal condition.
  • a “wellderly” subject refers to any subject that is at least 80 years old and has not suffered from any major chronic diseases. Selection of wellderly individuals as normal control subjects makes it statistically less likely that such individuals will go on to develop a degenerative condition. Thus, iPSCs derived from such individuals are less likely to exhibit a cellular phenotype that is associated or predictive of the disease being analyzed, and therefore provide a more reliable “normal control” phenotype for purposes of comparison to patient-derived iPSCs and iPSC-derived cells. In other embodiments, elderly individuals are selected for control iPSC generation that while not having suffered from a degenerative disease under study, may have suffered other unrelated degenerative diseases. In other cases, age-matched subjects free of the disease to be analyzed are used to generate normal control subject iPSCs.
  • efficacy is tested in larger panels of patient iPSC-derived cells to identify potential variation in efficacy or toxicity of the candidate therapeutic agent.
  • efficacy is tested in iPSCs or iPSC-derived cells from at least about 20 to about 500 patients, e.g., at least about 25, 30, 40, 50, 60, 70, 100, 200, 250, 300, 400, or another number of patients from at least about 20 to about 500 patients.
  • biomarkers associated with responsiveness to a candidate therapeutic agent or lack of responsiveness to a candidate therapeutic agent are identified and used to stratify a patient population into, e.g., “high responders” (HR) and “low responders” (LR), as schematized in FIG. 4 .
  • HR high responders
  • LR low responders
  • biomarkers are used to identify suitable patients for clinical trials of a candidate therapeutic agent.
  • biomarkers are used to predict the responsiveness or potential toxicity of a therapeutic agent for particular patients.
  • biomarkers include genomic biomarkers (e.g., SNPs, a CNVs, or other genetic polymorphisms).
  • the biomarkers include an expression profile signature (e.g., an mRNA expression profile).
  • the biomarkers may include a protein expression profile or even a single protein expression level. In some cases, where the biomarkers are expression profile biomarkers, these may be determined directly from a patient sample (e.g., blood, urine, sputum, hair, skin, or other biological sample taken directly from the patient). In other embodiments, expression profile biomarkers, are specific to patient iPSCs or iPSC-derived cells in which the candidate therapeutic agent or therapeutic agent is tested.
  • iPSCs are derived from patients and control subjects, and the iPSCs are differentiated into disease-relevant cell types thereby allowing a comparison of cellular phenotypes in patient-derived cells versus normal subject derived cells.
  • the cellular phenotype that is compared may include a mitochondrial phenotype, e.g., ATP synthesis, ATP/ADP ratio, mitochondrial potential, calcium buffering, production of reactive oxygen species, mitochondrial fusion and fission, mitochondrial morphology, and mitochondrial movement.
  • the cellular phenotype that is compared is the fraction and rate at which a particular cell type is undergoing apoptosis in the presence of a stressor.
  • the cellular phenotype is protein aggregation (e.g., the formation of lewy bodies).
  • gene expression e.g., microRNA expression is compared between patient-derived cells and normal subjects.
  • relational databases are constructed that integrate multiple data streams relating to each patient and control iPSC line.
  • data include, but are not limited to, one or more of the following: patient medical history and family medical history, patient medical data (e.g., blood pressure, liver enzyme levels), patient adverse drug reactions, patient drug responsiveness, partial or complete genomic sequence, sequence of all genes with known disease-associated alleles, comprehensive SNP genotypes (e.g., genotypes for all SNPs with known disease associations), gene copy number variation (CNV) polymorphisms, expression profiles for iPSCs and for cells differentiated from the iPSCs (e.g., dopaminergic neurons, cortical neurons, motor neurons, pancreatic cells, hepatocytes, cardiomyocytes, and vascular epithelial cells) under resting and under various stimulus paradigms (e.g., in the presence of a stressor), all cellular phenotype assay data used for initial pathway discovery and for drug screening, including, e.g., cellular
  • a user can query such a database based on any set of criteria with user define limits. For example, a user may wish to identify polymorphisms associated with patients whose iPSC-derived dopaminergic neurons did or did not respond to a candidate therapeutic agent. In another example, a user may wish to identify a common gene expression profile that distinguishes motor neurons that showed a severe apoptotic response to a stressor versus a mild apoptotic response, etc. Such databases are very useful for data mining and establishing robustly predictive signatures for specific disease states and their response to candidate therapeutic agents.
  • SMA Spinal Muscular Atrophy
  • SMA Survival Motor Neuron
  • the region of chromosome 5 that contains the SMN (survival motor neuron) gene has a large duplication. A large sequence that contains several genes occurs twice—i.e. once in each of the adjacent segments. The two copies of the gene—known as SMN1 and SMN2-differ by only a few base pairs.
  • the SMN2 gene contains a mutation that occurs at the splice junction of intron 6 to exon 7 resulting in about 90% of SMN2 pre-mRNA transcripts being spliced into a form that excludes exon 7. This shorter mRNA transcript codes for a truncated SMN protein, which is rapidly degraded.
  • SMA results from the loss of the SMN1 gene from both chromosomes, and its severity, ranging from SMA 1 to SMA 3, largely depends on whether the level of SMN2 E7 transcript can make up for low levels or absence of exon 7-inclusive SMN 1 transcript.
  • the mutations that cause the loss of SMN 1 are of two types. Deletion mutations, in which both copies of the SMN1 are missing. The other type of mutation is a conversion mutation in which both copies of the SMN1 gene have a point mutation resulting in the same splicing pattern as the SMN2 gene.
  • SMN2 E7 exon 7-inclusive SMN2 transcript
  • Induction of iPSCs was initiated by transduction of SMN1 ⁇ / ⁇ and SMN1 ⁇ /+ fibroblast cultures with four MoMLV VSV-G-pseudotyped viruses for expression of human OCT4, SOX2, KLF4, and c-MYC, each at an MOI of about 10.
  • fibroblasts were switched from human fibroblast medium into human ES cell supportive medium and monitored daily for the appearance of putative iPSC colonies based on morphological criteria.
  • the ability of the SM8c line to differentiate into ectodermal, mesodermal, and endodermal lineages in vitro was confirmed by immunostaining for the ectodermal marker Tun, the mesodermal marker Desmin, and the endodermal marker AFP, as shown in FIG. 7 . Further, the SM8c iPSC line was shown to differentiate into mature motor neurons as shown by double immunolabeling for Islet and Neuro-N (data not shown).
  • iPSCs can be generated from SMA patients and differentiated into motor neurons, as required for the screening assay described in Example 2.
  • SMA patient-specific motor neurons are obtained by first generating panels of iPS cell lines from Type I, Type II, and Type III SMA patients, as described in Example 1, and subsequently differentiating iPSCs into motor neurons. Prior to motor neuron differentiation SMA patient SMN2 minigene reporter iPSC lines are established to provide a convenient readout for the level of SMN2 E7 transcript in motor neurons.
  • standard dermal punch biopsies 2-4 mm in diameter and thickness are obtained from approximately 30 Type I, 30 Type II, and 30 Type III SMA patients, all of whom have an SMA1 ⁇ / ⁇ genotype, and 10 healthy, age-matched control subjects that have an SMA1 ⁇ /+ genotype.
  • iPSC line database For each SMA-iPSC line to be generated, the following corresponding patient information is collected and annotated in an iPSC line database: disease severity ranking (i.e., Type I, II, or III), age of disease onset, patient medical history, family medical history including incidence of ALS, blood level of SMN protein, SMN1 and SMN2 genotypes, MUNE Motor Unit Number Estimation, Hammersmith SMA Functional Motor Scale ranking, breathing test evaluation (only for children >5 yrs), symptom progression evaluation (e.g., how outcome of motor tests has changed over time), muscle mass index, description of therapeutic interventions to date, and therapy response.
  • disease severity ranking i.e., Type I, II, or III
  • age of disease onset i.e., patient medical history, family medical history including incidence of ALS, blood level of SMN protein, SMN1 and SMN2 genotypes
  • MUNE Motor Unit Number Estimation Hammersmith SMA Functional Motor Scale ranking
  • breathing test evaluation only
  • Additional data may be added to each record as they are acquired, including, e.g., SMN protein levels and SMN2 E7 transcript levels under various experimental conditions (e.g., in the presence or absence of a candidate therapeutic compound), informative SNP genotypes, genomic sequence, and tissue/cell-type specific expression profiles.
  • Biopsy samples are stored for up to 5-7 days at 4° C. in a “biopsy medium” containing KO-DMEM and supplemented with 10% fetal bovine serum (FBS), Earl's Salts, nucleosides, beta-mercaptoethanol (BME), non-essential amino acids, glutamine, and penicillin/streptomycin.
  • Biopsies are minced into 4-5 pieces, and the pieces are then transferred to a 60 mm dish. The pieces are then “sandwiched” under an acid-washed coverslip and cultured in biopsy medium for five days.
  • the sandwiched biopsy explants are cultured in human fibroblast (“hFib”) medium containing KO-DMEM, Earl's Salts, 10% FBS, glutamine, penicillin/streptomycin, and medium is replaced every 3-4 days until the coverslip is confluent.
  • hFib human fibroblast
  • SMA iPSCs are generated, as described in Example 1, from fibroblasts obtained from each biopsy.
  • An SMN2 splicing minigene reporter construct is generated that incorporates exons 6, 7, and 8, and utilizing the SMN2 promoter is generated essentially as described in Zhang et al (2001), Gene Ther., 8:1532-1538 and Wilson et al, Stem Cells and Development, 16:1027-1041.
  • the SMN2 reporter construct will incorporate the DD-AmCyan1 fluorescent protein reporter to maximize the signal to noise ratio in a compound screening reporter gene assay.
  • the DD-AmCyan1 protein contains a degradation (“DD”) domain that conditionally destabilizes the protein thereby keeping “background” levels of the reporter protein prior to a test compound screening assay very low.
  • the reporter protein upon addition of the cell-permeable “Shield1” ligand (Invitrogen), which selectively binds to the DD domain, the reporter protein is stabilized and can therefore accumulate.
  • DD-AmCyan1 reporter levels in the presence or absence of test compounds are maximized by measuring almost exclusively reporter protein produced after the beginning of the screening assay, i.e., after the addition of the Shield1 ligand and test compound.
  • Additional reporter constructs will include AmCyan1 or luciferase as the reporter.
  • Other constructs will include the CMV promoter to drive SMN2 minigene expression.
  • the SMN2 reporter construct is then stably transfected into type I, type II, and type III SMA-iPSCs and healthy control (SMN1 WT/WT ) iPSCs to generate SMN2-reporter SMA-iPSC lines of varying disease severity backgrounds, and SMN2 reporter control iPSC lines, respectively.
  • Primary screening of test compounds for the ability to increase properly spliced SMN2 transcript levels is conducted initially in motor neurons derived from Type I SMA reporter iPSCs.
  • SMN2 reporter SMA-iPSCs are trypsinized and then washed/resuspended in embryoid body (EB) medium containing KO DMEM (Invitrogen, catalog #10829-018), Knockout Serum Replacement (Invitrogen, catalog# A1099202), Plasmanate (Talecris), Glutamax (Invitrogen, catalog# 35050079), non-essential amino acids (Invitrogen, cat#11140050).
  • EB embryoid body
  • KO DMEM Invitrogen, catalog #10829-018
  • Knockout Serum Replacement Invitrogen, catalog# A1099202
  • Plasmanate Plasmanate
  • Glutamax Invitrogen, catalog# 35050079
  • non-essential amino acids Invitrogen, cat#11140050
  • N2 base medium DMEM/F12, Glutamax (Invitrogen, catalog#10565), N-2 Supplement (Invitrogen, catalog#17502-048), D-Glucose (Sigma, catalog # G8769), Ascorbic Acid (Sigma, catalog# A4403-100mG)
  • RA Retinoic Acid
  • PM Purmorphamine
  • RA (1 ⁇ M)/PM (1 ⁇ M)-supplemented EB medium is replaced every 3-5 days, as needed, until approximately day 28. Afterwards, EBs are dissociated by dilute papain treatment and gentle trituration, and then replated on new ULF 6-well plates followed by gentle trituration every 10 minutes over a period of 45 minutes.
  • the resulting cell suspension is collected and transferred to a 50 ml conical tube containing motor neuron maturation medium (DMEM/F12, Glutamax, N-2 Supplement (Invitrogen, catalog#17502-048), B-27 Supplement (Invitrogen, catalog# 17504-044), D-Glucose, Ascorbic Acid (Sigma, catalog# A4403-100mG), 2 ng/mL each GDNF (R&D, catalog#212-GD), BDNF (R&D, catalog#248-BD), and CNTF (R&D, catalog#257-NT/CF).
  • motor neuron maturation medium DMEM/F12, Glutamax, N-2 Supplement (Invitrogen, catalog#17502-048), B-27 Supplement (Invitrogen, catalog# 17504-044), D-Glucose, Ascorbic Acid (Sigma, catalog# A4403-100mG), 2 ng/mL each GDNF (R&D, catalog#212-GD), BDNF (R&
  • the cell suspension is pelleted by centrifugation at 1000 RPM for five minutes, and is then resuspended in motor neuron maturation medium at a cell concentration of approximately 1.6 ⁇ 10 6 cells/ml. Aliquots (50 ⁇ l) of cell suspension are then plated on laminin-coated wells of optical grade 96 well plates. Beginning on day 31, half-medium changes are conducted every other day or every day depending on how quickly the medium becomes spent. The differentiated cultures are maintained in motor neuron differentiation medium for another four weeks prior to beginning SMN2-reporter assays to allow expansion and maturation of the motor neuron population.
  • Candidate therapeutic compounds are screened in secondary assays for their ability to increase SMN2 E7 transcript levels and for their ability to promote SMA motor neuron survival over a period of about two weeks. Candidate therapeutic compounds are then tested on motor neurons derived from additional type I SMA SMN2-reporter iPSC lines, and from type II and type III iPSC lines to validate the effect of the therapeutic candidate compounds on motor neurons from diverse genetic backgrounds extant in the SMA patient population.
  • Parkinson's Disease is one of the most common neurodegenerative diseases of aging, affecting 1-2% of the population over 65 years of age. Clinical symptoms include rest tremor, bradykinesia, and rigidity. We seek to generate a PD patient iPSC model to identify candidate therapeutic agents that slow, halt, or reverse PD progression.
  • iPSC lines are generated from skin biopsies obtained from 10 healthy control subjects with no known family history of PD, 10 patients with sporadic PD, patients each with mutations in the genes that encode ⁇ -synuclein (PARK1), parkin (PARK2), PINK 1 (PARK6), or LRRK2 (PARKS) for a total of 10 patients for each mutation.
  • iPSCs are generated as described in Example 1. Afterwards, dopaminergic neurons are derived by differentiating each of the patient iPSC lines and control subject iPSCs.
  • a dopaminergic phenotype is established by immunocytochemical staining for tyrosine hydroxylase positivity, and assaying the differentiated cells for the ability to synthesize and release dopamine.
  • Dopaminergic neurons are obtained by differentiating the iPSCs according to the method of Perrier et al (2004), Proc Natl Acad Sci USA 101, 12543-12548. After validating the dopaminergic phenotype of neurons differentiated from each of the above iPSC lines, cultures of the patient iPSC-derived dopaminergic neurons are tested in a battery of cellular phenotype assays and compared to control subject dopaminergic neurons.
  • one of more of the PD-associated cellular phenotypes is used as the basis of a screen for candidate therapeutic agents that can reverse or ameliorate these cellular phenotypes. Further, it is expected that as the PD cellular phenotypes are identified in disease relevant cells (dopaminergic neurons) from human PD patients, their predictive value and reliability for the development of therapeutic agents will be more robust than those based on heterologous assay models.

Abstract

The present invention involves methods for identifying an agent that corrects a phenotype associated with a health condition or a predisposition for a health condition. The invention also involves methods for identifying a diagnostic cellular phenotype, determining the risk of a health condition in a subject, methods for reducing the risk of drug toxicity in a human subject, and methods for identifying a candidate gene that contributes to a human disease. The invention also discloses human induced pluripotent stem cell lines.

Description

    CROSS-REFERENCE
  • This application is a continuation of U.S. application Ser. No. 12/484,163, filed Jun. 12, 2009, which is a continuation-in-part of U.S. application Ser. No. 12/157,967, filed Jun. 13, 2008, which claims the benefit of U.S. Provisional Application No. 61/040,646, filed Mar. 28, 2008, and which also claims the benefit of International Application No. PCT/EP2007/010019, filed Nov. 20, 2007, and which also claims the benefit of Japanese Application No. JPO-2007-159382, filed Jun. 15, 2007; this application also claims the benefit of International Application No. PCT/IB2008/002540, filed Jun. 13, 2008, International Application No. PCT/EP2008/005047, filed Jun. 13, 2008, U.S. Provisional Application No. 61/061,592, filed Jun. 13, 2008, and U.S. Provisional Application No. 61/061,594, filed Jun. 13, 2008, all of which are herein incorporated by reference in their entirety.
  • BACKGROUND OF THE INVENTION
  • The pharmaceutical industry has expended vast technical and financial resources to develop novel therapeutic agents. Yet, the failure rate (more than 90%) for lead compounds remains persistently high. Often, lead drug compounds that meet expectations in preclinical models, such as inbred animal models, or a small number of cell lines, are toxic or ineffective when administered to a human clinical trial patient population. A fundamental deficiency in most current drug development efforts is that they do not evaluate candidate drug efficacy and toxicity in the context of the extreme genetic diversity of the human patient population. In other words, in the present drug development paradigm, drug efficacy and toxicity are not tested on many, if not most, of the relevant genotype/phenotype combinations present in the human population. Indeed, even after successful trials in a relatively small human clinical trial population, unexpected adverse effects can be revealed once these drugs are administered to a broader human patient population.
  • SUMMARY OF THE INVENTION
  • The present invention involves methods for identifying an agent that corrects a phenotype associated with a health condition or a predisposition for a health condition comprising contacting a first population of cells from a human induced pluripotent stem cell line, or cells differentiated from the human induced pluripotent stem cell line, with a candidate agent; contacting a second population of cells from a human induced pluripotent stem cell line, or cells differentiated from the human induced pluripotent stem cell line, with a control agent; wherein the cells in both populations comprise at least one endogenous allele associated with the health condition or predisposition for the health condition; assaying the two populations and identifying candidate agents as correcting the phenotype if the first population is closer to a normal phenotype following treatment than the second population. The condition may be selected from health conditions such as a neurodegenerative disorder, a neurological disorder, a mood disorder, a cardiovascular disease, a metabolic disorder, a respiratory disease, a drug sensitivity condition, an eye disease, an immunological disorder, or a hematological disease. The cells may be differentiated from induced stem cells to neural stem cells, neurons, cardiomyocytes, hepatic stem cells, or hepatocytes. The phenotype described may be apoptosis, intracellular calcium level, calcium flux, protein kinase activity, enzyme activity, cell morphology, receptor activation, protein trafficking, intracellular protein aggregation, organellar composition, motility, intercellular communication, protein expression, or gene expression.
  • The invention also involves methods for identifying a diagnostic cellular phenotype comprising comparing a set of cells from a subject to cells from a subject free of the health condition wherein both sets of cells were induced pluripotent stem cells, or were cells differentiated from induced pluripotent stem cells, and wherein the comparison is performed on a computer. The cells may be differentiated from induced stem cells to neural stem cells, neurons, cardiomyocytes, hepatic stem cells, or hepatocytes.
  • The invention also involves methods for determining the risk of a health condition in a subject comprising comparing at least one phenotype determined in a first set of cells derived from the subject to the at least one phenotype determined in a second set of cells derived from subjects free of the health condition and to the at least one phenotype determined in a third set of cells derived from subjects suffering from the health condition; and indicating that the subject is at high risk for the health condition if the at least one phenotype determined in the first set of cells is more similar to the at least one phenotype determined in the third set of cells than the at least one phenotype determined in the second set of cells, wherein the first, second, and third sets of cells were induced pluripotent stem cells, or were cells differentiated from induced pluripotent stem cells, and wherein the comparison is performed on a computer.
  • The invention also involves methods for reducing the risk of drug toxicity in a human subject, comprising contacting one or more cells differentiated from an induced pluripotent stem cell line generated from the subject with a dose of a pharmacological agent, assaying the contacted one or more differentiated cells for toxicity, and prescribing or administering the pharmacological agent to the subject if, and only if, the assay is negative for toxicity in the contacted cells. The cells differentiated from the induced pluripotent stem cell line may be hepatocytes, cardiomyocytes, or neurons.
  • The invention also involves methods for identifying a candidate gene that contributes to a human disease, comprising comparing a global gene expression profile of cultured human cells of a differentiated cell type from a plurality of healthy individuals to a global gene expression profile of cultured human cells of the differentiated cell type from a plurality of individuals suffering from the human disease and identifying one or more genes that have different expression levels as candidate genes that contribute to the human disease, wherein the comparison is performed on a computer.
  • The invention also discloses a human induced pluripotent stem cell line generated from a subject diagnosed as suffering from a health condition, or comprising at least one endogenous allele associated with a health condition or a predisposition for the health condition. The invention also discloses an isolated population of human cells comprising neural stem cells or neurons from a subject having at least one endogenous allele associated with a neurodegenerative disorder, a neurological disorder, or a mood disorder, or from a subject diagnosed with the neurodegenerative disorder, neurological disorder, or mood disorder. The invention also discloses an isolated population of human cells comprising human cardiac progenitor cells or cardiomyocytes from a subject having at least one endogenous allele associated with a cardiovascular disease, or from a subject diagnosed with the cardiovascular disease. The invention also discloses an isolated population of human cells comprising hepatic stem cells or hepatocytes from a subject having at least one endogenous allele associated with a drug sensitivity condition, or from a subject diagnosed with the drug sensitivity condition.
  • The invention further discloses a panel of genetically diverse human induced pluripotent stem cell lines, comprising human induced pluripotent stem cell lines generated from a plurality of individuals each of which carry at least one polymorphic allele that is unique among the plurality of individuals.
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, and patent application was specifically and individually indicated to be incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
  • FIG. 1 is a schematic comparison of a traditional drug discovery scheme (left) in which lead compounds are tested against a disease target in heterologous systems (e.g., animal models) prior to testing compound efficacy and safety in patients versus a new drug discovery paradigm (right) in which lead compounds are first identified based on their efficacy in correcting a disease-relevant cellular phenotype in patient-derived, disease-relevant cell types.
  • FIG. 2 is an overview of an exemplary, non-limiting, scheme for patient iPSC-based disease modeling and drug discovery.
  • FIG. 3 is an overview of an exemplary, non-limiting, scheme for patient iPSC-based testing of lead drug candidate efficacy and safety in cells from a genetically diverse cohort of patient iPSC lines.
  • FIG. 4 is an overview of an exemplary, non-limiting, scheme for patient iPSC-based identification of predictive biomarkers for drug efficacy and toxicity. Such biomarkers are used in, e.g., patient stratification for clinical trials of drug candidates, and also for optimal dosing and safety of approved therapeutics in specific patients or patient populations, which is sometimes referred to as “personalized medicine.”
  • FIG. 5 (Top Panel) shows photomicrographs of fibroblasts from three SMN1−/− SMA patients and two SMN1−/+ healthy control subjects; (Bottom Panel) shows photomicrographs of iPSC colonies derived from the corresponding SMA case and control subject fibroblasts illustrated in the top panel.
  • FIG. 6 shows photomicrographs of embryoid bodies obtained from the SMA case and control iPSC lines shown in FIG. 5.
  • FIG. 7 shows immunofluorescence photomicrographs of staining for ectodermal (TuJ1), mesodermal (Desmin), and endodermal (AFP) lineage markers in cells differentiated from SM10d iPSCs.
  • DETAILED DESCRIPTION OF THE INVENTION I. Introduction
  • Genetic variations (e.g., polymorphic alleles) within and among human patient populations underlie, to a large extent, differences in individual disposition to diseases, disease manifestation, disease severity, and response to treatment (e.g., to drug treatment). The prevalent animal and cellular models for human disease and drug discovery provide a poor representation of the genotypic/phenotypic spectrum extant in the patient populations to be treated. For example, strains of mice and rats commonly used in drug discovery are highly inbred, and thus only represent a very narrow range of possible genotype/phenotype combinations in mice or rats, let alone humans. Likewise, the relatively small number of human cell lines used for drug screening may reflect the genotypic/phenotypic scope of the individuals from which they were derived, but not that of a genetically diverse population. Further, most human cell lines are quite limited in their capacity to generate or phenocopy specific differentiated cell types (e.g., neurons, cardiomyocytes, and hepatocytes) affected by a particular health condition. Also, the cell lines are not representative of cell populations in a subject, since cell lines have been altered to indefinitely replicate. Importantly, in many cases animal models or genetically modified cell models of disease simply fail to adequately recapitulate the cellular disease phenotypes as they actually occur in a human patient's cells. Thus, typical preclinical drug discovery strategies miss many genotype/phenotypes that are present in the human population and will have a direct impact on the therapeutic efficacy and toxicity of a candidate drug compound. A practical consequence of these facts is that more often than not lead compounds fail in human clinical trials despite successful preclinical testing in animal models and transformed cell line models, as mentioned above. Ideally, drug screening and drug target discovery would be performed in biological models that recapitulate the genetic and phenotypic diversity present in a human patient population and the appropriate disease state at the cellular level, well before the clinical trial stage. These drug discovery paradigms are illustrated schematically in FIG. 1. In the traditional drug discovery model (left), candidate therapeutic agents are selected for clinical trials in patients based on their action on specific drug targets and their efficacy/lack of toxicity in animal models. In an alternative drug discovery model (right) the disease-relevant cells derived from patient iPSC lines, as described herein, are the starting point for identification of lead compounds based on their ability to ameliorate a disease-relevant cellular phenotype in patient derived cells.
  • Accordingly, the present disclosure describes human induced pluripotent stem cell lines from selected individuals (e.g., patients), genetically diverse panels of such cell lines, differentiated cells derived from such cell lines, and methods for their use in disease modeling, drug discovery, diagnostics, and individualized therapy.
  • II. Definitions
  • “Candidate drug compound,” as used herein, refers to any test compound to be assayed for its ability to affect a functional endpoint. Some examples of such functional endpoints are ligand binding to a receptor, receptor antagonism, receptor agonism, protein-protein interactions, enzymatic activities, transcriptional responses, etc.
  • “Correcting” a phenotype, as used herein, refers to altering a phenotype such that it more closely approximates a normal phenotype.
  • “iPSC donor,” as used herein, refers to a subject, e.g., a human patient from which one or more induced stem cell lines have been generated. Generally, the genome of an iPSC line corresponds to that of its iPSC donor.
  • “Phenomic analysis,” as used herein, refers to the analysis of phenotypes (e.g., resting calcium level, gene expression profiles, apoptotic index, electrophysiological properties, sensitivity to free radicals, compound uptake and extrusion, kinase activity, second messenger pathway responses) exhibited by a particular type of cell (e.g., cardiomyocytes).
  • “Phenome,” as used herein refers to the set of phenotypes that is subject and cell-type specific. For example, the phenome of hepatocytes and cardiomyocytes from the same individual will be quite distinct even though they share the same genome.
  • An “endogenous allele,” as used herein, refers to a naturally occurring allele that is native to the genome of a cell, i.e., an allele that is not introduced by recombinant methodologies.
  • An “iPSC-derived cell,” as used herein, refers to a cell that is generated from an iPSC either by proliferation of the iPSC to generate more iPSCs, or by differentiation of the iPSC into a different cell type. iPSC-derived cells include cells not differentiated directly from an iPSC, but from an intermediary cell type, e.g., a glial progenitor cell, a neural stem cell, or a cardiac progenitor cell.
  • A “normal” phenotype, as used herein, refers to a phenotype (e.g., apoptotic rate, resting calcium level, kinase activity, gene expression level) that falls within a range of phenotypes found in healthy individuals or that are not associated with (e.g., predictive of) a health condition.
  • III. Induced Stem Cell Lines for Drug Screening and Drug Target Discovery A. Overview
  • The present disclosure provides human induced pluripotent stem cell (iPSC) lines, panels of stem cell lines, and methods for their use in drug discovery, diagnostic, and therapeutic methods as described in detail below. The induced pluripotent stem cell lines disclosed herein are characterized by long term self renewal, a normal karyotype, and the developmental potential to differentiate into a wide variety of cell types (e.g., neurons, cardiomyocytes, and hepatocytes). Induced pluripotent stem cell lines can be differentiated into cell lineages of all three germ layers, i.e., ectoderm, mesoderm, and endoderm.
  • An important nexus exists between a subject (e.g., a patient) and iPSC lines generated from that subject. First, all of the genotypes of iPSC lines and those of the corresponding subject are identical. Thus, genotype-phenotype correlations, uncovered in one are informative for the other, and vice versa. Second, differentiated cells (e.g., neurons) derived ex vivo from an iPSC line will exhibit a complete set of cellular phenotypes (referred to herein as a “phenome”) that are very similar, if not identical, to those of differentiated cells in vivo in the corresponding subject. This point is particularly relevant for developing therapeutics targeted to cells that cannot be routinely obtained from patients (e.g., neurons, cardiomyocytes, hepatocytes, or pancreatic cells). For example, in the case of a patient suffering from a neurodegenerative disease (e.g., parkinson's disease), dopaminergic neurons, which are typically affected by this condition, can be obtained non-invasively by differentiating an iPSC line from the subject, and can then be screened in multiple assays. Thus, iPSC lines provide a renewable source of differentiated cells (e.g., inaccessible differentiated cells) in which pathological cellular phenotypes that are associated with a disease, cell type, and individual may be examined and screened against test compounds. An exemplary, non-limiting embodiment of this approach to disease modeling and drug discovery is schematically illustrated in FIG. 2. iPSC lines and iPSC-derived cells (e.g., motor neurons) are also useful for predicting the efficacy and/or adverse side effects of a candidate drug compound in specific individuals or groups of individuals, as schematically illustrated in FIG. 3. For example, test compounds can be tested for toxicity in hepatocytes differentiated from a genetically diverse panel of induced pluripotent stem cells. Toxicity testing in iPSC-derived hepatocytes can reveal both the overall likelihood of toxicity of a test compound in a target patient population, and the likelihood of toxicity in specific patients within that population.
  • In effect, iPSC lines and iPSC-derived cells (e.g., pancreatic cells) can serve as “cellular avatars,” that reveal cellular phenotypes that are disease, cell-type, and subject-specific to the extent the phenotypes are determined or predisposed by the genome. Collectively, panels of patient induced stem cell lines will represent a wide range of genotype/phenotype combinations in a patient population. Thus, they are useful for developing therapeutics that are effective and safe across a wide range of the relevant target population, or for determining which individuals can be treated effectively and safely with a given therapeutic agent.
  • B. Screening and Selection of Subject Samples
  • Some of the methods described herein utilize induced stem cell lines or panels of induced stem cell lines derived from subjects that meet one or more pre-determined criteria. In some cases subjects and cellular samples from such subjects may be selected for the generation of induced stem cell lines and panels of induced stem cell lines based on one or more of such pre-determined criteria. These include, but are not limited to, the presence or absence of a health condition in a subject (e.g., spinal muscular atrophy, Parkinson's disease, or amyotrophic lateral sclerosis), one or more positive diagnostic criteria for a health condition, a family medical history indicating a predisposition or recurrence of a health condition, the presence or absence of a genotype associated with a health condition, or the presence of at least one polymorphic allele that is not already represented in a panel of induced stem cell lines.
  • In some cases, a panel of induced stem cell lines is generated specifically from individuals diagnosed with a health condition, and from subjects that are free of the health condition. Such health conditions include, without limitation, neurodegenerative disorders; neurological disorders such as cognitive impairment, and mood disorders; auditory disease such as deafness; osteoporosis; cardiovascular diseases; diabetes; metabolic disorders; respiratory diseases; drug sensitivity conditions; eye diseases such as macular degeneration; immunological disorders; hematological diseases; kidney diseases; proliferative disorders; genetic disorders, traumatic injury, stroke, organ failure, or loss of limb.
  • Examples of neurodegenerative disorders include, but are not limited to, Alexander's disease, Alper's disease, Alzheimer's disease, amyotrophic lateral sclerosis, ataxia telangiectasia, Batten disease, bovine spongiform encephalopathy, Canavan disease, Cockayne syndrome, corticobasal degeneration, Creutzfeldt-Jakob disease, Huntington's disease, HIV-associated dementia, Kennedy's disease, Krabbe's disease, lewy body dementia, Machado-Joseph disease, multiple sclerosis, multiple system atrophy, narcolepsy, neuroborreliosis, Parkinson's disease, Pelizaeus-Merzbacher Disease, Pick's disease, primary lateral sclerosis, prion diseases, Refsum's disease, Sandhoffs disease, Schilder's disease, subacute combined degeneration of spinal cord secondary to pernicious anaemia, schizophrenia, spinocerebellar ataxia, spinal muscular atrophy, Steele-Richardson-Olszewski disease, and tabes dorsalis.
  • Examples of neurological disorders include, stroke, cognitive impairment, and mood disorders.
  • Examples of immunological disorders include but are not limited to acquired immune deficiency, leukemia, lymphoma, hypersensitivities (allergy), autoimmune diseases, and severe combined immune deficiency.
  • Examples of autoimmune diseases include but are not limited to acute disseminated encephalomyelitis, addison's disease, ankylosing spondylitis, antiphospholipid antibody syndrome, autoimmune hemolytic anemia, autoimmune hepatitis, bullous pemphigoid, coeliac disease, dermatomyositis, diabetes mellitus type 1, Goodpasture's syndrome, Graves' disease, Guillain-Barré syndrome, Hashimoto's disease, idiopathic thrombocytopenic purpura, lupus erythematosus, multiple sclerosis, myasthenia gravis, pemphigus, pernicious anaemia, polymyositis, primary biliary cirrhosis, rheumatoid arthritis, Sjögren's syndrome, temporal arthritis (also known as “giant cell arthritis”), vasculitis, Wegener's granulomatosis.
  • Examples of cardiovascular diseases include but are not limited to aneurysm, angina, arrhythmia, atherosclerosis, cardiomyopathy, cerebrovascular accident (stroke), cerebrovascular disease, congenital heart disease, congestive heart failure, myocarditis, valve disease coronary, artery disease dilated, cardiomyopathy, diastolic dysfunction, endocarditis, high blood pressure (hypertension), hypertrophic cardiomyopathy, mitral valve prolapse, myocardial infarction (heart attack), and venous thromboembolism.
  • Examples of metabolic disorders include but are not limited to acid lipase disease, amyloidosis, Barth Syndrome, biotinidase deficiency, carnitine palmitoyl transferase deficiency type II, central pontine myelinolysis, metabolic diseases of muscle including muscular dystrophy, Farber's Disease, glucose-6-phosphate dehydrogenase deficiency, gangliosidoses, trimethylaminuria, Lesch-Nyhan syndrome, lipid storage diseases, metabolic myopathies, methylmalonic aciduria, mitochondrial myopathies, mucopolysaccharidoses, mucolipidoses, mucolipidoses, mucopolysaccharidoses, multiple CoA carboxylase deficiency, nonketotic hyperglycinemia, Pompe disease, propionic acidemia, type I glycogen storage disease, urea cycle disorders, hyperoxaluria, and oxalosis.
  • Examples of proliferative disorders include but are not limited to one or more of the following: carcinomas, sarcomas, lymphomas, leukemias, germ cell tumors, blastic tumors, prostate cancer, lung cancer, colorectal cancer, bladder cancer, cutaneous melanoma, breast cancer, endometrial cancer, and ovarian cancer.
  • Further examples of diseases or disorders may be found in U.S. application Ser. Nos. 12/157,967, WSGR Docket Number 36588-704.201; filed on Jun. 13, 2008; First Inventor Kazuhiro Sakurada, 61/061,594, WSGR Docket Number 36588-707.101; filed on Jun. 13, 2008; First Inventor Kazuhiro Sakurada, and WSGR Docket Number 36588-704.502, filed Jun. 12, 2009; First Inventor Kazuhiro Sakurada, which are hereby incorporated by reference. It is also anticipated that the methods of the present invention include marketing and selling products and services for the treatment of diseases and disorders including, but not limited to, those mentioned herein.
  • Such subjects may be identified in, e.g., gene association studies, clinical studies, and hospitals, preferably after a final diagnosis of a health condition has been made. Preferably, subjects are identified in gene association studies that include non-affected control individuals.
  • In other cases, iPSC lines are generated from subjects screened for the presence or absence of at least one allele associated with a health condition or a predisposition for a health condition. Such alleles indicate that an individual, though not exhibiting overt symptoms of a health condition, has a high risk of developing the health condition. For example, BRCA1 have been used to indicate a high likelihood of developing breast cancer. Genotyping of subjects may be performed on samples from a number of sources, e.g., blood banks, sperm banks, gene-association studies, hospitals, clinical trials, or any other source as long as a living cellular sample can be obtained from the individual that is genotyped. While not wishing to be bound by theory, it is believed that one or more that cellular phenotypes from individuals carrying alleles associated with health conditions will exhibit abnormalities that can serve as more reliable prognostic indicators of a health condition in combination with a genotype than a genotype alone. Further, identification of specific abnormal cellular phenotypes associated with a health condition may indicate a target pathway for screening of prophylactic and therapeutic agents for the health condition.
  • There is an ongoing effort to identify associations between polymorphic alleles present in the human population, e.g., single polymorphisms (SNPs) and the occurrence of common health conditions, e.g., neurodegenerative diseases, psychiatric disorders, metabolic disorders, and cardiovascular diseases. Various types of polymorphic alleles can be found in the human genome as summarized in Table 1.
  • TABLE 1
    Types of Interindividual Variation in the Human Genome
    Genetic Frequency in
    change/variation Abbreviation Description human genome
    Single nucleotide SNP Typically two different nucleotides (biallelic 12,000,000
    polymorphism SNPs) at one defined position, but more rarely
    also triallelic variants occur
    Deletions/Insertions InDel Deletions (or insertions, depending on the allele >1,000,000
    frequencies) of between 1 to 1000 nucleotides.
    More frequent are deletions of one or three
    basepairs
    Varying number of VNTR Microsatellites also termed short tandem repeat   >500,000
    tandem recaps (STR) polymorphisms are typically tandem
    repeats of two, three or four nucleotides, but
    repeats up to ten nucleotides in length may also
    classified in this group.
    Minisatellites are VNTR polymorphisms in
    which 10-100 nucleotides are repeated in
    variable numbers. Repeated segments often do
    not have exactly identical sequences.
    VNTRs with larger repeat units (100-1000 bp)
    are termed satellites.
    Copy number CNV Inheritable deletion of multiplication of DNA >1500 loci
    variation segments larger than 1 kb. Currently, about 1500 covering 12% of
    CNVs distributed through all chromosomes are the genome
    known; estimated to cover 12% of the human
    genome length.
  • A number of studies have identified alleles associated with a health condition or a predisposition towards a health condition.
  • Examples of alleles associated with health conditions are known in the art. See, e.g., the databases listed in Table 2.
  • TABLE 2
    List of Publicly Available Databases Containing Alleles Associated
    with a Health Condition or Predisposition to a Health Condition
    Name of
    Website Website URL Brief Description
    Alzgene www.alzforum.org/res/com/gen/alzgene Collection of published
    genetic association studies
    performed on Alzheimer
    Disease phenotypes, from
    database searches and
    journals' contents lists. Case
    and control data presented.
    Cytokine Gene www.nanea.dk/cytokinesnps/ Regularly updated database
    Polymorphism with Medline-based records
    in Human from a systematic review of
    Disease cytokine gene
    polymorphisms associated
    with human disease. Data
    extracted from two
    publications about the study.
    HuGE Navigator hugenavigator.net HuGE Navigator provides
    access to a continuously
    updated knowledge base in
    human genome
    epidemiology, including
    information on population
    prevalence of genetic
    variants, gene-disease
    associations, gene-gene and
    gene-environment
    interactions, and evaluation
    of genetic tests.
    GenAtlas www.genatlas.org Regularly updated database
    of genes, phenotypes and
    references. Among
    numerous databases are
    brief sections on disorders
    associated with genes, with
    lists of citations. May be
    biased towards statistically
    significant results.
    GeneCanvas genecanvas.idf.inserm.fr Database of cardiovascular
    candidate genes and their
    polymorphisms investigated
    at INSERM (Paris, France).
    Data include gene
    frequencies and linkage
    disequilibrium statistics.
    Genetic geneticassociationdb.nih.gov Database of human genetic
    Association association studies of
    Database complex diseases and
    disorders, based on Medline
    records. Data extracted from
    publications.
    Human Obesity obesitygene.pbrc.edu Database of obesity-related
    Gene Map genes, including P values
    Database for association and
    references. Biased in favour
    of statistically significant
    results.
    Infevers fmf.igh.cnrs.fr/infevers Database of genetic
    associations in hereditary
    inflammatory disorders,
    with voluntarily submitted
    entries. Submissions are
    validated by an editorial
    board member.
    MedGene medgene.med.harvard.edu/MEDGENE/ Automated database of gene
    disease association studies
    in Medline.
    OMIM www.ncbi.nlm.nih.gov/omim/ Database of human genes
    and genetic disorders,
    containing textual
    information with links to
    Medline and sequence
    records in the Entrez
    system, and links to
    additional related resources
    at NCBI and elsewhere.
    PharmGKB www.pharmgkb.org Database of genomic data
    and clinical information
    from participants in
    pharmacogenetics research
    studies. Welcomes
    submission of primary data.
    T1DBase t1dbase.org/ Database of type 1 diabetes
    data, including information
    from collaborating
    laboratories. Some
    indication given of
    unpublished data.
  • Some examples of health condition-associated alleles and their corresponding studies are provided in Table 3.
  • TABLE 3
    Some Examples of Alleles Associated with a Health Condition
    Polymorphism(s)
    Disease identified References
    Bipolar rs420259 The Wellcome Trust Case Control
    disorder Consortium (2007), Nature,
    447: 661-678
    Coronary rs1333049 The Wellcome Trust Case Control
    artery Consortium (2007), Nature,
    disease 447: 661-678
    Crohn's rs17221417 The Wellcome Trust Case Control
    disease rs11209026 Consortium (2007), Nature,
    rs10210302 447: 661-678
    rs9858542
    rs17234657
    rs1000113
    rs10761659
    rs10883365
    rs17221417
    rs2542151
    Hypertension The Wellcome Trust Case Control
    Consortium (2007), Nature,
    447: 661-678
    Rheumatoid rs6679677 The Wellcome Trust Case Control
    arthritis rs6457617 Consortium (2007), Nature,
    447: 661-678
    Type 1 rs11761231 The Wellcome Trust Case Control
    Diabetes rs6679677 Consortium (2007), Nature,
    rs9272346 447: 661-678
    rs11171739
    rs17696736
    rs12708716
    Type 2 rs4506565 The Wellcome Trust Case Control
    Diabetes rs9465871 Consortium (2007), Nature,
    rs9939609 447: 661-678
    Gallstone rs1187534 Bush, et al., (2007), Nat Genet,
    disease (D19H) 39: 995-999
    Myocardial rs10757278 Helgadottir, et al., (2007), Science,
    Infarction 316: 1491-1493
    Atrial rs2200733 Gudbjartsson, et al., (2007), Nature,
    fibrillation 448: 353-357
    Type 2 rs1801282 Warren, et al., (2007)
    diabetes rs13266634 Pharmacogenomics, 7: 180-189
    rs1111875
    rs7903146
    rs5219
    rs4402960
    rs7754840
    rs10811661
    rs9300039
    rs8050136
    Type 2 rs13266634 Saxena, et al., (2007), Science,
    diabetes rs1111875 316: 1331-1336
    rs7903146
    rs5219
    rs1801282
    rs10811661
    rs4402960
    rs7754840
    Rheumatoid rs3761847 Plenge, et al., (2007), N Engl J Med,
    arthritis 357: 1199-209
    Exfoliation rs1048661 + Thorleifsson, et al., (2007), Science,
    Glaucoma rs3825942 317: 1397-1400
    Breast rs2981582 Easton, et al., (2007), Nature,
    Cancer rs12443620 447: 1087-1093
    rs8051542
    rs889312
    rs3817198
    rs2107425
    rs13281615
    Colorectal rs6983267 Tomlinson, et al., (2007), Nat Genet,
    cancer 39: 984-988
  • The sequence and other information for any rs-identified SNP can be accessed on the world wide web through the SNP database of the National Center for Biotechnology Information (www.ncbi.nlm.nih.gov/); pulldown menu=“SNP.”
  • Subjects may be screened for alleles in genes that affect response to a therapeutic agent or to a class of therapeutic agents. Examples of such alleles include, but are not limited to, alleles of drug metabolizing enzymes, such as Glucose 6 phosphate dehydrogenase (G6PDH), Butyrlcholine esterase, N-acetyltransferase, Cytochrome P450 isoforms (e.g., 2B6, 2D6, C19, 2C9), Thiopurine S-methyltransferase, Dihydropyrimidine dehydrogenase, and Uridin diphospho-glucuronic acid transferase type 1A1. Alleles of Cytochrome P450 enzyme isoforms can be found, e.g., in a database provided under the “Home Page of the Human Cytochrome P450 (CYP) Allele Nomenclature Committee,” at “www.cypalleles.ki.se/.” Some alleles occur in genes that affect drug transport, including, e.g., multiple drug resistance conferring transporters (MDRs), breast cancer resistance protein (BRCP), multidrug resistance-associated-associated proteins (MRPs), and organic anion-transporting polypeptide (OATP1B1). Other alleles occur in genes that encode drug targets, including, but not limited to, Vitamin K epoxide reductase, Factor V, G-protein coupled receptors (GPCRs). Of note, GPCRs are one of the most common drug targets. Examples of polymorphic alleles in GPCRs can be found in, e.g., the GPCR Natural Variants (“NaVa”) Database, which is accessible on the internet at “nava.liacs.nl/” The GPCR NaVa database describes sequence variants within the family of human G Protein-Coupled Receptors (GPCRs). It includes: rare mutations (frequency <1%); polymorphisms (frequency >1%), including Single Nucleotide Polymorphisms (SNPs); variants without estimates of allele frequency.
  • Polymorphic alleles of interest may be detected and scored in a nucleic acid sample from a subject by any of a number of methods known in the art. For example, detection of multiple alleles may be performed by conducting a nucleic acid array-based assay on a nucleic acid sample from a subject, where the nucleic acid array comprises allele-specific probes (e.g., SNP-specific probes), which, under high stringency hybridization conditions, selectively hybridize with and discriminate between the nucleic acid sequences of two or more polymorphic alleles of interest, e.g., alleles of G-protein coupled receptors.
  • The nucleic acid arrays used to detect polymorphisms may be commercially available nucleic acid arrays. For example, the Affymetrix® Genome-Wide SNP Array 6.0 includes probes for more than 906,000 SNPs and more than 946,000 probes for the detection of copy number variation. Alternatively, the nucleic acid arrays may be custom-made to include to a limited subset of alleles of interest. The design of suitable probe arrays for analysis of predetermined polymorphisms and interpretation of the hybridization patterns is described in detail in WO 95/11995; EP 717,113; and WO 97/29212. Such arrays typically contain first and second groups of probes which are designed to be complementary to different allelic forms of the polymorphism. Each group contains a first set of probes, which is subdivided into subsets, one subset for each polymorphism. Each subset contains probes that span a polymorphism and proximate bases and are complementary to one allelic form of the polymorphism. Thus, within the first and second probe groups there are corresponding subsets of probes for each polymorphism. The hybridization patterns of these probes to target samples can be analyzed by footprinting or cluster analysis, as described above. For example, if the first and second probes groups contain subsets of probes respectively complementarity to first and second allelic forms of a polymorphic site spanned by the probes, then on hybridization of the array to a sample that is homozygous for the first allelic form all probes in the subset from the first group show specific hybridization, whereas probes in the subset from the second group that span the polymorphism show only mismatch hybridization. The mismatch hybridization is manifested as a footprint of probe intensities in a plot of normalized probe intensity (i.e., target/reference intensity ratio) for the subset of probes in the second group. Conversely, if the target sample is homozygous for the second allelic form, a footprint is observed in the normalized hybridization intensities of probes in the subset from the first probe group. If the target sample is heterozygous for both allelic forms then a footprint is seen in normalized probe intensities from subsets in both probe groups although the depression of intensity ratio within the footprint is less marked than in footprints observed with homozygous alleles. Analysis of the hybridization pattern of a nucleic acid array to a nucleic acid sample indicates which allelic form is present at some or all of the SNP sequences represented on the array. Thus, an individual or an iPSC line generated from an individual can be characterized with a polymorphic profile representing allelic variants of interest, e.g., alleles associated with a health condition.
  • In other embodiments, an allele is detected using a primer extension reaction or amplification reaction. For example, a nucleic acid sample containing (or suspected of containing) a target nucleic acid molecule can be contacted with an oligonucleotide primer that, upon further contact with a polymerase, can be extended up to and, if desired, beyond the position of the SNP. In addition, the nucleic acid sample can be contacted with an amplification primer pair, comprising a first primer and a second primer, which selectively hybridize to complementary strands of a target nucleic acid molecule and, in the presence of polymerase, allow for generation of an amplification product. For convenience, the primers of an amplification primer pair are referred to as a “first primer” and a “second primer”; however, reference herein to a “first primer” or a “second primer” is not intended to indicate any importance, order of addition, or the like. It will be further recognized that an amplification primer pair requires that the first and second primer comprise what are commonly referred to as a forward primer and a reverse primer.
  • A primer extension or PCR amplification reaction can be designed such that the presence of a particular nucleotide at an SNP position can be determined by the presence or size of the extension and/or amplification product, in which case the SNP can be determined using a method such as gel electrophoresis, capillary gel electrophoresis, or mass spectrometry; or the amplification product can be sequenced to determine the nucleotide at the SNP position. In addition, the SNP can be detected indirectly, for example, by further contacting the sample with a detector oligonucleotide, which can selectively hybridize to a nucleotide sequence of the first amplification product comprising the SNP position; and detecting selective hybridization of the detector oligonucleotide, as above.
  • Various other methods useful for genotyping are known to the art and can be applied to the present methods. For example, PCR can be performed using TaqMan® reagents, followed by reading the plates at this endpoint. Molecular beacons, Amplifluor® or TriStar® reagents and methods similarly can be used (Stratagene; Intergen). Amplification products also can be detected using an ELISA format, for example, using a design in which one primer is biotinylated and the other contains digoxygenin. The amplification products are then bound to a streptavidin plate, washed, reacted with an enzyme-conjugated antibody to digoxygenin, and developed with a chromogenic, fluorogenic, or chemiluminescent substrate for the enzyme. Alternatively, a radioactive method can be used to detect generated amplification products, for example, by including a radiolabeled deoxynucleoside triphosphate into the amplification reaction, then blotting the amplification products onto DEAE paper for detection. In addition, if one primer is biotinylated, then streptavidin-coated scintillation proximity assay plates can be used to measure the PCR products. Additional methods of detection can use a chemiluminescent label, for example, a lanthanide chelate such as used in the DELFIA® assay (Pall Corp.), an electrochemiluminescent label such as ruthenium tris-bipyridy (ORI-GEN), or a fluorescent label, for example, using fluorescence correlation spectroscopy.
  • An assay system that is commercially available and can be used to identify a nucleotide occurrence of one or more SNPs is the SNP-IT® assay system (Orchid BioSciences, Inc.; Princeton N.J.). In general, the SNP-IT® method is a three step primer extension reaction. In the first step a target nucleic acid molecule is isolated from a sample by hybridization to a capture primer, which provides a first level of specificity. In a second step the capture primer is extended from a terminating nucleotide triphosphate at the target SNP site, which provides a second level of specificity. In a third step, the extended nucleotide triphosphate can be detected using a variety of known formats, including, for example, by direct fluorescence, indirect fluorescence, an indirect colorimetric assay, mass spectrometry, or fluorescence polarization. Reactions conveniently can be processed in 384 well format in an automated format using a SNP stream® instrument (Orchid BioSciences, Inc.).
  • Various methods for genotyping SNP alleles, selected as described herein, are readily adaptable to high throughput assays. For example, an amplification reaction such as PCR can be performed using inexpensive robotic thermocyclers for a specified number of cycles, then the amplification product generated can be determined at the endpoint of the reaction. Furthermore, the methods can be performed in a multiplex format, for example, using differentially labeled oligonucleotide probes, or performing oligonucleotide ligation assays that result in different sized ligation products, or amplification reactions that result in different sized amplification products. In another example, high-throughput mass spectrometry is used to detect SNP alleles in a target nucleic acid sample. Mass spectrometric methods for SNP genotyping are described in, e.g., U.S. Pat. Nos. 7,132,519, 6,994,998; and U.S. Patent Application No 20060275789.
  • Where hybridization-based methods are used, high stringency conditions are those that result in perfect matches remaining in hybridization complexes, while imperfect matches melt off. Similarly, low stringency conditions are those that allow the formation of hybridization complexes with both perfect and imperfect matches. High stringency conditions are known in the art; see for example Maniatis et al. (1989), Molecular Cloning: A Laboratory Manual, 2d Edition; and Short Protocols in Molecular Biology, ed. Ausubel, et al. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen (1993), Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Acid Probes, “Overview of principles of hybridization and the strategy of nucleic acid assays.” Generally, stringent conditions are selected to be about 5-10 C lower than the thermal melting point (Tm) for the specific sequence at a defined ionic strength pH. The Tm is the temperature (under defined ionic strength, pH and nucleic acid concentration) at which 50% of the probes complementary to the target hybridize to the target sequence at equilibrium (as the target sequences are present in excess, at Tm, 50% of the probes are occupied at equilibrium). Stringent conditions will be those in which the salt concentration is less than about 1.0 M sodium ion, typically about 0.01 to 1.0 M sodium ion concentration (or other salts) at pH 7.0 to 8.3 and the temperature is at least about 3° C. for short probes (e.g. 10 to 50 nucleotides) and at least about 6° C. for long probes (e.g. greater than 50 nucleotides). Stringent conditions may also be achieved with the addition of destabilizing agents such as formamide. In another embodiment, less stringent hybridization conditions are used; for example, moderate or low stringency conditions may be used, as are known in the art. See, e.g., Maniatis and Ausubel, supra, and Tijssen, supra.
  • C. Methods for Inducing Pluripotent Stem Cell Lines
  • iPSC lines may be induced from a wide variety of mammalian cells, e.g., human somatic cells, such as fibroblasts, bone marrow-derived mononuclear cells, skeletal muscle cells, adipose cells, peripheral blood mononuclear cells, macrophages, hepatocytes, keratinocytes, oral keratinocytes, hair follicle dermal cells, gastric epithelial cells, lung epithelial cells, synovial cells, kidney cells, skin epithelial cells or osteoblasts. Methods for inducing multipotent and pluripotent stem cell lines are further disclosed in U.S. application Ser. Nos. 12/157,967, WSGR docket number 36588-704.201; filed Jun. 13, 2008; first inventor Kazuhiro Sakurada, 61/061,594, WSGR Docket Number 36588-707.101; filed on Jun. 13, 2008; First Inventor Kazuhiro Sakurada, and 61/061,565, WSGR Docket Number 36588-702.101; filed on Jun. 13, 2008; First Inventor Kazuhiro Sakurada, which are hereby incorporated by reference in their entirety.
  • The cells to be induced can originate from many different types of tissue, e.g., bone marrow, skin (e.g., dermis, epidermis), muscle, adipose tissue, peripheral blood, foreskin, skeletal muscle, or smooth muscle. The cells can also be derived from neonatal tissue, including, but not limited to: umbilical cord tissues (e.g., the umbilical cord, cord blood, cord blood vessels), the amnion, the placenta, or other various neonatal tissues (e.g., bone marrow fluid, muscle, adipose tissue, peripheral blood, skin, skeletal muscle etc.).
  • The cells can be derived from neonatal or post-natal tissue collected from a mammal within the period from birth, including cesarean birth, to death. For example, the tissue may be from a mammal who is >10 minutes old, >1 hour old, >1 day old, >1 month old, >2 months old, >6 months old, >1 year old, >2 years old, >5 years old, >10 years old, >15 years old, >18 years old, >25 years old, >35 years old, >45 years old, >55 years old, >65 years old, <80 years old, <70 years old, <60 years old, <50 years old, <40 years old, <30 years old, <20 years old or <10 years old. In some examples, the tissue is from a human age 18, 20, 21, 23, 24, 25, 28, 29, 31, 33, 34, 35, 37, 38, 40, 41, 42, 43, 44, 47, 51, 55, 61, 63, 65, 70, 77, or 85 years old.
  • The cells may be from non-embryonic tissue, e.g., at a stage of development later than the embryonic stage. In some cases, the cells may be derived from a fetus. In some cases, the cells are not from a fetus. In some cases, the cells are from an embryo. In some cases, the cells are not from an embryo.
  • The cells can be obtained from a single cell or a population of cells. The population may be homogenous or heterogeneous. The cells may be a population of cells found in a human cellular sample, e.g., a biopsy or blood sample. In some cases, the cells are a cell line. In some cases, the cells are somatic cells. In some cases, the cells are derived from cells fused to other cells. In some cases, the cells are not derived from cells fused to other cells. In some cases, the cells are not derived from cells artificially fused to other cells. In some cases, the cells are not: a cell that has been fused with an embryonic stem cell, or a cell that has undergone the procedure known as somatic cell nuclear transfer.
  • The cellular population may include both differentiated and undifferentiated cells. In some cases, the population primarily contains differentiated cells. In other cases, the population primarily contains undifferentiated cells, e.g., undifferentiated stem cells. The undifferentiated cells within the population may be induced to become pluripotent or multipotent. In some cases, differentiated cells within the cellular population are induced to become pluripotent or multipotent.
  • The cellular population may include undifferentiated cells such as mesenchymal stem cells (MSCs), see, e.g., Pittenger et al. (1999), Science 284 (5411): 143-7, multipotent adult progenitor cells (MAPCs), see, e.g., Jahagirdar et al. (2005), Stem Cell Rev. 1(1): 53-9, and/or marrow-isolated adult multilineage inducible (MIAMI) cells (D'Ippolioto et al., (2004), J. Cell Sci. 117 (Pt 14): 2971-81. MSCs are multipotent cells that arise from the mesenchyme during development. In some cases, the undifferentiated stem cells (e.g., mesenchymal stem cells, MAPCs and MIAMI cells) are stem cells that have not undergone epigenetic inactivating modification by heterochromatin formation due to DNA methylation or histone modification of at least four genes, at least three genes, at least two genes, at least one gene, or none of the following: Nanog, Oct3/4, Sox2 and Tert. Activation, or expression of such genes, e.g., Tert, Nanog, Oct3/4 or Sox2, may occur when human pluripotent stem cells are induced from undifferentiated stem cells present in a human postnatal tissue.
  • Methods for obtaining human somatic cells are well established, as described in, e.g., Schantz and Ng (2004), A Manual for Primary Human Cell Culture, World Scientific Publishing Co., Pte, Ltd. In some cases, the methods include obtaining a cellular sample, e.g., by a biopsy, blood draw, or alveolar or other pulmonary lavage. Other suitable methods for obtaining various types of human somatic cells include, but are not limited to, the following exemplary methods:
  • Bone Marrow
  • The donor is given a general anesthetic and placed in a prone position. From the posterior border of the ilium, a collection needle is inserted directly into the skin and through the iliac surface to the bone marrow, and liquid from the bone marrow is aspirated into a syringe. A mononuclear cell fraction is then prepared from the aspirate by density gradient centrifugation. The collected crude mononuclear cell fraction is then cultured prior to use in the methods described herein for induction pluripotency. For convenience, methods for induction of pluripotency, as described herein, are collectively referred to as “induction.”
  • Postnatal Skin
  • Skin tissue containing the dermis is harvested, for example, from the back of a knee or buttock. The skin tissue is then incubated for 30 minutes at 37° C. in 0.6% trypsin/DMEM (Dulbecco's Modified Eagle's Medium)/F-12 with 1% antibiotics/antimycotics, with the inner side of the skin facing downward.
  • After the skin tissue is turned over to scrub slightly the inner side with tweezers, the skin tissue is finely cut into 1 mm2 sections using scissors, which are then centrifuged at 1200 rpm and room temperature for 10 minutes. The supernatant is removed, and to the tissue precipitate is added 25 ml of 0.1% trypsin/DMEM/F-12/1% antibiotics, antimycotics, and stirred using a stirrer at 37° C. and 200-300 rpm for 40 minutes. After confirming that the tissue precipitate is fully digested, 3 ml fetal bovine serum (FBS) (manufactured by JRH) is added, and filtered sequentially with gauze (Type I manufactured by PIP), a 100 μm nylon filter (manufactured by FALCON) and a 40 μm nylon filter (manufactured by FALCON). After centrifuging the resulting filtrate at 1200 rpm and room temperature for 10 minutes to remove the supernatant, DMEM/F-12/1% antibiotics, antimycotics is added to wash the precipitate, and then centrifuged at 1200 rpm and room temperature for 10 minutes. The cell fraction thus obtained is then cultured prior to induction.
  • Postnatal Skeletal Muscle
  • After the epidermis of a connective tissue containing muscle such as the lateral head of the biceps brachii muscle or the sartorius muscle of the leg is cut and the muscle tissue is excised, it is sutured. The whole muscle obtained is minced with scissors or a scalpel, and then suspended in DMEM (high glucose) containing 0.06% collagenase type IA and 10% FBS, and incubated at 37° C. for 2 hours.
  • By centrifugation, cells are collected from the minced muscle, and suspended in DMEM (high glucose) containing 10% FBS. After passing the suspension through a microfilter with a pore size of 40 μm and then a microfilter with a pore size of 20 μm, the cell fraction obtained may be cultured according to the method described in 6. below as crude purified cells containing undifferentiated stem cells, and used for the induction of human pluripotent stem cells of the present invention.
  • Postnatal Adipose Tissue
  • Cells derived from adipose tissue for use in the present invention may be isolated by various methods known to a person skilled in the art. For example, such a method is described in U.S. Pat. No. 6,153,432, which is incorporated herein in its entirety. A preferred source of adipose tissue is omental adipose tissue. In humans, adipose cells are typically isolated by fat aspiration.
  • In one method of isolating cells derived from adipose cells, adipose tissue is treated with 0.01% to 0.5%, preferably 0.04% to 0.2%, and most preferably about 0.1% collagenase, 0.01% to 0.5%, preferably 0.04%, and most preferably about 0.2% trypsin and/or 0.5 ng/ml to 10 ng/ml dispase, or an effective amount of hyaluronidase or DNase (DNA digesting enzyme), and about 0.01 to about 2.0 mM, preferably about 0.1 to about 1.0 mM, most preferably 0.53 mM concentration of ethylenediaminetetraacetic acid (EDTA) at 25 to 50° C., preferably 33 to 40° C., and most preferably 37° C. for 10 minutes to 3 hours, preferably 30 minutes to 1 hour, and most preferably 45 minutes.
  • Cells are passed through nylon or a cheese cloth mesh filter of 20 microns to 800 microns, more preferably 40 microns to 400 microns, and most preferably 70 microns. Then the cells in the culture medium are subjected to differential centrifugation directly or using Ficoll or Percoll or another particle gradient. The cells are centrifuged at 100 to 3000×g, more preferably 200 to 1500×g, most preferably 500×g for 1 minute to 1 hours, more preferably 2 to 15 minutes and most preferably 5 minutes, at 4 to 50° C., preferably 20 to 40° C. and more preferably about 25° C.
  • The adipose tissue-derived cell fraction thus obtained may be cultured according to the method described herein as crude purified cells containing undifferentiated stem cells, and used for the induction of human pluripotent or multipotent stem cells.
  • Blood
  • About 50 ml to about 500 ml vein blood or cord blood is collected, and a mononuclear cell fraction is obtained by the Ficoll-Hypaque method, as described in, e.g., Kanof et al. (1993), Current Protocols in Immunology (J. E. Coligan, A. M. Kruisbeek, D. H. Margulies, E. M. Shevack, and W. Strober, eds.), ch. 7.1.1.-7.1.5, John Wiley & Sons, New York).
  • After isolation of the mononuclear cell fraction, approximately 1×107 to 1×108 human peripheral blood mononuclear cells are suspended in a RPMI 1640 medium containing 10% fetal bovine serum, 100 μg/ml streptomycin and 100 units/ml penicillin, and after washing twice, the cells are recovered. The recovered cells are resuspended in RPMI 1640 medium and then plated in a 100 mm plastic petri dish at a density of about 1×107 cells/dish, and incubated in a 37° C. incubator at 8% CO2. After 10 minutes, cells remaining in suspension are removed and adherent cells are harvested by pipetting. The resulting adherent mononuclear cell fraction is then cultured prior to the induction period as described herein. In some cases, the peripheral blood-derived or cord blood-derived adherent cell fraction thus obtained may be cultured according to the method described herein as crude purified cells containing undifferentiated stem cells, and used for the induction of human pluripotent stem cells of the present invention.
  • Induction
  • During the induction process, forced expression of certain polypeptides is carried out in cultured cells for a period of time, after which the iPSCs are screened for a number of morphological and gene expression properties that characterize multipotent and pluripotent stem cells. Induced cells that meet these screening criteria may then be subcloned and expanded. In some cases, the cells to be induced may be cultured for a period of time prior to the induction procedure. Alternatively, the cells to be induced may be used directly in the induction process without a prior culture period. In some embodiments, the type of cell culture medium used is the same or very similar before, during, and after the induction process. In other cases, different cell culture media are used at different points. For example, one type of culture medium may be used directly before the induction process, while a second type of media is used during the induction process. At times, a third type of culture medium is used during the induction process.
  • Cells may be cultured in medium supplemented with a particular serum. In some embodiments, the serum is fetal bovine serum (FBS). The serum can also be fetal calf serum (FCS). In some cases, the serum may be Human AB serum. Mixtures of serum may also be used, e.g. mixture of FBS and Human AB, FBS and FCS, or FCS and Human AB.
  • Culture of cells may be carried out under a low serum culture conditions prior to, during, or following induction. A “low serum culture condition” refers to the use of a cell culture medium containing a concentration of serum ranging from 0% (v/v) (i.e., serum-free) to about 5% (v/v), e.g., 0% to 2%, 0% to 2.5%, 0% to 3%, 0% to 4%, 0% to 5%, 0.1% to 2%, 0.1% to 5%, 0.1%, 0.5%, 1%, 1.2%, 1.5%, 2%, 2.5%, 3%, 3.5%, or 4%. In some embodiments, the serum concentration is from about 0% to about 2%. In some cases, the serum concentration is about 2%. In some cases, the serum concentration is preferably 2% or less. In other embodiments, cells are cultured under a “high serum condition,” i.e., greater than 5% serum to about 20% serum, e.g., 6%, 7%, 8%, 10%, 12%, 15%, or 20%. Culturing under high serum conditions may occur prior to, during, and/or after induction.
  • Some representative media that the cells can be cultured in include: MAPC, FBM, ES, MEF-conditioned ES (MC-ES), and mTeSR™ (available, e.g., from StemCell Technologies, Vancouver, Canada), See Ludwig et al (2006), Nat Biotechnol, 24(2):185-187. In other cases, alternative culture conditions for growth of human ES cells are used, as described in, e.g., Skottman et al (2006), Reproduction, 132(5):691-698. In some embodiments, the cells are cultured in MAPC, FBM, MC-ES, or mTeSR™ prior to and/or during the introduction of induction factors to the cells; and the cells are cultured in MC-ES or mTeSR™ medium later in the induction process.
  • MAPC (2% FBS) Medium may comprise: 60% Dulbecco's Modified Eagle's Medium-low glucose, 40% MCDB 201, Insulin Transferrin Selenium supplement, (0.01 mg/ml insulin; 0.0055 mg/ml transferrin; 0.005 μg/ml sodium selenite), 1× linolenic acid albumin (1 mg/mL albumin; 2 moles linoneic acid/mole albumin), 1 nM dexamethasone, 2% fetal bovine serum, 1 nM dexamethasone, 10−4 M ascorbic acid, and 10 μg/ml gentamycin.
  • FBM (2% FBS) Medium may comprise: MCDB202 modified medium, 2% fetal bovine serum, 5 μg/ml insulin, 50 mg/ml gentamycin, and 50 ng/ml amphotericin-B.
  • ES Medium may comprise: 40% Dulbecco's Modified Eagle's Medium (DMEM) 40% F12 medium, 2 mM L-glutamine, 1× non-essential amino acids (Sigma, Inc., St. Louis, Mo.), 20% Knockout Serum Replacement™ (Invitrogen, Inc., Carlsbad, Calif.), and 10 μg/ml gentamycin.
  • MC-ES medium may be prepared as follows. ES medium is conditioned on mitomycin C-treated murine embryonic fibroblasts (MEFs), harvested, filtered through a 0.45-μM filter, and supplemented with about 0.1 mM β mercaptoethanol, about 10 ng/ml bFGF or FGF-2, and, optionally, about 10 ng/ml activin A. In some cases, irradiated MEFs are used in place of the mitomycin C-treated MEFs.
  • When either low or high serum conditions are used for culturing the cells, one or more growth factors such as fibroblast growth factor (FGF)-2; basic FGF (bFGF); platelet-derived growth factor (PDGF), epidermal growth factor (EGF); insulin-like growth factor (IGF); or insulin can be included in the culture medium. Other growth factors that can be used to supplement cell culture media include, but are not limited to one or more: Transforming Growth Factor β-1 (TGF β-1), Activin A, Noggin, Brain-derived Neurotrophic Factor (BDNF), Nerve Growth Factor (NGF), Neurotrophin (NT)-1, NT-2, or NT 3. In some cases, one or more of such factors is used in place of the bFGF or FGF-2 in the MC-ES medium or other cell culture medium.
  • In some cases, the concentration of growth factors in the culture media described herein (e.g., MAPC, FBM, MC-ES, mTeSR™) is from about 2 ng/ml to about 20 ng/ml, e.g., about 2 ng/ml, 3 ng/ml, 4 ng/ml, 5 ng/ml, 6 ng/ml, 7 ng/ml, 8 ng/ml, 10 ng/ml, 12 ng/ml, 14 ng/ml, 15 ng/ml, 17 ng/ml, or 20 ng/ml. In some embodiments, the concentration of bFGF or FGF2 is from about 2 ng/ml to about 5 ng/ml; from about 5 ng/ml to about 8 ng/ml; from about 9 ng/ml to about 11 ng/ml; from about 11 ng/ml to about 15 ng/ml; or from about 15 ng/ml to about 20 ng/ml.
  • The growth factors may be used alone or in combination. For example, FGF-2 may be added alone to the medium; in another example, both PDGF and EGF are added to the culture medium.
  • In some examples, following initiation of the forced expression of genes or polypeptides (e.g., immediately after a retroviral infection period) in cells, the “iPSCs” are maintained in MC-ES medium as described herein.
  • In some embodiments, cells are maintained in the presence of a rho, or rho-associated, protein kinase (ROCK) inhibitor to reduce apoptosis. In some cases, an inhibitor of Rho associated kinase is added to the culture medium. For example, the addition of Y-27632 (Calbiochem; water soluble) or Fasudil (HA1077: Calbiochem), an inhibitor of Rho associated kinase (Rho associated coiled coil-containing protein kinase) may be used to culture the human pluripotent stem cells of the present invention. In some cases the concentration of Y-27632 or Fasudil, is from about 5 μM to about 20 e.g., about 5 μM, 10 μM, 15 μM, or 20 μM.
  • The cells may be cultured for about 1 to about 12 days e.g., 2 days, 3 days, 4.5 days, 5 days, 6.5 days, 7 days, 8 days, 9 days, 10 days, or any other number of days from about 1 day to about 12 days prior to undergoing the induction methods described herein.
  • In some cases, the iPSCs are cultured in complete ES medium in a 37° C., 5% CO2 incubator, with medium changes about every 1 to 2 days. In some embodiments, induced the iPSCs are cultured and observed for about 14 days to about 40 days, e.g., 15, 16, 17, 18, 19, 20, 23, 24, 27, 28, 29, 30, 31, 33, 34, 35, 36, 37, 38 days, or any other period from about 14 days to about 40 days prior to identifying and selecting clones comprising “iPSCs” based on morphological characteristics. Morphological characteristics for identifying iPSC clones include, but are not limited to, a small cell size with a high nucleus-to-cytoplasm ratio; formation of small monolayer colonies within the space between parental cells (e.g., between fibroblasts).
  • The cells may be plated at a cell density of about 1×103 cells/cm2 to about 1×104 cells/cm2, e.g., 2×103 cells/cm2, 3.5×103 cells/cm2, 6×103 cells/cm2, 7×103 cells/cm2, 9×103 cells/cm2, or any other cell density from about 1×103 cells/cm2 to about 1×104 cells/cm2.
  • The cells can be plated and cultured directly on tissue culture-grade plastic. Alternatively, cells are plated and cultured on a coated substrate, e.g., a substrate coated with fibronectin, gelatin, matrigel™, collagen, or laminin. Suitable cell culture vessels include, e.g., 35 mm, 60 mm, 100 mm, and 150 mm cell culture dishes, 6-well cell culture plates, and other size-equivalent cell culture vessels. In some cases, the cells are cultured with feeder cells. For example, the cells may be cultured on a layer, or carpet, of MEFs.
  • Media with low concentrations of serum may be particularly useful to enrich for undifferentiated stem cells. The undifferentiated cells cultured under low serum conditions may or may not share certain properties with MSCs, MAPCs, and/or MIAMI cells. Differences in phenotype may be due, in part, to culture methods used to obtain MSCs, MAPCs and MIAMI cells. For example, MSCs are often obtained by isolating the non-hematopoeitic cells (e.g., interstitial cells) adhering to a plastic culture dish when tissue, e.g., bone marrow, fat, muscle, or skin etc., is cultured in a culture medium containing a high-concentration serum (5% or more). However, even under these culture conditions, a very small number of undifferentiated cells can be maintained, especially if the cells were passaged under certain culture conditions (e.g., low passage number or low-density culturing).
  • In some embodiments, in order to culture and grow human pluripotent stem cells induced from the undifferentiated stem cells of the present invention present in a human postnatal tissue, it is preferred that the cells are subcultured every 5 to 7 days in a culture medium containing the additives described herein on a MEF-covered plastic culture dish or a matrigel-coated plastic culture dish. In some cases, the cells may be cultured at a low density, which may be accomplished by splitting the cells from about 1:6 to 1:3 or by plating the cells at 103 cells/cm2 to 3×104 cells/cm2.
  • Primary culture ordinarily occurs immediately after the cells are isolated from a donor, e.g., human. The primary cells can be subjected to a second subculture, a third subculture, a fourth subculture, and greater than four subcultures. A “second” subculture describes primary culture cells subcultured once, a “third” subculture describes primary cultures subcultured twice, a “fourth” subculture describes primary cells subcultured three times, etc. The culture techniques described herein may generally include culturing from the period between the primary culture and the fourth subculture, but other culture periods may also be employed. Preferably, cells are cultured from primary culture to second subculture.
  • Inducing a cell to become pluripotent can be accomplished in numerous ways. In some embodiments, the methods for induction of pluripotency in one or more cells include forcing expression of a set of induction factors (Ws). In some cases, the set of IFs includes one or more: an Oct3/4 polypeptide, a Sox2 polypeptide, a Klf4 polypeptide, or a c-Myc polypeptide. In some cases, the set does not include a c-Myc polypeptide. For example, the set of IFs can include: an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide, but not a c-Myc polypeptide. In some cases, the set of IFs does not include polypeptides that might increase the risk of cell transformation.
  • In some cases, the set may include a c-Myc polypeptide. In certain cases, the c-Myc polypeptide is a constitutively active variant of c-Myc. In some instances, the set includes a c-Myc polypeptide capable of inducible activity, e.g., a c-Myc-ER polypeptide, see, e.g., Littlewood, et al. (1995) Nucleic Acid Res. 23(10):1686-90.
  • In other cases, the set of IFs may include: an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide, but not a TERT polypeptide, a SV40 Large T antigen polypeptide, HPV16 E6 polypeptide, a HPV16 E7 polypeptide, or a Bmi1 polypeptide. In some cases, the set of IFs does not include a TERT polypeptide. In some cases, the set of IFs does not include a SV40 Large T antigen. In other cases, the set of IFS does not include a HPV16 E6 polypeptide or a HPV16 E7 polypeptide.
  • In some cases, the set of Ws includes three Ws, wherein two of the three IFs are an Oct3/4 polypeptide and a Sox2 polypeptide. In other cases, the set of IFs includes two Ws, wherein the two polypeptides are a c-Myc polypeptide and a Sox2 polypeptide In some cases, the set of induction factors is limited to Oct 3/4, Sox2, and Klf4 polypeptides. In other cases, the set of induction factors may be limited to a set of four Ws: an Oct3/4 polypeptide, a Sox2 polypeptide, a Klf4 polypeptide, and a c-Myc polypeptide.
  • A set of Ws may include Ws in addition to an Oct 3/4, a Sox2, and a Klf4 polypeptide. Such additional Ws include, but are not limited to Nanog, TERT, LIN28, CYP26A1, GDF3, FoxD3, Zfp42, Dnmt3b, Ecat1, and Tel1 polypeptides. In some cases, the set of additional Ws does not include a c Myc polypeptide. In some cases, the set of additional IFs does not include polypeptides that might increase the risk of cell transformation.
  • Forced expression of Ws may be maintained for a period of at least about 7 days to at least about 40 days, e.g., 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days, 20 days, 21 days, 25 days, 30 days, 33 days, or 37 days.
  • The efficiency of inducing pluripotency in cells of a human population of cells is from at least about 0.001% to at least about 0.01% of the total number of cells to be induced, e.g., 0.002%, 0.0034%, 0.004%, 0.005%, 0.0065%, 0.007%, 0.008%, or 0.0085%.
  • D. HDAC Inhibitor
  • Induction of the cells may be accomplished by combining histone deacetylase (HDAC) inhibitor treatment with forced expression of sets of Ws. The cells to be induced may be undifferentiated stem cells present in a human postnatal tissue. In other cases, the cells to be induced are differentiated cells or are a mixture of differentiated or undifferentiated cells.
  • The HDAC may be combined with the forced expression of a specific set of Ws, e.g., Oct 3/4, a Sox2, and a Klf4. For example, a human somatic cell is induced to become pluripotent after HDAC inhibitor treatment is combined with forced expression of Oct3/4, Sox2 and Klf4 or forced expression of Oct3/4, Sox2, Klf4, and c-Myc. In some cases, human pluripotent stem cells can be induced by introducing three genes of Oct3/4, Sox2 and Klf4 or three genes of Oct3/4, Sox2 and Klf4 plus the c-Myc gene or a HDAC inhibitor into undifferentiated stem cells present in a human postnatal tissue in which each gene of Tert, Nanog, Oct3/4 and Sox2 has not undergone epigenetic inactivation. In still other cases, human pluripotent stem cells are induced by introducing three genes of Oct3/4, Sox2 and Klf4 or three genes of Oct3/4, Sox2 and Klf4 plus the c-Myc gene or a histone deacetylase inhibitor into undifferentiated stem cells after the undifferentiated stem cells were amplified by a primary culture or a second subculture, or a subculture in a low density and subculturing in a culture medium comprising a low-concentration serum.
  • Cells may be treated with one or more HDACs for about 2 hours to about 5 days, e.g., 3 hours, 6 hours, 12 hours, 14 hours, 18 hours, 1 day, 2 days, 3 days, or 4 days. Treatment with HDAC inhibitor may be initiated prior to beginning forced expression of IFs in the cells. In some cases, HDAC inhibitor treatment begins during or after forced expression of IFs in the cells. In other cases, HDAC inhibitor treatment begins prior to forced expression and is maintained during forced expression.
  • Suitable concentrations of an HDAC inhibitor range from about 0.001 nM to about 10 mM, depending on the particular HDAC inhibitor to be used, but are selected so as to not significantly decrease cell survival in the treated cells. The HDAC concentration may range from 0.01 nM, to 1000 nM. In some embodiments, the HDAC concentration ranges from about 0.01 nM to about 1000 nM, e.g., about 0.05 nM, 0.1 nM, 0.5 nM, 0.75 nM, 1.0 nM, 1.5 nM, 10 nM, 20 nM, 40 nM, 50 nM, 100 nM, 200 nM, 300 nM, 500 nM, 600 nM, 700 nM, 800 nM, or other concentration from about 0.01 nM to about 1000 nM. Cells are exposed for 1 to 5 days or 1 to 3 days. For example, cells are exposed 1 day, 2 days, 3 days, 4 days or 5 days.
  • Multiple varieties of HDAC inhibitors can be used for the induction experiments. In a preferred embodiment, the HDAC inhibitor MS-275 is used. Examples of suitable HDAC inhibitors include, but are not limited to, any the following:
  • A. Trichostatin A and its analogs, for example: trichostatin A (TSA); and trichostatin C (Koghe et al. 1998, Biochem. Pharmacol. 56: 1359-1364).
  • B. Peptides, for example: oxamflatin [(2E)-5-[3-[(phenylsulfonyl)aminophenyl]-pent-2-ene-4-inohydroxamic acid (Kim et al., Oncogene 18: 2461-2470 (1999)); Trapoxin A (cylco-(L-phenylalanyl-L-phenylalanyl-D-pipecolinyl-L-2-amino-8-oxo-9,10-epoxy-decanoyl) (Kijima et al., J. Biol. Chem. 268: 22429-22435 (1993)); FR901228, depsipeptide (Nakajima et al., Ex. Cell RES. 241: 126-133 (1998)); FR225497, cyclic tetrapeptide (H. Mori et al., PCT International Patent Publication WO 00/08048 (Feb. 17, 2000)); apicidin, cyclic tetrapeptide [cyclo-(N—O-methyl-L-tryptophanyl-L-isoleucinyl-D-pipecolinyl-L-2-amino-8-oxodecanoyl)] (Darkin-Rattray et al., Proc. Natl. Acad. Sci. U.S.A. 93: 13143-13147 (1996); apicidin Ia, apicidin Ib, apicidin Ic, apicidin IIa, and apicidin IIb (P. Dulski et al., PCT International Patent Publication WO 97/11366); HC-toxin, cyclic tetrapeptide (Bosch et al., Plant Cell 7: 1941-1950 (1995)); WF27082, cyclic tetrapeptide (PCT International Patent Publication WO 98/48825); and chlamydocin (Bosch et al., supra).
  • C. Hybrid polar compounds (HPC) based on hydroxamic acid, for example: salicyl hydroxamic acid (SBHA) (Andrews et al., International J. Parasitology 30: 761-8 (2000)); suberoylanilide hydroxamic acid (SAHA) (Richon et al., Proc. Natl. Acad. Sci. U.S.A. 95: 3003-7 (1998)); azelaic bishydroxamic acid (ABHA) (Andrews et al., supra); azelaic-1-hydroxamate-9-anilide (AAHA) (Qiu et al., Mol. Biol. Cell 11: 2069-83 (2000)); M-carboxy cinnamic acid bishydroxamide (CBHA) (Ricon et al., supra); 6-(3-chlorophenylureido) carpoic hydroxamic acid, 3-Cl-UCHA) (Richon et al., supra); MW2796 (Andrews et al., supra); and MW2996 (Andrews et al., supra).
  • D. Short chain fatty acid (SCFA) compounds, for example: sodium butyrate (Cousens et al., J. Biol. Chem. 254: 1716-23 (1979)); isovalerate (McBain et al., Biochem. Pharm. 53: 1357-68 (1997)); valproic acid; valerate (McBain et al., supra); 4-phenyl butyric acid (4-PBA) (Lea and Tulsyan, Anticancer RESearch 15: 879-3 (1995)); phenyl butyric acid (PB) (Wang et al., Cancer RESearch 59: 2766-99 (1999)); propinate (McBain et al., supra); butylamide (Lea and Tulsyan, supra); isobutylamide (Lea and Tulsyan, supra); phenyl acetate (Lea and Tulsyan, supra); 3-bromopropionate (Lea and Tulsyan, supra); tributyrin (Guan et al., Cancer RESearch 60: 749-55 (2000)); arginine butyrate; isobutyl amide; and valproate.
  • E. Benzamide derivatives, for example: MS-275 [N-(2-aminophenyl)-4-[N-(pyridine-3-yl-methoxycarbonyl)aminomethyl]benzamide] (Saito et al., Proc. Natl. Acad. Sci. U.S.A. 96: 4592-7 (1999)); and a 3′-amino derivative of MS-275 (Saito et al., supra); and CI-994.
  • A histone deacetylase inhibitor treatment may be carried out, for example, as follows. The concentration of the HDAC inhibitor may depend on a particular inhibitor, but is preferably 0.001 nM to about 10 mM, and more preferably about 0.01 nM to about 1000 nM. The effective amount or the dosage of a histone deacetylase inhibitor is defined as the amount of the histone deacetylase inhibitor that does not significantly decrease the survival rate of cells, specifically undifferentiated stem cells. Cells are exposed for 1 to 5 days or 1 to 3 days. The exposure period may be less than one day. In a specific embodiment, cells are cultured for about 1 to 5 days, and then exposed to an effective amount of a histone deacetylase inhibitor. However, the histone deacetylase inhibitor may be added at the start of culturing. Within such a time frame, a gene-carrying vehicle such as a vector containing a nucleic acid encoding three genes (Oct3/4, Sox2 and Klf4) is introduced into cultured cells by a known method.
  • E. IF Expression Vectors
  • Forced expression of the IFs may comprise introducing one or more mammalian expression vectors encoding an Oct3/4, a Sox2, and a Klf4 polypeptide to a population of cells. The IFs may be introduced into the cells as exogenous genes. In some cases, the exogenous genes are integrated into the genome of a host cell and its progeny. In other cases, the exogenous genes persist in an episomal state in the host cell and its progeny. Exogenous genes are genes that are introduced to the cell from an external source. A gene as used herein is a nucleic acid that includes an open reading frame encoding a polypeptide of interest, e.g., an IF. The gene preferably includes a promoter operably linked to an open reading frame. In some cases, a natural version of the gene may already exist in the cell but an additional “exogenous gene” is added to the cell to induce polypeptide expression.
  • The one or more mammalian expression vectors may be introduced into greater than 20% of the total population of cells, e.g., 25%, 30%, 35%, 40%, 44%, 50%, 57%, 62%, 70%, 74%, 75%, 80%, 90%, or other percent of cells greater than 20%. A single mammalian expression vector may contain two or more of the just-mentioned IFs. In other cases, one or more expression vectors encoding an Oct 3/4, Sox2, Klf4, and c Myc polypeptide are used. In some embodiments, each of the IFs to be expressed is encoded on a separate mammalian expression vector.
  • In some cases, the IFs are genetically fused in frame with a transport protein amino acid sequence, e.g., that of a VP22 polypeptide as described in, e.g., U.S. Pat. Nos. 6,521,455, 6,251,398, and 6,017,735. Such VP22 sequences confer intercellular transport of VP22 fusion polypeptides from cells that have been transfected with a VP22 fusion polypeptide expression vector to neighboring cells that have not been transfected or transduced. See, e.g., Lemken et al (2007), Mol Ther, 15(2):310-319. Accordingly, the use of IF-VP22 fusion polypeptides can significantly increase the functional efficiency of transfected mammalian expression vectors in the induction methods described herein.
  • Examples of suitable mammalian expression vectors include, but are not limited to: recombinant viruses, nucleic acid vectors, such as plasmids, bacterial artificial chromosomes, yeast artificial chromosomes, human artificial chromosomes, cDNA, cRNA, and PCR product expression cassettes. Examples of suitable promoters for driving expression of IFs in include retroviral LTR elements; constitutive promoters such as CMV, HSV1-TK, SV40, EF-1a, 13 actin; PGK, and inducible promoters, such as those containing Tet-operator elements. In some cases, one or more of the mammalian expression vectors encodes, in addition to an IF, a marker gene that facilitates identification or selection of cells that have been transfected or infected. Examples of marker genes include, but are not limited to, fluorescent protein genes, e.g., for EGFP, DS-Red, YFP, and CFP; proteins conferring resistance to a selection agent, e.g., the neoR gene, and the blasticidin resistance gene.
  • 1. Recombinant Viruses
  • Forced expression of an IF may be accomplished by introducing a recombinant virus carrying DNA or RNA encoding an IF to one or more cells. Additionally, the recombinant virus may carry DNA or RNA encoding more than 1 IF. This includes multiple copies of a single IF or multiple Ws contained within a single virus. For ease of reference, at times a virus will be referred to herein by the IF it is encoding. For example, a virus encoding an Oct3/4 polypeptide, may be described as an “Oct3/4 virus.” In certain cases, a virus may encode more than one copy of an IF or may encode more than one IF, e.g., two IFs, at a time.
  • Different combinations or sets of recombinant viruses may be introduced to the cells. The set of recombinant viruses may include combinations included in any set of IFs described herein. The set of recombinant viruses may include at least: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus. The set of recombinant viruses may be limited to a set of four recombinant viruses: an Oct3/4 virus, a Sox2 virus, a Klf4 virus, and a c-Myc virus. In some cases, the set of recombinant viruses is limited to a set of at least: an Oct3/4 virus, a Sox2 virus, a Klf4 virus, and a c-Myc virus. In some cases, the set of recombinant viruses is limited to Oct 3/4, Sox2, and Klf4 viruses. The set of recombinant viruses may be limited a set of at least: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus. In some cases, the set of recombinant viruses includes three recombinant viruses, wherein two of the three recombinant viruses are an Oct3/4 virus and a Sox2 virus. In still other cases, the set of recombinant viruses may be limited to a Sox2 virus and a c-Myc virus.
  • In some cases, the set of recombinant viruses does not include a recombinant virus that encodes a polypeptide that might increase the risk of cell transformation, e.g., a c-Myc polypeptide. For example, the set of recombinant viruses can include: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus but not a c-Myc virus.
  • In other cases, the set of recombinant viruses includes a c-Myc virus. The c-Myc polypeptide encoded by the c-Myc virus may be wild-type c-Myc or a constitutively active variant of c-Myc. In some instances, the set includes a virus encoding c-Myc polypeptide capable of inducible activity, e.g., a c-Myc-ER polypeptide, see, e.g., Littlewood, et al. (1995) Nucleic Acid Res. 23(10):1686-90.
  • The set of recombinant viruses may include: an Oct3/4 virus, a Sox2 virus, and a Klf4 virus, but not a TERT virus, a SV40 Large T antigen virus, HPV16 E6 virus, a HPV16 E7 virus, or a Bmi1 virus. At times, the set of recombinant viruses does not include a TERT virus. In some cases, the set of recombinant viruses does not include a SV40 virus. In other cases, the set of recombinant viruses does not include a HPV16 E6 virus or a HPV16 E7 virus.
  • A set of recombinant viruses may include viruses in addition to an Oct 3/4, a Sox2, and a Klf4 virus. Such additional recombinant viruses include, but are not limited to Nanog, TERT, CYP26A1, GDF3, FoxD3, Zfp42, Dnmt3b, Ecat1, and Tel1 viruses. In some cases, the set of recombinant viruses includes any IF variant described herein.
  • Individual viruses may be added to the cells sequentially in time or simultaneously. In some cases, at least one virus, e.g., an Oct3/4 virus, a Sox2 virus, a Klf4 virus, or a c-Myc virus, is added to the cells at a time different from the time when one or more other viruses are added. In some examples, the Oct3/4 virus, Sox2 virus and KlF4 virus are added to the cells simultaneously, or very close in time, and the c-Myc virus is added at a time different from the time when the other viruses are added.
  • At least two recombinant viruses may be added to the cells simultaneously or very close in time. In some examples, Oct3/4 virus and Sox2 virus are added simultaneously, or very close in time, and the Klf4 virus or c-Myc virus is added at a different time. In some examples, Oct3/4 virus and Sox2 virus; Oct3/4 virus and Klf4 virus; Oct3/4 virus and c-Myc virus; Sox2 virus and Klf4 virus; Sox2 virus and c-Myc virus; or Klf4 and c-Myc virus are added simultaneously or very close in time.
  • In some cases, at least three viruses, e.g., an Oct3/4 virus, a Sox2 virus, and a Klf4 virus, are added to the cells simultaneously or very close in time. In other instances, at least four viruses, e.g., Oct3/4 virus, Sox2 virus, Klf4 virus, and c-Myc virus are added to the cells simultaneously or very close in time.
  • At times, the efficiency of viral infection can be improved by repetitive treatment with the same virus. In some cases, one or more Oct3/4 virus, Sox2 virus, Klf4 virus, or c-Myc virus is added to the cells at least two, at least three, or at least four separate times.
  • Examples of recombinant viruses include, but are not limited, to retroviruses (including lentiviruses); adenoviruses; and adeno-associated viruses. Often, the recombinant retrovirus is murine moloney leukemia virus (MMLV), but other recombinant retroviruses may also be used, e.g., Avian Leukosis Virus, Bovine Leukemia Virus, Murine Leukemia Virus (MLV), Mink-Cell focus-Inducing Virus, Murine Sarcoma Virus, Reticuloendotheliosis virus, Gibbon Abe Leukemia Virus, Mason Pfizer Monkey Virus, or Rous Sarcoma Virus, see, e.g., U.S. Pat. No. 6,333,195.
  • In other cases, the recombinant retrovirus is a lentivirus (e.g., Human Immunodeficiency Virus-1 (HIV-1); Simian Immunodeficiency Virus (SW); or Feline Immunodeficiency Virus (FIV)), See, e.g., Johnston et al (1999), Journal of Virology 73(6)” 4991-5000 (FIV); Négre D et al (2002) Current Topics in Microbiology and Immunology 261:53-74 (SIV); Naldini et al (1996) Science. 272:263-267 (HIV).
  • The recombinant retrovirus may comprise a viral polypeptide (e.g., retroviral env) to aid entry into the target cell. Such viral polypeptides are well-established in the art, see, e.g., U.S. Pat. No. 5,449,614. The viral polypeptide may be an amphotropic viral polypeptide, e.g., amphotropic env, that aids entry into cells derived from multiple species, including cells outside of the original host species. See, e.g., id. The viral polypeptide may be a xenotropic viral polypeptide that aids entry into cells outside of the original host species. See, e.g., id. In some embodiments, the viral polypeptide is an ecotropic viral polypeptide, e.g., ecotropic env, that aids entry into cells of the original host species. See, e.g., id.
  • Examples of viral polypeptides capable of aiding entry of retroviruses into cells include but are not limited to: MMLV amphotropic env, MMLV ecotropic env, MMLV xenotropic env, vesicular stomatitis virus-g protein (VSV-g), HIV-1 env, Gibbon Ape Leukemia Virus (GALV) env, RD114, FeLV-C, FeLV-B, MLV 10A1 env gene, and variants thereof, including chimeras. See e.g., Yee et al (1994), Methods Cell Biol. Pt A:99-112 (VSV-G); U.S. Pat. No. 5,449,614. In some cases, the viral polypeptide is genetically modified to promote expression or enhanced binding to a receptor.
  • In general, a recombinant virus is produced by introducing a viral DNA or RNA construct into a producer cell. In some cases, the producer cell does not express exogenous genes. In other cases, the producer cell is a “packaging cell” comprising one or more exogenous genes, e.g., genes encoding one or more gag, pol, or env polypeptides and/or one or more retroviral gag, pol, or env polypeptides. The retroviral packaging cell may comprise a gene encoding a viral polypeptide, e.g., VSV-g that aids entry into target cells. In some cases, the packaging cell comprises genes encoding one or more lentiviral proteins, e.g., gag, pol, env, vpr, vpu, vpx, vif, tat, rev, or nef. In some cases, the packaging cell comprises genes encoding adenovirus proteins such as E1A or E1B or other adenoviral proteins. For example, proteins supplied by packaging cells may be retrovirus-derived proteins such as gag, pol, and env, lentivirus-derived proteins such as gag, pol, env, vpr, vpu, vpx, vif, tat, rev, and nef; and adenovirus-derived proteins such as E1A and E1B. In many examples, the packaging cells supply proteins derived from a virus that differs from the virus from which the viral vector derives.
  • Packaging cell lines include but are not limited to any easily-transfectable cell line. Packaging cell lines can be based on 293T cells, NIH3T3, COS or HeLa cell lines. As packaging cells, any cells may be used that can supply a lacking protein of a recombinant virus vector plasmid deficient in at least one gene encoding a protein required for virus packaging. Examples of packaging cell lines include but are not limited to: Platinum-E (Plat-E); Platinum-A (Plat-A); BOSC 23 (ATCC CRL 11554); and Bing (ATCC CRL 11270), see, e.g., Morita et al (2000) Gene Therapy 7:1063-1066; Onishi et al (1996) Experimental Hematology 24:324-329; U.S. Pat. No. 6,995,009. Commercial packaging lines are also useful, e.g., Ampho-Pak 293 cell line, Eco-Pak 2-293 cell line, RetroPack PT67 cell line, and Retro-X Universal Packaging System (all available from Clontech).
  • The retroviral construct may be derived from a range of retroviruses, e.g., MMLV, HIV-1, SIV, Fly, or other retrovirus described herein. The retroviral construct may encode all viral polypeptides necessary for more than one cycle of replication of a specific virus. In some cases, the efficiency of viral entry is improved by the addition of other factors or other viral polypeptides. In other cases, the viral polypeptides encoded by the retroviral construct do not support more than one cycle of replication, e.g., U.S. Pat. No. 6,872,528. In such circumstances, the addition of other factors or other viral polypeptides can help facilitate viral entry. In an exemplary embodiment, the recombinant retrovirus is HIV-1 virus comprising a VSV-g polypeptide but not comprising a HIV-1 env polypeptide.
  • The retroviral construct may comprise: a promoter, a multi-cloning site, and/or a resistance gene. Examples of promoters include but are not limited to CMV, SV40, EF1α, β actin; retroviral LTR promoters, and inducible promoters. The retroviral construct may also comprise a packaging signal (e.g., a packaging signal derived from the MFG vector; a psi packaging signal). Examples of retroviral constructs known in the art include but are not limited to: pMX, pBabeX or derivatives thereof. See e.g., Onishi et al (1996) Experimental Hematology 24:324-329. In some cases, the retroviral construct is a self-inactivating lentiviral vector (SIN) vector, see, e.g., Miyoshi et al., (1998) J Virol. 72(10): 8150-8157. In some cases, the retroviral construct is LL-CG, LS-CG, CL-CG, CS-CG, CLG or MFG. Miyoshi et al., (1998) J Virol. 72(10): 8150-8157; Onishi et al (1996) Experimental Hematology 24:324-329; Riviere et al. (1995) PNAS 92: 6733-6737. Virus vector plasmids (or constructs), include: pMXs, pMXs-IB, pMXs-puro, pMXs-neo (pMXs-IB is a vector carrying the blasticidin-resistant gene in stead of the puromycin-resistant gene of pMXs-puro) [Experimental Hematology, 2003, 31 (11): 1007-14], MFG [Proc. Natl. Acad. Sci. U.S.A. 92, 6733-6737 (1995)], pBabePuro [Nucleic Acids Research 18, 3587-3596 (1990)], LL-CG, CL-CG, CS-CG, CLG [Journal of Virology 72: 8150-8157 (1998)] and the like as the retrovirus system, and pAdex1 [Nucleic Acids Res. 23: 3816-3821 (1995)] and the like as the adenovirus system. In exemplary embodiments, the retroviral construct comprises blasticidin (e.g., pMXs-IB), puromycin (e.g., pMXs-puro, pBabePuro); or neomycin (e.g., pMXs-neo). See, e.g., Morgenstern et al. (1990) Nucleic Acids Research 18: 3587-3596.
  • The retroviral construct may encode one or more IFs. In an exemplary embodiment, pMX vectors encoding Oct3/4, Sox2, Klf4, or c-Myc polypeptides, or variants thereof, are generated or obtained. For example, Oct3/4 is inserted into pMXs-puro to create pMX-Oct3/4; Sox2 is inserted into pMXs-neo to create pMX-Sox2; Klf4 is inserted into pMXs-IB to create pMX-Klf4; and c-Myc is inserted into pMXs-IB to create pMX-c-Myc.
  • Methods of producing recombinant viruses from packaging cells and their uses are well-established, see, e.g., U.S. Pat. Nos. 5,834,256; 6,910,434; 5,591,624; 5,817,491; 7,070,994; and 6,995,009, incorporated herein by reference. Many methods begin with the introduction of a viral construct into a packaging cell line. The viral construct may be introduced by any method known in the art, including but not limited to: the calcium phosphate method [Kokai (Japanese Unexamined Patent Publication) No. 2-227075], the lipofection method [Proc. Natl. Acad. Sci. U.S.A. 84: 7413 (1987)], the electroporation method, microinjection, Fugene transfection, and the like, and any method described herein.
  • In one example, pMX-Oct3/4, pMX-Sox2, pMX-Klf4 or pMX-c-Myc is introduced into PlatE cells by Fugene HD (Roche) transfection. The cell culture medium may be replaced with fresh medium comprising FBM (Lonza) supplemented with FGM-2 Single Quots (Lonza). In some embodiments, the medium is replaced from about 12 to about 60 hours following the introduction of the viral construct, e.g., from about 12 to about 18 hours; about 18 to about 24; about 24 to about 30; about 30 to about 36; about 36 to about 42; about 42 to about 48; about 48 to about 54; or about 54 to about 60 hours following introduction of the viral construct to the producer cells. The medium may be replaced from about 24 to about 48 hours after introduction of the viral construct to the producer cells. The supernatant can be recovered from about 4 to about 24 hours following the addition of fresh media, e.g., about 4 hours. In some cases, the supernatant may be recovered about every 4 hours following the addition of fresh media. The recovered supernatant may be passed through a 0.45 uM filter (Millipore). In some cases, the recovered supernatant comprises retrovirus derived from one or more: pMX-Oct3/4, pMX-Sox2, pMX-Klf4 or pMX-c-Myc.
  • Adenoviral transduction may be used to force expression of the sets of IFs. Methods for generating adenoviruses and their use are well established as described in, e.g., Straus, The Adenovirus, Plenum Press (NY 1984), 451 496; Rosenfeld, et al, Science, 252:431-434 (1991); U.S. Pat. Nos. 6,203,975, 5,707,618, and 5,637,456. In other cases, adenoviral-associated viral transduction is used to force expression of the sets of Ws. Methods for preparing adeno-associated viruses and their use are well established as described in, e.g., U.S. Pat. Nos. 6,660,514 and 6,146,874.
  • In an exemplary embodiment, an adenoviral construct is obtained or generated, wherein the adenoviral construct, e.g., Adeno-X, comprises DNA encoding Oct3/4, Sox2, Klf4, or c-Myc. An adenoviral construct may be introduced by any method known in the art, e.g., Lipofectamine 2000 (Invitrogen) or Fugene HD (Roche), into HEK 293 cells. In some cases, the method further comprises (1) collecting the cells when they exhibit a cytopathic effect (CPE), such effect occurring from about 10 to about 20 days, e.g., about 11, 13, 14, 15, 18, or 20 days after transfection (2) subjecting the cells to from about 2 to about 5 freeze-thaw cycles, e.g., about 3, (3) collecting the resulting virus-containing liquid; (4) purifying the virus using an adenovirus purification kit (Clontech) and (5) storing the virus at −80° C. In some cases, the titer, or plaque-forming unit (PFU), of the adenoviral stocks is determined using an Adeno-X rapid titer kit (Clontech), as described herein.
  • The cells may be infected with a recombinant retrovirus that naturally targets a different cell type or cells originating from a different host. To aid infection efficiency, an exogenous receptor may be first introduced into the human cells. For example, an exogenous mouse receptor may be added to human cells, e.g., postnatal dermal fibroblasts, in order help entry of murine moloney leukemia virus (MMLV). The exogenous receptor may improve infection efficiency by facilitating viral entry, especially if the receptor recognizes a viral polypeptide, e.g., MMLV env, or HIV env. Examples of exogenous receptors include but are not limited to any receptor recognized by a specific retrovirus or lentivirus known in the art. For example, a murine receptor, mCAT1, GenBank Accession No NM007513 protein is used in order to aid MMLV infection of a human target cell. In another example, a CXCR4 or CCR5 receptor is used to aid HIV-1 infection of a target cell.
  • The exogenous receptor may be introduced by methods described herein. Methods of introducing the exogenous receptor include but are not limited to: calcium phosphate transfection, Lipofectamine transfection, Fugene transfection, microinjection, or electroporation. In exemplary embodiments, a virus, e.g., recombinant adenovirus or retrovirus (including lentivirus), is used to introduce the exogenous receptor to the target cell. In a further exemplary embodiment, a recombinant adenovirus is used to introduce MCAT1 to human cells and then a recombinant retrovirus, e.g., MMLV, is used to introduce the IF genes, e.g., Oct 3/4, a Sox2, a Klf4, or c-Myc, to the cells.
  • In some cases, a solution of adenovirus comprising DNA encoding the mCAT1 protein, e.g., an adenovirus generated by using a pADEX-mCAT1 construct, is generated or obtained. The adenovirus solution can comprise Hanks' balanced salt solution. In exemplary embodiments, infection of cells is accomplished by: (1) contacting the p-ADEX-mCAT1 adenovirus solution with cells, e.g., human, non-embryonic fibroblasts, at a multiplicity of infection (m.o.i.) from about 1:5 to about 1:50, e.g., about 1:5, about 1:7; about 1:10; about 1:15, about 1:20, about 1:25; about 1:30, about 1:35; about 1:40; about 1:45, or about 1:50; (2) incubating the cells with the adenovirus solution at room temperature from about 15 minutes to about 2 hours, e.g., about 15 minutes, about 30 minutes, about 45 minutes, about 1 hour, about 1.25 hours, about 1.5 hours, about 1.75 hours, or about 2 hours; and (3) culturing the somatic cell population in culture medium from about 24 hours to about 60 hours, e.g., about 24 hours, about 30 hours, about 36 hours, about 42 hours, about 48 hours, about 54 hours, or about 60 hours.
  • The cells can be infected using a wide variety of methods. In some cases, the infection of cells occurs by (1) combining one or more, two or more, three or more, or all four: pMX-Oct3/4 retrovirus, pMX-Sox2 retrovirus, pMX-Klf4, or pMX-c-Myc to obtain a retrovirus solution (2) supplementing the retrovirus solution with from about 2 ug/ml to about 15 ug/ml Polybrene, e.g., about 2 ug/ml, about 3 ug/ml, about 5 ug/ml, about 7 ug/ml, about 10 ug/ml, about 12 ug/ml, or about 15 ug/ml Polybrene; (3) contacting the retroviral solution with the somatic cells, at a m.o.i. of from about 1:100 to about 1:500, e.g., about 1:100, about 1:150, about 1:200, about 1:250, about 1:300, about 1:350, about 1:400, about 1:450, or about 1:500 m.o.i.; (4) allowing the contacting of step (3) to continue at 37° C. from about 2 hours to about 24 hours, e.g., about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours, about 23 hours, or about 24 hours; (5) soon after the contacting of step (4), changing the medium to MC-ES medium, as described herein; and (6) changing the MC-ES medium with fresh medium every 1 to 2 days. In some cases, infection of somatic cells occurs by following steps (1) through (6) described herein, with the added step of pre-incubating the somatic cells for a length of time, e.g., about 48 hours, prior to contacting the cells with the retroviral solution. Such pre-incubation may be necessary when the somatic cell expresses an exogenous receptor that was introduced by viral transduction, transfection, or other method. Thus, in some embodiments, if an adenovirus or lentivirus is used to introduce an exogenous receptor, e.g., mCAT1, to the somatic cell; such cells may need to be cultured for a length of time from at least about 30 hours to at least about 60 hours, e.g., about 30, about 35, about 40, about 48, about 52, about 55, or about 60 hours.
  • The infection of cells may be accomplished by any method known in the art. e.g., Palsson, B., et al. WO95/10619. Apr. 20, 1995; Morling, F. J. et al. (1995). Gene Therapy. 2: 504-508; Gopp et al. (2006) Methods Enzymol. 420:64-81. For example, the infection may be accomplished by spin-infection or “spinoculation” methods that involve subjecting the cells to centrifugation during the period closely following the addition of virus to the cells. In some cases, virus may be concentrated prior to the infection, e.g., by ultracentrifugation. In some cases, other technologies may be used to aid or improve entry of retroviruses into the target cell. For example, the retrovirus may be contacted with a liposome or immunoliposome to aid or direct entry into a specific cell type. See, e.g., Tan et al. (2007) Mol. Med. 13(3-4): 216-226.
  • The methods of infecting cells described herein may be used to infect cells expressing an exogenous receptor, e.g., MCAT1 or other exogenous receptor described herein. Depending on how the exogenous receptor was introduced, the preincubation period of the cells prior to infection may need to be varied. In some cases, cells that do not express an exogenous receptor are used. Some recombinant retroviruses, e.g., VSV-G pseudotyped recombinant retroviruses, may not need the aid of an exogenous receptor in order to efficiently enter cells. In some examples, VSV-G pseudotyped recombinant retrovirus is introduced to cells following the method described herein, except that the timing of the preculturing of the cells may vary.
  • 2. Nucleic Acid Vectors
  • Nucleic acid vector transfection (e.g., transient transfection) methods may be used to introduce IFs into human cells. Methods for preparation of transfection-grade nucleic acid expression vectors are well established. See, e.g., Sambrook and Russell (2001), “Molecular Cloning: A Laboratory Manual,” 3rd ed, (CSHL Press). Examples of high efficiency transfection efficiency methods include “nucleofection,” as described in, e.g., Trompeter (2003), J Immunol Methods, 274(1-2):245-256, and in international patent application publications WO2002086134, WO200200871, and WO2002086129, transfection with lipid-based transfection reagents such as Fugene® 6 and Fugene® HD (Roche), DOTAP, and lipofectamine™ LTX in combination with the PLUS™ (Invitrogen, Carlsbad, Calif.), Dreamfect™ (OZ Biosciences, Marseille, France), GeneJuice™ (Novagen, Madison, Wis.), polyethylenimine (see, e.g., Lungwitz et al (2005), Eur J Pharm Biopharm, 60(2):247-266), and GeneJammer™ (Stratagene, La Jolla, Calif.), and nanoparticle transfection reagents as described in, e.g., U.S. patent application Ser. No. 11/195,066.
  • 3. Protein Transduction
  • The induction methods may use protein transduction to introduce at least one of the IFs directly into cells. In some cases, protein transduction method includes contacting cells with a composition containing a carrier agent and at least one purified polypeptide comprising the amino acid sequence of one of the above-mentioned IFs. Examples of suitable carrier agents and methods for their use include, but are not limited to, commercially available reagents such as Chariot™ (Active Motif, Inc., Carlsbad, Calif.) described in U.S. Pat. No. 6,841,535; Bioport® (Gene Therapy Systems, Inc., San Diego, Calif.), GenomeONE (Cosmo Bio Co., Ltd., Tokyo, Japan), and ProteoJuice™ (Novagen, Madison, Wis.), or nanoparticle protein transduction reagents as described in, e.g., in U.S. patent application Ser. No. 138,593.
  • The protein transduction method may comprise contacting a cells with at least one purified polypeptide comprising the amino acid sequence of one of the above-mentioned TAs fused to a protein transduction domain (PTD) sequence (IF-PTD fusion polypeptide). The PTD domain may be fused to the amino terminal of an IF sequence; or, the PTD domain may be fused to the carboxy terminal of an IF sequence. In some cases, the IF-PTD fusion polypeptide is added to cells as a denatured polypeptide, which may facilitate its transport into cells where it is then renatured. Generation of PTD fusion proteins and methods for their use are established in the art as described in, e.g., U.S. Pat. Nos. 5,674,980, 5,652,122, and 6,881,825. See also, Becker-Hapak et al (2003), Curr Protocols in Cell Biol, John Wiley & Sons, Inc. Exemplary PTD domain amino acid sequences include, but are not limited to, any of the following: YGRKKRRQRRR (SEQ ID NO:1); RKKRRQRR (SEQ ID NO:2); YARAAARQARA (SEQ ID NO:3); THRLPRRRRRR (SEQ ID NO:4); and GGRRARRRRRR (SEQ ID NO:5).
  • In some cases, individual purified IF polypeptides are added to cells sequentially at different times. In other embodiments, a set of at least three purified IF polypeptides, but not a purified c-Myc polypeptide, e.g., an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide are added to cells. In some embodiments, a set of four purified IF polypeptides, e.g., purified Oct3/4, Sox2, Klf4, and c-Myc polypeptides are added to cells. In some embodiments, the purified IF polypeptides are added to cells as one composition (i.e., a composition containing a mixture of the IF polypeptides). In some embodiments, cells are incubated in the presence of a purified IF polypeptide for about 30 minutes to about 24 hours, e.g., 1 hours, 1.5 hours, 2 hours, 2.5 hours, 3 hours, 3.5 hours 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 12 hours, 16 hours, 18 hours, 20 hours, or any other period from about 30 minutes to about 24 hours. In some embodiments, protein transduction of cells is repeated with a frequency of about every day to about every 4 days, e.g., every 1.5 days, every 2 days, every 3 days, or any other frequency from about every day to about every four days
  • Forced expression of IFs may also be achieved by using nucleic acid-free IF-containing protein transducing nanoparticles (PTN). Details of methods for generating and using PTNs are found in, e.g., Link et al (2006), Nuc Acids Res, 34(2):e16.
  • In some cases, the methods described herein utilize protein transduction and expression vector transduction/transfection in any combination to force expression of a set of IFs as described herein. In some embodiments, retroviral expression vectors are used to force expression of Oct 3/4, a Sox2, and a Klf4 polypeptides in cells, and purified c-Myc purified polypeptide is introduced into cells by protein transduction as described herein. HDAC inhibitor treatment can be used in addition to the purified IF polypeptide. In some cases, a set of at least three purified IF polypeptides, but not a purified c-Myc polypeptide, e.g., an Oct3/4 polypeptide, a Sox2 polypeptide, and a Klf4 polypeptide are added to cells which are also subjected to HDAC inhibitor treatment.
  • F. Subcloning Induced Cell Colonies
  • Cell colonies may be subcloned by any method known in the art. In some cases, the iPSCs are cultured and observed for about 14 days to about 40 days, e.g., 15, 16, 17, 18, 19, 20, 23, 24, 27, 28, 29, 30, 31, 33, 34, 35, 36, 37, 38 days, or any other period from about 14 days to about 40 days prior to identifying and selecting clones comprising “iPSCs” based on morphological characteristics. Morphological characteristics for identifying iPSC clones include, but are not limited to, a small cell size with a high nucleus-to-cytoplasm ratio; formation of small monolayer colonies within the space between parental cells (e.g., between fibroblasts).
  • After washing cell cultures with a physiological buffer, e.g., Hank's balanced salt solution, colonies displaying the morphological characteristics of interest are surrounded by a cloning ring to the bottom of which silicone grease has been applied. About 100 μl (or 50 μl to 150 μl) of “Detachment Medium For Primate ES Cells” (manufactured by ReproCELL, Tokyo Japan) is then added to the cloning ring and incubated at 37° C. for about 20 minutes to form a cell suspension. The cell suspension in the ring containing the detached colonies is then added to about 2 ml of MC ES medium (or other medium described herein), and plated in one well of a MEF-coated 24-well plate or other cell culture vessel of equivalent surface area. After culturing the colony-derived cells in a 5% CO2 cell culture incubator at 37° C. for about 14 hours, the medium is replaced. Subsequently, the medium is replaced about every two days until about 8 days later when a second subculture is carried out.
  • In some embodiments, in the first subculture, the medium is removed, the cells are washed with Hank's balanced salt solution, and Detachment Medium For Primate ES Cells (ReproCell, Tokyo, Japan) is then added to the cells and incubated at 37° C. for 10 minutes. After the incubation, MC-ES medium (2 ml) is added to the resulting cell suspension to quench the activity of the Detachment Medium. The cell suspension is then transferred to a centrifuge tube, and centrifuged at 200×g at 4° C. for 5 minutes. The supernatant is removed, the cell pellet is resuspended in MC ES medium, and the resuspended cells are plated on four wells of a MEF-coated 24-well plate and cultured for about seven days until a second subculture is prepared.
  • In the second subculture, prepared by the method described above, cells are plated on a 60 mm cell culture dish coated with matrigel at a concentration of 20 μg/cm2. About eight days later (approximately 5 weeks after initiating forced expression of IFs), a third subculture is prepared in which cells are plated on two matrigel-coated 60 mm cell culture dishes, one of which can subsequently be used for gene expression analysis and the other for continued passaging as described below. One of the subcultures is used for gene expression analysis, as described herein, and the other is passaged as needed to maintain a cell line derived from the iPSC clone.
  • G. Passaging and Maintaining Induced Cells
  • After subcloning, the iPSCs may be subcultured about every 5 to 7 days. In some cases, the cells are washed with Hank's balanced salt solution, and dispase or Detachment Medium For Primate ES Cells is added, and incubated at 37° C. for 5 to 10 minutes. When approximately more than half of the colonies are detached, MC-ES medium is added to quench enzymatic activity of the detachment medium, and the resulting cell/colony suspension is transferred to a centrifuge tube. Colonies in the suspension are allowed to settle on the bottom of the tube, the supernatant is carefully removed, and MC-ES medium is then added to resuspend the colonies. After examining the size of the colonies, any extremely large ones are broken up into smaller sizes by slow up and down pipetting. Appropriately sized colonies are plated on a matrigel-coated plastic culture dish with a base area of about 3 to 6 times that before subculture.
  • Examples of culture media useful for culturing human pluripotent stem cells induced from undifferentiated stem cells present in a human postnatal tissue of the present invention include, but are not limited to, the ES medium, and a culture medium suitable for culturing human ES cells such as MEF-conditioned ES medium (MC-ES) or other medium described herein, e.g., mTeSRT™. In some examples, the cells are maintained in the presence of a ROCK inhibitor, as described herein.
  • IV. Analysis of Induced Cells
  • Cell colonies subcultured from those initially identified on the basis of morphological characteristics may be assayed for any of a number of properties associated with pluripotent stem cells, including, but not limited to, expression of alkaline phosphatase activity, expression of ES cell marker genes, expression of protein markers, hypomethylation of Oct3/4 and Nanog promoters relative to a parental cells, long term self-renewal, normal diploid karyotype, and the ability to form a teratoma comprising ectodermal, mesodermal, and endodermal tissues.
  • A number of assays and reagents for detecting alkaline phosphatase activity in cells (e.g., in fixed cells or in living cells) are known in the art. In an exemplary embodiment, colonies to be analyzed are fixed with a 10% formalin neutral buffer solution at room temperature for about 5 minutes, e.g., for 2 to 5 minutes, and then washed with PBS. A chromogenic substrate of alkaline phosphatase, 1 step BCIP (5-Bromo-4-Chloro-3′-Indolylphosphate p-Toluidine Salt) and NBT (Nitro-Blue Tetrazolium Chloride) manufactured by Pierce (Rockford, Ill.) is then added and reacted at room temperature for 20 to 30 minutes. Cells having alkaline phosphatase activity are stained blue-violet.
  • Putative iPS cell colonies tested for alkaline phosphatase activity may be then assayed for expression of a series of human embryonic stem cell marker (ESCM) genes including, but not limited to, Nanog, TDGF1, Dnmt3b, Zfp42, FoxD3, GDF3, CYP26A1, TERT, Oct 3/4, Sox2, Sa114, and HPRT. See, e.g., Assou et al (2007), Stem Cells, 25:961-973. Many methods for gene expression analysis are known in the art. See, e.g., Lorkowski et al (2003), Analysing Gene Expression, A Handbook of Methods: Possibilities and Pitfalls, Wiley-VCH. Examples of suitable nucleic acid-based gene expression assays include, but are not limited to, quantitative RT-PCR (qRT-PCR), microarray hybridization, dot blotting, RNA blotting, RNAse protection, and SAGE.
  • In some embodiments, levels of ESCM gene mRNA expression levels in putative iPS cell colonies are determined by qRT-PCR. Putative iPS cell colonies are harvested, and total RNA is extracted using the “Recoverall total nucleic acid isolation kit for formaldehyde- or paraformaldehyde-fixed, paraffin-embedded (FFPE) tissues” (manufactured by Ambion, Austin, Tex.). In some instances, the colonies used for RNA extraction are fixed colonies, e.g., colonies that have been tested for alkaline phosphatase activity. The colonies can be used directly for RNA extraction, i.e., without prior fixation. In an exemplary embodiment, after synthesizing cDNA from the extracted RNA, the target gene is amplified using the TaqMan® PreAmp mastermix (manufactured by Applied Biosystems, Foster City, Calif.). Real-time quantitative PCR is performed using an ABI Prism 7900HT using the following PCR primer sets (from Applied Biosystems) for detecting mRNA of the above-mentioned ESCM genes: Nanog, Hs02387400_g1, Dnmt3b, Hs00171876_m1, FoxD3, Hs00255287_s1, Zfp42, Hs01938187_s1, TDGF1, Hs02339499_g1, TERT, Hs00162669_m1, GDF3, Hs00220998_m1, CYP26A1, Hs00175627_m1, GAPDH, Hs99999905_ml).
  • Putative iPS cell colonies may be assayed by an immunocytochemistry method for expression of protein markers including, but not limited to, SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, CD9, CD24, Thy-1, and Nanog. A wide range of immunocytochemistry assays, e.g., fluorescence immunocytochemistry assays, are known as described in, e.g., Harlow et al (1988), Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 353-355, and see also, The Handbook—A Guide to Fluorescent Probes and Labeling Technologies, Molecular Probes, Inc., Eugene, Oreg., (2004).
  • In an exemplary embodiment, expression of one or more of the above-mentioned protein markers in putative iPS cell colonies is assayed as follows. Cultured cells are fixed with 10% formaldehyde for 10 min and blocked with 0.1% gelatin/PBS at room temperature for about an hour. The cells are incubated overnight at 4° C. with primary antibodies against SSEA-3 (MC-631; Chemicon), SSEA-4 (MC813-70; Chemicon), TRA-1-60 (ab16288; abcam), TRA-1-81 (ab16289; abcam), CD9 (M-L13; R&D systems), CD24 (ALB9; abcam), Thy1 (5E10; BD Bioscience), or Nanog (MAB1997; R&D Systems). For Nanog staining, cells are permeabilized with 0.1% Triton X-100/PBS before blocking. The cell colonies are washed with PBS three times, then incubated with AlexaFluor 488-conjugated secondary antibodies (Molecular Probes) and Hoechst 33258 (Nacalai) at room temperature for 1 h. After further washing, fluorescence is detected with a fluorescence microscope, e.g., Axiovert 200M microscope (Carl Zeiss).
  • Expression of embryonic stem cell (ESC) marker genes in iPSC colonies may be assayed in live cells, which increases the efficiency of identifying iPSC colonies following an induction method as described herein. Examples of ESC marker genes useful for identifying induced stem cell colonies include, e.g., Oct3/4, Nanog, Klf4, Lin28, Sox2, c-Myc, or TERT. In some embodiments, mRNA for one or more of these genes is detected in live cells. In other embodiments, mRNAs for two or more of the ESC marker genes is detected. In one approach, cells are contacted with one or more molecular beacon probes that hybridize to and signal the presence of one or more stem cell marker genes. Molecular beacons (MBs) are single-stranded oligonucleotide hybridization probes that form a stem-and-loop structure. The loop contains a probe sequence that is complementary to a target sequence, and the stem is formed by the annealing of complementary arm sequences that are located on either side of the probe sequence. A fluorophore is covalently linked to the end of one arm and a quencher is covalently linked to the end of the other arm. MBs do not fluoresce when they are free in solution. However, when they hybridize to a target sequence they undergo a conformational change that enables them to fluoresce brightly. The probe sequence may range in length from about 15 to about 30 nucleotides depending on the GC content of the target probe sequence. Generally, the GC content of the target probe sequence should be from about 40 to about 60%. The flanking stem sequences may range from about 5 to about 7 nucleotides with a GC content of about 75 to about 100 percent. The design of MBs and their use to detect mRNA expression in living cells is known in the art, as described in, e.g., Rhee et al (2008), Nuc Acid Res, 36(5):e30. Useful algorithms for determining melting temperatures of an MB duplex and an MB/target duplex are known in the art. See, e.g., the “Mfold” algorithm described in Zucker (2003), Nuc Acids Res 31(13): 3406-3415, which is public available on a web server: frontend.bioinfo.rpi.edu/applications/mfold/cgi-bin/dna-form1.cgi. See also the Hyther Server at: ozone3.chem.wayne.edu/. Typical parameters for use in these algorithms are 200 nM concentration for beacons and nucleic acid target, a folding temperature of 37° C., and ionic condition of 10 mM KCl and 5 mM MgCl2. The iPSC colonies to be evaluated may be contacted about 14 days to about 50 days after initiating induction, e.g., 14 days to 21 days, 14 days to 28 days, 20 days to 45 days, 25 days to 40 days, 30 days to 35 days, 30 days to 50 days after induction. Preferably, cells are contacted with as low a concentration of an MB and as short a period as compatible with reliably detecting a signal. In some embodiments, the concentration of an MB of about 0.1 μM to about 5 μM (for each MB), e.g., 0.1 μM to 0.5 μM, 0.2 μM to 1 μM, 0.5 μM to 2 μM, or 3 μM to 5 μM. Incubation periods with a MB may range from about 5 minutes to about two hours, e.g., 15 minutes to 30 minutes, 20 minutes to one hour, 30 minutes to 1.5 hours, 45 minutes to 2 hours, or any other time period form about 5 minutes to two hours. In some cases, MBs are introduced into the cells without the use of a transfection reagent. In other cases, a transfection reagent optimized for oligonucleotide transfection is utilized, e.g., TransIT® oligo transfection reagent kit or any other transfection reagents known in the art. In other cases, streptolysin-O is used to transiently permealize cells to allow entry of the MBs into the cells. This method is described in, e.g., Rhee et al supra and Santangelo et al (2004), Nuc Acids Res, 32(6): e57.
  • In some cases, MBs are added to adherent cell cultures and cell colonies found to be positive for expression of one or more ESC marker genes are picked off the substrate as described above. In other cases, MBs are added to iPSCs in suspension and ESC-positive cells are selected by FACS or any other fluorescence based sorting method. Alternatively, MBs are added to adherent iPSCs, which are then dispersed prior to FACS selection. Use of FACS for selection of iPSCs is particularly useful for high throughput generation of iPSC lines and panels of iPSC lines.
  • A. Methylation Analysis
  • In some embodiments, a characteristic of the iPSCs is reduced methylation of the genomic promoters of Oct3/4 and Nanog relative to those of their parental cells. Suitable Oct3/4 promoter regions to be analyzed include, but are not limited to, the Oct3/4 proximal promoter including conserved region 1 (CR1) and the Oct3/4 promoter distal enhancer including CR4. Suitable Nanog promoter regions to be analyzed include, but are not limited to, the Nanog proximal promoter including the Oct3/4 and Sox2 binding sites. See, e.g., Rodda et al (2005), J Biol Chem, 280:24731-24737 and Yang et al (2005), J Cell Biochem, 96:821-830. A number of methods for the quantitative analysis of genomic DNA are known as described in, e.g., Brena et al (2006), J Mol Med, 84(5):365-377. In an exemplary embodiment, genomic DNA isolated from putative iPSCs and cells used for a comparison is isolated and treated with bisulfate. Bisulfite-treated genomic DNA is then PCR-amplified with primers containing a T7 promoter sequence. Afterwards, RNA transcripts are generated using T7 polymerase and then treated with RNAse A to generate methylation-specific cleavage products. Methylation of individual CpG sites is assessed by MALDI-TOF mass spectrometry of the cleavage products. A detailed description of the method is provided in, e.g., Ehich et al (2005), Proc Natl Acad Sci USA, 102:15785-15790.
  • B. Self-Renewal Assay
  • One of the characteristics of stem cells is their ability to proliferate continuously without undergoing senescence. Accordingly, iPSCs are assessed for their ability to be passaged continuously in vitro. In some cases, the iPSCs are assayed for their ability to be passaged for at least about 30 to at least about 100 times in vitro, e.g., about 33, 35, 40, 45, 51, 56, 60, 68, 75, 80, 90, 93, 100, or any other number of passages from at least about 30 to at least about 100 passages.
  • In another evaluation, iPSCs are assayed for their ability to proliferate for a period of about 30 days to about 500 days from initiation of forced expression of IFs in parental cells, e.g., 40 days, 50 days, 60 days, 70 days, 80 days, 100 days, 150 days, 180 days, 200 days, 250 days, 300 days, 400 days, 450 days or any other period from about 30 days to about 500 days from initiation of forced expression of IFs in the parental cells. In some embodiments, long-term self-renewal of iPSCs is determined when the cells are passaged in a defined medium (e.g., mTeSR1 medium) and in the absence of feeder cells, e.g., mTeSR1 medium as described herein. In other embodiments, cells are passaged in MC-ES medium as described herein.
  • C. Karyotype Analysis
  • As another possible analysis, iPSCs are assessed for diploidy and a normal, stable karyotype, e.g., stable after the cells of have been passaged for at least one year in vitro. A number of karotype analysis methods are known in the art. In some embodiments, the karyotype analysis method is multicolor FISH as described in, e.g., Bayani et al (2004), Curr Protoc Cell Biol, Chapter 22: Unit 22.5. In other embodiments, the karyotype analysis includes a molecular karyotype analysis as described in, e.g., Vermeesch et al (2007), Eur J Hum Genet, 15(11):1105-1114. In an exemplary embodiment, iPSCs are pretreated with 0.02 μg/ml colecemid for about 2 to about 3 hours, incubated with about 0.06 to about 0.075M KCl for about 20 minutes, and then fixed with Carnoy's fixative. Afterwards, for multicolor FISH analysis, cells are hybridized with multicolor FISH probes, e.g., those in the Star*FISH© Human Multicolour FISH (M-FISH) Kit from Cambio, Ltd (Cambridge, UK).
  • D. Teratoma Analysis
  • It is generally believed that pluripotent stem cells have the ability to form a teratoma, comprising ectodermal, mesodermal, and endodermal tissues, when injected into an immunocompromised animal. Induced cells or induced pluripotent stem cells (iPS) or ES cell-like pluripotent stem cells may refer to cells having an in vitro long-term self-renewal ability and the pluripotency of differentiating into three germ layers, and said pluripotent stem cells may form a teratoma when transplanted into a test animal such as mouse.
  • The iPSCs may be assessed for pluripotency in a teratoma formation assay in an immunocompromised animal model. The immunocompromised animal may be a rodent that is administered an immunosuppressive agent, e.g., cyclosporin or FK-506. For example, the immunocompromised animal model may be a SCID mouse. About 0.5×106 to about 2.0×106, e.g., 0.6×106, 0.8×106, 1.0×106, 1.2×106, 1.5×106, 1.7×106, or other number of iPSCs from about 0.5×106 to about 2.0×106 iPSCs/mouse may be injected into the medulla of a testis of a 7- to 8-week-old immunocompromised animal. After about 6 to about 8 weeks, the teratomas are excised after perfusing the animal with PBS followed by 10% buffered formalin. The excised teratomas are then subjected to immunohistological analysis. One method of distinguishing human teratoma tissue from host (e.g., rodent) tissue includes immunostaining for the human-specific nuclear marker HuNu. Immunohistological analysis includes determining the presence of ectodermal (e.g., neuroectodermal), mesodermal, and endodermal tissues. Protein markers for ectodermal tissue include, but are not limited to, nestin, GFAP, and integrin β1. Protein markers for mesodermal tissue include, but are not limited to, collagen II, Brachyury, and osteocalcin. Protein markers for endodermal tissue include, but are not limited to, α-fetoprotein (α FP) and HNF3beta.
  • E. Global Gene Expression
  • In some embodiments, global gene expression analysis is performed on putative iPS cell colonies. Such global gene expression analysis may include a comparison of gene expression profiles from a putative iPS cell colony with those of one or more cell types, including but not limited to, (i) parental cells, i.e., one or more cells from which the putative iPS cell colony was induced; (ii) a human ES cell line; or (iii) an established iPS cell line. As known in the art, gene expression data for human ES cell lines are available through public sources, e.g., on the world wide web in the NCBI “Gene Expression Omnibus” database. See, e.g., Barrett et al (2007), Nuc Acids Res, D760-D765. Thus, in some embodiments, comparison of gene expression profiles from a putative iPS colony to those of an ES cell line entails comparison experimentally obtained data from a putative iPS cell colony with gene expression data available through public databases. Examples of human ES cell lines for which gene expression data are publicly available include, but are not limited to, hE14 (GEO data set accession numbers GSM151739 and GSM151741), Sheff4 (GEO Accession Nos GSM194307, GSM194308, and GSM193409), h_ES 01 (GEO Accession No. GSM194390), h_ES H9 (GEO Accession No. GSM194392), and h_ES BG03 (GEO Accession No. GSM194391).
  • It is also possible to accomplish global gene expression by analyzing the total RNA isolated from one or more iPS cell lines by a nucleic acid microarray hybridization assay. Examples of suitable microarray platforms for global gene expression analysis include, but are not limited to, the Human Genome U133 plus 2.0 microarray (Affymetrix) and the Whole Human Genome Oligo Micoarray (Agilent). A number of analytical methods for comparison of gene expression profiles are known as described in, e.g., Suarez-Farinas et al (2007), Methods Mol Biol, 377:139-152, Hardin et al (2007), BMC Bioinformatics, 8:220, Troyanskaya et al (2002), Bioinformatics, 18(11):1454-1461, and Knudsen (2002), A Biologist's Guide to Analysis of DNA Microarray Data, John Wiley & Sons. In some embodiments, gene expression data from cells produced by the methods described herein are compared to those obtained from other cell types including, but not limited to, human ES cell lines, parental cells, and multipotent stem cell lines. Suitable statistical analytical metrics and methods include, but are not limited to, the Pearson Correlation, Euclidean Distance, Hierarchical Clustering (See, e.g., Eisen et al (1998), Proc Natl Acad Sci USA, 95(25): 14863-14868), and Self Organizing Maps (See, e.g., Tamayo et al (1999), Proc Natl Acad Sci USA, 96(6):2907-2912.
  • F. Methods for Differentiating Induced Stem Cell Lines
  • iPSC lines may be differentiated into cell-types of various lineages. Examples of differentiated cells include any differentiated cells from ectodermal (e.g., neurons and fibroblasts), mesodermal (e.g., cardiomyocytes), or endodermal (e.g., pancreatic cells) lineages. The differentiated cells may be one or more: pancreatic beta cells, neural stem cells, neurons (e.g., dopaminergic neurons), oligodendrocytes, oligodendrocyte progenitor cells, hepatocytes, hepatic stem cells, astrocytes, myocytes, hematopoietic cells, or cardiomyocytes.
  • The differentiated cells derived from the iPSCs may be terminally differentiated cells, or they may be capable of giving rise to cells of a specific lineage. For example, iPSCs can be differentiated into a variety of multipotent cell types, e.g., neural stem cells, cardiac stem cells, or hepatic stem cells. The stem cells may then be further differentiated into new cell types, e.g., neural stem cells may be differentiated into neurons; cardiac stem cells may be differentiated into cardiomyocytes; and hepatic stem cells may be differentiated into hepatocytes. Methods for differentiating iPSCs are further disclosed in U.S. application Ser. Nos. 12/157,967, WSGR docket number 36588-704.201; filed Jun. 13, 2008; first inventor Kazuhiro Sakurada, 61/061,594, WSGR Docket Number 36588-707.101; filed on Jun. 13, 2008; First Inventor Kazuhiro Sakurada, and 61/061,565, WSGR Docket Number 36588-702.101; filed on Jun. 13, 2008; First Inventor Kazuhiro Sakurada, which are hereby incorporated by reference in their entirety.
  • There are numerous methods of differentiating the iPSCs into a more specialized cell type. Methods of differentiating iPSCs may be similar to those used to differentiate other stem cells, particularly ES cells, MSCs, MAPCs, MIAMI, hematopoietic stem cells (HSCs). In some cases, the differentiation occurs ex vivo; in some cases the differentiation occurs in vivo.
  • Any known method of generating neural stem cells from ES cells may be used to generate neural stem cells from iPSCs, See, e.g., Reubinoff et al. (2001) Nat. Biotechnol. 19(12):1134-40. For example, neural stem cells may be generated by culturing the iPSCs as floating aggregates in the presence of noggin, or other bone morphogenetic protein antagonist, see e.g., Itsykson et al. (2005) Mol Cell Neurosci. 30(1):24-36. In another example, neural stem cells may be generated by culturing the iPSCs in suspension to form aggregates in the presence of growth factors, e.g., FGF-2, Zhang et al. (2001), Nat. Biotech. (19) 1129-1133. In some cases, the aggregates are cultured in serum-free medium containing FGF-2. In another example, the iPSCs are co-cultured with a mouse stromal cell line, e.g., PA6 in the presence of serum-free medium comprising FGF-2. In yet another example, the iPSCs are directly transferred to serum-free medium containing FGF-2 to directly induce differentiation.
  • Neural stems derived from the iPSCs may be differentiated into neurons, oligodendrocytes, or astrocytes. Dopaminergic neurons play a central role in Parkinson's Disease and are thus of particular interest. In order to promote differentiation into dopaminergic neurons, iPSCs may be co-cultured with a PA6 mouse stromal cell line under serum-free conditions, see, e.g., Kawasaki et al. (2000) Neuron 28(1):31-40. Other methods have also been described, see, e.g., Pomp et al. (2005), Stem Cells 23(7):923-30; U.S. Pat. No. 6,395,546.
  • Oligodendrocytes may also be generated from the iPSCs. For example, oligodendrocytes may be generated by co-culturing iPSCs or neural stem cells with stromal cells, e.g., Lee et al. (2000) Nature Biotechnol 18:675-679. In another example, oligodendrocytes may be generated by culturing the iPSCs or neural stem cells in the presence of a fusion protein, in which the Interleukin (IL)-6 receptor, or derivative, is linked to the IL-6 cytokine, or derivative thereof.
  • Astrocytes may also be produced from the iPSCs. Astrocytes may be generated by culturing iPSCs or neural stem cells in the presence of neurogenic medium with bFGF and EGF, see e.g., Brustle et al. (1999) Science 285:754-756.
  • Induced cells may be differentiated into pancreatic beta cells by methods known in the art, e.g., Lumelsky et al. (2001) Science 292:1389-1394; Assady et al., (2001) Diabetes 50:1691-1697; D′Amour et al (2006) Nat Biotechnol: 1392-1401′ D'Amour et al. (2005) Nat Biotechnol 23:1534-1541. The method may comprise culturing the iPSCs in serum-free medium supplemented with Activin A, followed by culturing in the presence of serum-free medium supplemented with all-trans retinoic acid, followed by culturing in the presence of serum-free medium supplemented with bFGF and nicotinamide, e.g., Jiang et al. (2007) Cell Res 4:333-444. In other examples, the method comprises culturing the iPSCs in the presence of serum-free medium, activin A, and Wnt protein from about 0.5 to about 6 days, e.g., about 0.5, 1, 2, 3, 4, 5, 6, days; followed by culturing in the presence of from about 0.1% to about 2%, e.g., 0.2%, FBS and activin A from about 1 to about 4 days, e.g., about 1, 2, 3, 4 days; followed by culturing in the presence of 2% FBS, FGF-10, and KAAD-cyclopamine (keto-N-aminoethylaminocaproyl dihydro cinnamoylcyclopamine and retinoic acid from about 1 to about 5 days, e.g., 1, 2, 3, 4, or 5 days; followed by culturing with 1% B27, gamma secretase inhibitor and extendin-4 from about 1 to about 4 days, e.g., 1, 2, 3, or 4 days; and finally culturing in the presence of 1% B27, extendin-4, IGF-1, and HGF for from about 1 to about 4 days, e.g., 1, 2, 3, or 4 days.
  • Hepatic cells or hepatic stem cells may be differentiated from the iPSCs. For example, culturing the iPSCs in the presence of sodium butyrate may generate hepatocytes, see e.g., Rambhatla et al. (2003) Cell Transplant 12:1-11. In another example, hepatocytes may be produced by culturing the iPSCs in serum-free medium in the presence of Activin A, followed by culturing the cells in fibroblast growth factor-4 and bone morphogenetic protein-2, e.g., Cai et al. (2007) Hepatology 45(5):1229-39. In an exemplary embodiment, the iPSCs are differentiated into hepatic cells or hepatic stem cells by culturing the iPSCs in the presence of Activin A from about 2 to about 6 days, e.g., about 2, about 3, about 4, about 5, or about 6 days, and then culturing the iPSCs in the presence of hepatocyte growth factor (HGF) for from about 5 days to about 10 days, e.g., about 5, about 6, about 7, about 8, about 9, or about 10 days.
  • The method may also comprise differentiating iPSCs into cardiac muscle cells. In an exemplary embodiment, the method comprises culturing the iPSCs in the presence of noggin for from about two to about six days, e.g., about 2, about 3, about 4, about 5, or about 6 days, prior to allowing formation of an embryoid body, and culturing the embryoid body for from about 1 week to about 4 weeks, e.g., about 1, about 2, about 3, or about 4 weeks.
  • In other examples, cardiomyocytes may be generated by culturing the iPSCs may in the presence of LIF, or by subjecting them to other methods in the art to generate cardiomyocytes from ES cells, e.g., Bader et al. (2000) Circ Res 86:787-794, Kehat et al. (2001) J Clin Invest 108:407-414; Mummery et al. (2003) Circulation 107:2733-2740.
  • Examples of methods to generate other cell-types from iPSCs include: (1) culturing iPSCs in the presence of retinoic acid, leukemia inhibitory factor (LIF), thyroid hormone (T3), and insulin in order to generate adipoctyes, e.g., Dani et al. (1997) J. Cell Sci 110:1279-1285; (2) culturing iPSCs in the presence of BMP-2 or BMP-4 to generate chondrocytes, e.g., Kramer et al. (2000) Mech Dev 92:193-205; (3) culturing the iPSCs under conditions to generate smooth muscle, e.g., Yamashita et al. (2000) Nature 408: 92-96; (4) culturing the iPSCs in the presence of beta-mercaptoethanol to generate keratinocytes, e.g., Bagutti et al. (1996) Dev Biol 179: 184-196; Green et al. (2003) Proc Natl Acad Sci USA 100:15625-15630; (5) culturing the iPSCs in the presence of Interleukin-3(IL-3) and macrophage colony stimulating factor to generate macrophages, e.g., Lieschke and Dunn (1995) Exp Hemat 23:328-334; (6) culturing the iPSCs in the presence of IL-3 and stem cell factor to generate mast cells, e.g., Tsai et al. (2000) Proc Natl Acad Sci USA 97:9186-9190; (7) culturing the iPSCs in the presence of dexamethasone and stromal cell layer, steel factor to generate melanocytes, e.g., Yamane et al. (1999) Dev Dyn 216:450-458; (8) co-culturing the iPSCs with fetal mouse osteoblasts in the presence of dexamethasone, retinoic acid, ascorbic acid, beta-glycerophosphate to generate osteoblasts, e.g., Buttery et al. (2001) Tissue Eng 7:89-99; (9) culturing the iPSCs in the presence of osteogenic factors to generate osteoblasts, e.g., Sottile et al. (2003) Cloning Stem Cells 5:149-155; (10) overexpressing insulin-like growth factor-2 in the iPSCs and culturing the cells in the presence of dimethyl sulfoxide to generate skeletal muscle cells, e.g., Prelle et al. (2000) Biochem Biophys Res Commun 277:631-638; (11) subjecting the iPSCs to conditions for generating white blood cells, e.g., Rathjen et al. (1998) Reprod Fertil Dev 10:31-47; or (12) culturing the iPSCs in the presence of BMP4 and one or more: SCF, FLT3, IL-3, IL-6, and GCSF to generate hematopoietic progenitor cells, e.g., Chadwick et al. (2003) Blood 102:906-915.
  • In some cases, sub-populations of differentiated cells may be purified or isolated. In some cases, one or more monoclonal antibodies specific to the desired cell type are incubated with the cell population and those bound cells are isolated. In other cases, the desired subpopulation of cells expresses a reporter gene that is under the control of a cell type specific promoter.
  • In a specific embodiment, the hygromycin B phosphotransferase-EGFP fusion protein is expressed in a cell type specific manner. The method of purifying comprises sorting the cells to select green fluorescent cells and reiterating the sorting as necessary, in order to obtain a population of cells enriched for cells expressing the construct (e.g., hygromycin B phosphotransferase-EGFP) in a cell-type-dependent manner. Selection of desired sub-populations of cells may also be accomplished by negative selection of proliferating cells with the herpes simplex virus thymidine kinase/ganciclovir (HSVtk/GCV) suicide gene system or by positive selection of cells expressing a bicistronic reporter, e.g., Anderson et al. (2007) Mol. Ther. (11):2027-2036.
  • G. Panels of Induced Stem Cell Lines
  • In some cases, the methods described herein utilize a panel of iPSC lines or a panel of cells differentiated from iPSC lines. A panel of iPSC lines comprises multiple iPSC lines, e.g., iPSC lines, that meet certain selection criteria. Also provided herein are panels of cells differentiated from iPSC lines as described herein. Such panels of differentiated cells include, but are not limited to, panels of neural stem cells, neurons, retinal cells, glial progenitor cells, glial cells, cardiac progenitor cells, cardiomyocytes, pancreatic progenitor cells, pancreatic beta cells, hepatic stem cells, hepatocytes or lung progenitor cells. In some cases, the selection criteria for inclusion of an iPSC line in a panel of iPSC lines are determined prior to generating the iPSC lines that will constitute the panel. In other cases, the selection criteria are applied to iPSC lines generated before hand, e.g., a bank of iPSC lines. Selection criteria include, but are not limited to, the presence or absence of a particular health condition in an iPSC donor, a positive drug response in an iPSC donor, negative, positive, or adverse drug responses in an iPSC donor, the presence or absence of a particular phenotype in an iPSC line or in cells differentiated from the iPSC line, and the presence or absence of one or more polymorphic alleles in the cell lines or their corresponding donors.
  • In some embodiments, where selection criteria include the presence or absence of one or more polymorphic alleles, the panel includes genetically diverse human iPSC lines in which each iPSC line carries at least one polymorphic allele that is unique among the iPSCs to be included in the panel, e.g., 5 to 10, 20 to 50, 50 to 200, 200 to 500, 500 to 1000, 1000 to 5000, 5000 to 20000, or 20000 to 50000 polymorphic alleles that are unique within the panel of iPSC lines. Such polymorphic alleles may include, e.g., a SNP allele, a promoter allele, or a protein-encoding allele. Polymorphic alleles can be screened and scored for by genotyping using any of a number of known genotyping assays. In some cases, the genotyping assay is a multiplexed genotyping assay, e.g., a nucleic acid microarray assay platform such as a “SNP chip.” In some cases, the one or more polymorphic alleles are pre-selected. In some embodiments, the one or more preselected alleles are polymorphic alleles associated with a health condition or a predisposition to a health condition. Examples of polymorphic alleles associated with a health condition or a predisposition to a health condition, include, but are not limited to, polymorphic alleles associated with a neurodegenerative disorder, a neurological disorder, an eye disease, a mood disorder, a respiratory disease, a cardiovascular disease, an immunological disorder, a hematological disease, a metabolic disorder, or a drug sensitivity condition. Some examples of polymorphic alleles associated with a health condition are provided in Table 3 above. Polymorphic alleles may include polymorphic alleles in an encoded protein or a regulatory sequence affecting the expression of the encoded protein. In some cases, the encoded protein is a drug target. Examples of drug target proteins include, but are not limited to, GPCRs, ion channels, kinases, enzymes, and transcription factors.
  • In other embodiments, the one or more polymorphic alleles are pre-selected based on the presence of a high degree of surrounding linkage disequilibrium in the genome, which has been proposed as a signature of genomic loci that are likely to impact many common health conditions. Methods for identifying SNPs having a high surrounding linkage disequilibrium and genes near such SNPs are described in, e.g., Wang et al (2006), Proc Natl Acad Sci USA, 103(1):135-140.
  • In some cases, a panel of iPSC lines includes iPSC lines generated from subjects that are diagnosed as suffering from one or more health conditions. The one or more health conditions may be one or more health conditions that are common to all of the iPSC donors, or they may be health conditions that are different between the iPSC donors.
  • In certain cases, a panel of iPSC lines includes iPSC lines generated from subjects that are both diagnosed as suffering from a health condition and carry a polymorphic allele associated with a health condition, e.g., a polymorphic allele associated with the diagnosed health condition.
  • A panel of iPSC lines may include iPSC lines from at least about 10 individuals to at least about 50,000 individuals, e.g., 10 to 50, 20 to 100, 50 to 250, 100 to 1000, 250, to 2000, 500 to 5000, 1000 to 10,000, 2500 to 20,000, 10,000, to 30,000, 20,000 to 40,000, or 30,000 to 50,000 individuals.
  • A panel of iPSC lines may include iPSC lines from at least two ethnic groups, e.g., 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 20, 25, 30, or 50 ethnic groups. Examples of ethnic groups include, but are not limited to, Europeans, Japanese, Chinese, and the Yoruba of Nigeria, and ethnic groups listed in Table 4.
  • TABLE 4
    Exemplary Ethnic Groups
    Africa
    Bantu
    Biaka
    Mandenka
    Mbuti pygmy
    Mozabite
    San
    Yoruba
    Native America
    Colombian
    Karitiana
    Maya
    Pima
    Surui
    Asia
    Ctrl/South
    Balochi
    Brahui
    Burusho
    Hazara
    Kalash
    Makrani
    Pathan
    Sindhi
    Uyghur
    Western Asia
    Bedouin
    Druze
    Eastern Asia
    Cambodian
    Dai
    Daur
    Han (N. China)
    Han (S. China)
    Hezhen
    Japanese
    Lahu
    Miao
    Mongola
    Naxi
    Oroqen
    She
    Tu
    Tujia
    Xibo
    Yakut
    Yi
    Europe
    Adygei
    Basque
    French
    North Italian
    Orcadian
    Russian
    Sardinian
    Tuscan
    Oceania
    Melanesian
    Papuan
  • IV. Methods for Use of Induced Stem Cell Lines and Panels of Induced Stem Cell Lines A. Overview
  • The iPSC lines and panels of iPSC lines described herein are useful in a number of methods relating to drug discovery and development. Typically, a drug candidate compound will be evaluated in a biochemical assay (e.g., a receptor binding assay) that evaluates only a single or very few sequence variants of the drug target expressed in a patient population. Thus, such assays provide little information as to how effective the drug candidate compound is likely to be in patients that express a drug target allele that differs from the particular drug target allele that was originally screened. Along the same lines, drug candidate compounds often undergo functional cellular screens in one or few cell lines engineered to express a specific allele of the drug target, again ignoring the genetic diversity of a human patient population not only with respect to the drug target itself, but also to that of the various downstream signal transduction proteins that play a role in the response endpoint of cells to a drug. Likewise, adverse effects of candidate drug compounds (e.g., liver toxicity) are generally evaluated in inbred animal models, which are likely to be uninformative for a variable fraction of a human patient population. In contrast, drug screening in panels of genetically diverse iPSC lines, as described herein, addresses the lack of genetic diversity in the prevailing drug screening models.
  • The panels of genetically diverse iPSC lines described herein (e.g., human iPSC lines) or cells differentiated from panels of genetically diverse iPSC lines, as described herein, may be used to identify test compounds that act on a drug target of interest. In some embodiments, the panels of iPSCs cell lines include a sufficient number of iPSC lines such that at least two, e.g., at least 3, 5, 10, 20, 50, 100, or 200 polymorphic alleles of a drug target (e.g., a GPCR, ion channel, or kinase) are represented in the panel. In some embodiments, panels of iPSC lines are derived from subjects diagnosed as suffering from a health condition or identified as having a predisposition to the health condition. In other embodiments, the iPSC line panels comprise iPSC lines each of which that has at least one polymorphic allele associated with a health condition or a predisposition to the health condition.
  • Drug targets for many health conditions are known. Such drug targets may include, but are not limited to, receptors, GPCRs, growth factor receptors, neurotransmitter receptors, ion channels, enzymes, protein kinases, proteases, cytoskeletal proteins, and transcription factors. Test compounds can be assayed for their effect on a drug target by a number of assays known in the art. Such assays include cell-based assays including, but not limited to, assays for determining second messenger levels, e.g., intracellular calcium, cAMP, cGMP, arachidonic acid, and inositol phosphates; channel currents; apoptosis; proliferation; morphological changes; changes in adhesion. Examples of cell-based assays include, but are not limited to those described in, U.S. Pat. Nos. 7,319,009, 7,288,368, and 7,238,213, Cell based assays may also include determining the cellular localization of one or more proteins (e.g., protein kinases, receptors, and transcription factors) in cells in the presence or absence of a test compound. Test compounds may also be screened for their ability to alter a gene expression profile by any gene expression profiling method known in the art. In some cases, the cells to be screened may be genetically modified to express one or more reporter proteins that can indicate activation of a signaling pathway. For example protein-protein interactions between fusion proteins introduced into cells may be detected by a number of methods known in the art, e.g., by fluorescence resonance energy transfer (FRET) or enzyme fragment complementation.
  • In some cases, the mechanistic basis of a sporadic form of a disease is a combination of genetically-determined cell type-specific phenotype and epigenetic factors (e.g., oxidative stress). In other words, iPSC-derived differentiated cells from a patient with a sporadic form of a disease (e.g., Parkinson's) may bear a genetic predisposition to a pathological or pre-pathological cellular phenotype (e.g., apoptosis), but the phenotype may only manifest in vitro in the presence of an appropriate “stressor” that recapitulates environmental/epigenetic factors associated with the sporadic disease or a cellular phenotypes that precede a clinical manifestation of the disease (e.g., mitochondrial dysfunction, oxdidative stress, or nitrosylative stress). Accordingly, in some cases disease-relevant cellular phenotypes are induced by a stressor. Examples of stressors include, but are not limited to cellular oxidative stress, nitrosylative stress, proteasome inhibition, inhibition of mitochondrial electron transport, translation inhibition, decreased calcium buffering, high osmolarity, heat shock, heavy metals (e.g., Zn, Mn, Fe, Cd, Al, or Pb), protein misfolding. Examples of agents that induce, increase, or result from oxidative stress include, but are not limited to, H2O2, ascorbic acid/FeSO4, 4-hydroxynonenal, glutamate, kainate, NMDA, dopamine, okadaic acid, Aβ1-42 and isocyanate. Proteasome inhibitors include, but are not limited to lactacystin, ziram, MG132, and carbobenzoxy-L-isoleucyl-gamma-t-butyl-L-glutamyl-L-alanyl-L-leucinal (PSI). Mitochondrial stressors include, but are not limited to, rotenone, 3-nitropropionic acid (NPA), 1-methyl-4-phenylpyridinium (MPP+), antimycin, paraquat, methylglycoxal, and cyanide. Nitrosylative stressors include, but are not limited to, (+/−)-S-nitroso-N-acetylpenicillamine, sodium nitroprussiate, and peroxynitrite.
  • In some cases, the stressor is provided by expressing or overexpressing an exogenous wild type or mutated gene and/or protein. Examples of such genes include, α-synuclein, amyloid beta, Aβ1-42, Parkin, Pink1, Leucine-rich repeat kinase 2 (LRRK2), superoxide dismutase (SOD).
  • Assays of drug candidate compounds in an iPSC line or a panel of iPSC lines can include determining a dose-response. In some embodiments, the dose response of an iPSC line or that of one or more types of cells differentiated from the iPSC line provides an indication that of the likely efficacy of the compound in the corresponding iPSC donor. In some embodiments, the fraction of iPSC lines in a panel of iPSC lines that exhibit an acceptable dose-response to a test compound indicates an expected probability of an acceptable dose-response relationship in the target patient population of interest. In some cases, cell-based assays of drug candidate include a comparison of responses obtained in a panel of iPSC lines or iPSC-derived cells to one or more reference iPSC lines or cells that serve as a positive or negative control for the effect of a drug candidate compound. The reference iPSC lines or cells may be from a healthy iPSC donor, from an iPSC donor diagnosed as suffering from a health condition, or an iPSC donor carrying a polymorphic allele associated with a health condition. In other embodiments, assays of drug candidate compounds in an iPSC line or a panel of iPSC lines can include determining effective concentrations, maximum tolerated dose and minimum effective concentration. Additional methods and assays are disclosed in U.S. application No. 61/061,594, WSGR Docket Number 36588-707.101; filed Jun. 13, 2008; First Inventor Kazuhiro Sakurada, hereby incorporated by reference.
  • In some cases, the drug screening may be conducted on cells differentiated from iPSCs. Examples of such differentiated cells are described herein (e.g., hepatic cells, neural stem cells, neurons, pancreatic beta cells, cardiomyocytes, hepatic stem cells, oligodendrocytes). The drugs may be targeted to treat a specific disease or condition, e.g., a disease or condition described herein. For example, the iPSCs may be differentiated into dopaminergic neurons, which are used to screen drugs for Parkinson's disease. In other cases, neurons or neural stem cells differentiated from iPSCs may be used to screen drugs for treating Alzheimer's disease, multiple sclerosis, or other neurological disorders. In some cases the In other cases, the iPSCs may be transplanted directly into an immunocompromised animal, e.g., SCID mouse, which is then used to establish in vitro or in vivo assay systems that mimic physiologic conditions in humans or other animals. The in vitro or in vivo assay systems may be used to screen for drugs, e.g., drugs for Parkinson's disease, or as a means to identify biological mechanisms.
  • Screening of test compounds may also be conducted in iPSC-derived cells when an abnormal cellular phenotype (e.g., abnormal cell morphology, gene expression, or signaling), associated with a health condition or a predisposition to the health condition is known, but a drug target has not yet been identified. Such assays may include contacting a test population of iPSC-derived cells from one or more iPSC donors with a test compound and contacting with a negative control compound a negative control population of iPSC-derived cells from the same one or more iPSC donors. The assayed cellular phenotype associated with the health condition of interest in the test and negative control populations can then be compared to a normal cellular phenotype. Where the assayed cellular phenotype in the test population is determined as being closer to a normal cellular phenotype than that exhibited by the negative control population, the drug candidate compound is identified as normalizing the phenotype. A normal cellular phenotype with respect to a particular health condition or a predisposition for a health condition may be established in iPSC-derived cells from iPSC donors that do not suffer from the health condition or a predisposition for the health condition.
  • Test compounds identified as lead compounds, may be tested on a panel of iPSC-derived cells in a manner analogous to a clinical trial. In some cases, the efficacy of the lead compound versus a negative control compound, e.g., a placebo compound is determined in a panel of iPSC-derived cells from patients suffering from the same health condition. Preferably, such a panel of iPSC-derived cells is from subjects that are genetically diverse. For example, such patients may be carry at least one polymorphic allele that is unique among the iPSC-derived cells to be included in the panel, e.g., 5 to 10, 20 to 50, 50 to 200, 200 to 500, 500 to 1000, 1000 to 5000, 5000 to 20000, or 20000 to 50000 polymorphic alleles that are unique within the panel of iPSC lines. A number of methods for quantifying the genetic diversity of a population are known in the art, e.g., the analysis of molecular variance (AMOVA) and generalized analysis of molecular variance (GAMOVA). See, e.g., Excoffier et al (1992), Genetics, 131: 479-491; Nievergelt et al (2008), PLOS Genetics, 3(4):e51. Various clinical experimental designs known in the art may be used for comparing the effect of a lead compound versus a negative control compound. See, e.g., Chow et al (2004) “Design and Analysis of Clinical Trials: Concepts and Methodologies,” John Wiley & Sons, Inc., Hoboken, N.J.
  • The efficacy of the lead compound in iPSC-derived cells may be determined based on any cellular response endpoint, e.g., a response obtained in any of the cell-based assays or gene expression profiling assays mentioned herein.
  • In some cases, potential adverse effects of a lead compound are tested on a panel of iPSC-derived cells. The iPSC-derived cells may include any cell type that hepatocytes, cardiomyocytes, neurons,
  • Drug candidate compounds may be individual small molecules of choice (e.g., a lead compound from a previous drug screen) or in some cases, the drug candidate compounds to be screened come from a combinatorial library, i.e., a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical “building blocks.” For example, a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks. Indeed, theoretically, the systematic, combinatorial mixing of 100 interchangeable chemical building blocks results in the synthesis of 100 million tetrameric compounds or 10 billion pentameric compounds. See, e.g., Gallop et al. (1994), J. Med. Chem. 37(9), 1233. Preparation and screening of combinatorial chemical libraries are well known in the art. Combinatorial chemical libraries include, but are not limited to: diversomers such as hydantoins, benzodiazepines, and dipeptides, as described in, e.g., Hobbs et al. (1993), Proc. Natl. Acad. Sci. U.S.A. 90, 6909; analogous organic syntheses of small compound libraries, as described in Chen et al. (1994), J. Amer. Chem. Soc., 116: 2661; Oligocarbamates, as described in Cho, et al. (1993), Science 261, 1303; peptidyl phosphonates, as described in Campbell et al. (1994), J. Org. Chem., 59: 658; and small organic molecule libraries containing, e.g., thiazolidinones and metathiazanones (U.S. Pat. No. 5,549,974), pyrrolidines (U.S. Pat. Nos. 5,525,735 and 5,519,134), benzodiazepines (U.S. Pat. No. 5,288,514).
  • Numerous combinatorial libraries are commercially available from, e.g., ComGenex (Princeton, N.J.); Asinex (Moscow, Russia); Tripos, Inc. (St. Louis, Mo.); ChemStar, Ltd. (Moscow, Russia); 3D Pharmaceuticals (Exton, Pa.); and Martek Biosciences (Columbia, Md.).
  • B. Individualized Drug Therapy and Failed Drug “Rescue”
  • iPSC cell lines and iPSC-derived cells generated from a subject (e.g., a human subject) can be used to determine the likelihood that a particular drug will have sufficient efficacy in that subject and, if so, an appropriate dose range for that subject. This process is illustrated schematically in FIG. 3. iPSC-derived cells from a subject, e.g., differentiated iPSC-derived cells may be exposed ex vivo to a drug to be tested, and then assayed for their phenotypic response to the drug as described herein. The response of the iPSC-derived cells may be compared to a reference response obtained in iPSC-derived cells from one or more individuals in which the drug has been shown to be effective and/or a reference response in iPSC-derived cells from subjects in which the drug was found to be ineffective. In some cases, the subject to be tested is a subject suffering from a health condition or a predisposition to the health condition. For example, where the subject is suffering from a health condition, and multiple drugs are available to treat the health condition, the efficacies and adverse effects of the multiple drugs may be evaluated iPSC-derived cells from that individual. Preferably, the iPSC-derived cells used to test drug efficacy include cells that express at least one drug target (e.g., a neurotransmitter receptor). In other cases, the subject is not suffering from a health condition. In one embodiment, drugs for various health conditions are tested preemptively in iPSC-derived cells from a healthy subject to establish a pharmaco-phenomic profile for that subject. The pharmaco-phenomic profile may subsequently be used as needed for selecting optimal drugs and drug dosing for treatment of the particular subject.
  • C. Disease Pathway and Target Discovery
  • For many diseases, especially those that have primarily a sporadic form (e.g., Parkinson's disease), the underlying cellular phenotype(s) that precede and eventually result in pathology are unknown. In fact, for progressive degenerative conditions, it is likely that a causative or predictive cellular phenotype occurs well before the first manifestation of symptoms. However, for many types of diseases the relevant cells (e.g., neurons, cardiomyocytes, and pancreatic cells) are not directly accessible for analysis. Thus, depending on the cell type affected by a particular disease, it has not been possible to compare live cells from patients to those of normal subjects in order to identify disease-relevant, cellular phenotypes that cause or predispose for a disease. Identification of reproducible cellular phenotype differences between patient iPSC-derived and normal subject iPSC-derived cells allows the development of screening assays to identify candidate therapeutic agents. Candidate therapeutic agents are those that normalize a disease-associated cellular phenotype, i.e., alter the relevant cellular phenotype in the patient-derived cells so that it is closer to the corresponding cellular phenotype in cells derived from normal subjects under the same conditions. Alternatively, the therapeutic agent may alter a cellular phenotype of \patient-derived iPSCs so as to protect them from a stressor, as described herein.
  • Sets of data representing various cellular phenotypes (e.g., mitochondrial ROS production, expression profiles, protein aggregation) in patient iPSC-derived cells versus normal subject iPSC-derived cells constitute vectors in a multidimensional space, amenable to analysis by means of multivariate and univariate statistical and machine learning techniques. Thus cellular phenotypes distinguishing patient versus normal subject can be identified, for example, by means of univariate statistical methods, such as t-test, ANOVA, regression, as well as their non-parametric analogs. In some embodiments, cellular phenotype data are further filtered using various statistical criteria, e.g., p-value of significance (Type1 error), effect size, etc. Sets of cellular phenotypes which differ significantly between disease and normal states are further scrutinized by biological pathways analysis. In many cases, a pathway enrichment analysis is performed to further narrow the set of cellular phenotypes which are the most disease-informative. A number of statistical procedures such as Hypergeometric statistic, Kolmogorov-Smirnoff test, etc, can be used to perform pathway enrichment analysis.
  • In some cases, cellular phenotypes that are found to differ significantly in patient versus normal subjects (i.e., disease-relevant cellular phenotypes) need to be validated by means of orthogonal assays. In other cases, the identified disease-relevant cellular phenotypes are confirmed by performing validation/cross-validation analysis on the independent data sets from the same type of cellular phenotype assays. In some embodiments, disease-relevant cellular phenotypes are determined by first assaying and analyzing only a portion of the available patient and normal iPSC lines, and then validating disease-relevant cellular phenotypes in the remaining iPSC lines. In other embodiments, where it is not feasible to utilize independent sets of iPSC lines for disease-relevant cellular phenotype discovery versus validation, other statistical approaches, such as k-fold cross-validation techniques, are used instead. In some cases, one or more validated cellular phenotypes is then used to assess test agents for their ability to convert one or more cellular phenotypes reflecting a disease condition to cellular phenotypes reflecting a normal condition.
  • Where the disease under study is a progressive condition with a potentially late onset (e.g., age 60 and over), selection of “normal” control subjects is non-trivial, as it is usually not possible to know, prospectively, who will develop a progressive degenerative disorder. In other words, subjects that are apparently normal at a given age/time point (e.g., when a biopsy is obtained for iPSC derivation) may eventually develop the disease for which an associated cellular phenotype is sought. Thus, cells derived from such a subject would not be a valid “normal” control. Accordingly, in some embodiments, rather than selecting an age-matched normal control subject, a “wellderly” subject is selected for normal control iPSC derivation. As used herein, a “wellderly” subject refers to any subject that is at least 80 years old and has not suffered from any major chronic diseases. Selection of wellderly individuals as normal control subjects makes it statistically less likely that such individuals will go on to develop a degenerative condition. Thus, iPSCs derived from such individuals are less likely to exhibit a cellular phenotype that is associated or predictive of the disease being analyzed, and therefore provide a more reliable “normal control” phenotype for purposes of comparison to patient-derived iPSCs and iPSC-derived cells. In other embodiments, elderly individuals are selected for control iPSC generation that while not having suffered from a degenerative disease under study, may have suffered other unrelated degenerative diseases. In other cases, age-matched subjects free of the disease to be analyzed are used to generate normal control subject iPSCs.
  • In some cases, once a candidate therapeutic agent has been identified as effectively normalizing a cellular phenotype in a small number of patient iPSC lines and cells derived therefrom, efficacy is tested in larger panels of patient iPSC-derived cells to identify potential variation in efficacy or toxicity of the candidate therapeutic agent. In some cases, efficacy is tested in iPSCs or iPSC-derived cells from at least about 20 to about 500 patients, e.g., at least about 25, 30, 40, 50, 60, 70, 100, 200, 250, 300, 400, or another number of patients from at least about 20 to about 500 patients. In some embodiments, biomarkers associated with responsiveness to a candidate therapeutic agent or lack of responsiveness to a candidate therapeutic agent are identified and used to stratify a patient population into, e.g., “high responders” (HR) and “low responders” (LR), as schematized in FIG. 4. In some cases, biomarkers are used to identify suitable patients for clinical trials of a candidate therapeutic agent. In other cases, biomarkers are used to predict the responsiveness or potential toxicity of a therapeutic agent for particular patients. In some cases, biomarkers include genomic biomarkers (e.g., SNPs, a CNVs, or other genetic polymorphisms). In other cases, the biomarkers include an expression profile signature (e.g., an mRNA expression profile). The biomarkers may include a protein expression profile or even a single protein expression level. In some cases, where the biomarkers are expression profile biomarkers, these may be determined directly from a patient sample (e.g., blood, urine, sputum, hair, skin, or other biological sample taken directly from the patient). In other embodiments, expression profile biomarkers, are specific to patient iPSCs or iPSC-derived cells in which the candidate therapeutic agent or therapeutic agent is tested.
  • Thus, in some embodiments, iPSCs are derived from patients and control subjects, and the iPSCs are differentiated into disease-relevant cell types thereby allowing a comparison of cellular phenotypes in patient-derived cells versus normal subject derived cells. For example, the cellular phenotype that is compared may include a mitochondrial phenotype, e.g., ATP synthesis, ATP/ADP ratio, mitochondrial potential, calcium buffering, production of reactive oxygen species, mitochondrial fusion and fission, mitochondrial morphology, and mitochondrial movement. In other cases, the cellular phenotype that is compared is the fraction and rate at which a particular cell type is undergoing apoptosis in the presence of a stressor. In some embodiments, the cellular phenotype is protein aggregation (e.g., the formation of lewy bodies). In other embodiments, gene expression (e.g., microRNA expression) is compared between patient-derived cells and normal subjects.
  • In some cases, relational databases are constructed that integrate multiple data streams relating to each patient and control iPSC line. These data include, but are not limited to, one or more of the following: patient medical history and family medical history, patient medical data (e.g., blood pressure, liver enzyme levels), patient adverse drug reactions, patient drug responsiveness, partial or complete genomic sequence, sequence of all genes with known disease-associated alleles, comprehensive SNP genotypes (e.g., genotypes for all SNPs with known disease associations), gene copy number variation (CNV) polymorphisms, expression profiles for iPSCs and for cells differentiated from the iPSCs (e.g., dopaminergic neurons, cortical neurons, motor neurons, pancreatic cells, hepatocytes, cardiomyocytes, and vascular epithelial cells) under resting and under various stimulus paradigms (e.g., in the presence of a stressor), all cellular phenotype assay data used for initial pathway discovery and for drug screening, including, e.g., cellular phenotype data in the presence or absence of test compounds and compounds with known pharmacological properties (e.g., a cholinesterase inhibitor, a receptor ligand, a kinase inhibitor etc.). In some embodiments, a user can query such a database based on any set of criteria with user define limits. For example, a user may wish to identify polymorphisms associated with patients whose iPSC-derived dopaminergic neurons did or did not respond to a candidate therapeutic agent. In another example, a user may wish to identify a common gene expression profile that distinguishes motor neurons that showed a severe apoptotic response to a stressor versus a mild apoptotic response, etc. Such databases are very useful for data mining and establishing robustly predictive signatures for specific disease states and their response to candidate therapeutic agents.
  • EXAMPLES Example 1 Generation of iPSC Lines from Patients Suffering from Spinal Muscular Atrophy
  • Spinal Muscular Atrophy (SMA) is a neuromuscular disease characterized by degeneration of motor neurons that is among the leading causes of childhood paralysis and mortality. The disease exhibits a wide range of severity affecting infants through adults, and is subdivided into types I-IV based on the age of onset and severity of symptoms: Type I “Infantile” onset at ages 0-6 months and generally fatal); Type II “Intermediate,” onset at ages 7-15 months; inability to stand or walk, but some ability to maintain a sitting position; Type III “Juvenile” onset at ages 18 months to 17 years, with some ability to walk, though potentially transient; Type IV “Adult,” some muscle weakness, but no genetic basis is known.
  • The molecular basis of SMA is linked to the Survival Motor Neuron (SMN) gene. The region of chromosome 5 that contains the SMN (survival motor neuron) gene has a large duplication. A large sequence that contains several genes occurs twice—i.e. once in each of the adjacent segments. The two copies of the gene—known as SMN1 and SMN2-differ by only a few base pairs. The SMN2 gene contains a mutation that occurs at the splice junction of intron 6 to exon 7 resulting in about 90% of SMN2 pre-mRNA transcripts being spliced into a form that excludes exon 7. This shorter mRNA transcript codes for a truncated SMN protein, which is rapidly degraded. About 10% of pre-mRNA transcript from SMN2 is spliced into the full length transcript that codes for the fully functional SMN protein. This splicing defect occurs in multiple cell types, although, for unknown reasons, the survival of motor neurons appear to be particularly affected.
  • SMA results from the loss of the SMN1 gene from both chromosomes, and its severity, ranging from SMA 1 to SMA 3, largely depends on whether the level of SMN2E7 transcript can make up for low levels or absence of exon 7-inclusive SMN 1 transcript. The mutations that cause the loss of SMN 1 are of two types. Deletion mutations, in which both copies of the SMN1 are missing. The other type of mutation is a conversion mutation in which both copies of the SMN1 gene have a point mutation resulting in the same splicing pattern as the SMN2 gene. As an initial step towards developing an in vitro assay for identifying molecules that can increase levels of exon 7-inclusive SMN2 (SMN2E7) transcript, we generated several iPSC lines from Coriell fibroblast lines established from three SMN1−/− SMA patients and from two healthy SMN1−/+ subjects.
  • Induction of iPSCs was initiated by transduction of SMN1−/− and SMN1−/+ fibroblast cultures with four MoMLV VSV-G-pseudotyped viruses for expression of human OCT4, SOX2, KLF4, and c-MYC, each at an MOI of about 10. Five days after viral transduction, fibroblasts were switched from human fibroblast medium into human ES cell supportive medium and monitored daily for the appearance of putative iPSC colonies based on morphological criteria.
  • Initial putative SMA-iPSC colonies were picked after approximately three weeks and propagated clonally in the presence the presence of the ROCK inhibitor Y-27632 (10 μM) Calbiochem) to derive the SMN1−/− iPSC lines SM4p, SM7t, and SM8c, and the SMN1−/+ iPSC lines SM9a and SM10d, as shown in FIG. 5. Each of the iPSCs expressed the pluripotency associated markers, Nanog, Oct4, SSEA3, SSEA4, TRA1-60, and TRA1-81 (data not shown) as determined by immunocytochemistry. Q-PCR analysis showed that these iPSC lines expressed endogenous Oct 4, Sox2, and Klf4, but not the exogenous Oct4, Sox2, and Klf4 introduced by viral transduction. In addition, Q-PCR analysis also demonstrated expression of Nanog, SSEA-3, SSEA-4, TRA1-60, TRA1-81, DNMT3B, FOXD3, LIN28, ZNF206, LEFT2, TDGF1, and TDGF2 in all of the iPSC lines (data not shown). Importantly, all of the SMA iPSC lines were able to form embryoid bodies (EBs) as shown in FIG. 6, which indicated that these lines had good potential for differentiation as is expected for iPSCs. Indeed, the ability of the SM8c line to differentiate into ectodermal, mesodermal, and endodermal lineages in vitro was confirmed by immunostaining for the ectodermal marker Tun, the mesodermal marker Desmin, and the endodermal marker AFP, as shown in FIG. 7. Further, the SM8c iPSC line was shown to differentiate into mature motor neurons as shown by double immunolabeling for Islet and Neuro-N (data not shown).
  • Based on these results, we concluded that iPSCs can be generated from SMA patients and differentiated into motor neurons, as required for the screening assay described in Example 2.
  • Example 2 Assay for Identification of Molecules that Improve Molecular and Cellular Disease Phenotypes in Motor Neurons from Patients Suffering from Spinal Muscular Atrophy
  • We seek to identify molecules that increase the level of SMN2E7 transcript in motor neurons derived from patients suffering from SMA. In principle, increased levels of SMN2E7 transcript can be increased by boosting SMN2 transcription, reducing degradation of SMN2 mRNA, or by increasing the fraction of SMN2 pre-mRNA that is spliced into SMN2E7 mRNA. SMA patient-specific motor neurons are obtained by first generating panels of iPS cell lines from Type I, Type II, and Type III SMA patients, as described in Example 1, and subsequently differentiating iPSCs into motor neurons. Prior to motor neuron differentiation SMA patient SMN2 minigene reporter iPSC lines are established to provide a convenient readout for the level of SMN2E7 transcript in motor neurons.
  • Following parental informed consent, standard dermal punch biopsies 2-4 mm in diameter and thickness are obtained from approximately 30 Type I, 30 Type II, and 30 Type III SMA patients, all of whom have an SMA1−/− genotype, and 10 healthy, age-matched control subjects that have an SMA1−/+ genotype. For each SMA-iPSC line to be generated, the following corresponding patient information is collected and annotated in an iPSC line database: disease severity ranking (i.e., Type I, II, or III), age of disease onset, patient medical history, family medical history including incidence of ALS, blood level of SMN protein, SMN1 and SMN2 genotypes, MUNE Motor Unit Number Estimation, Hammersmith SMA Functional Motor Scale ranking, breathing test evaluation (only for children >5 yrs), symptom progression evaluation (e.g., how outcome of motor tests has changed over time), muscle mass index, description of therapeutic interventions to date, and therapy response. Additional data may be added to each record as they are acquired, including, e.g., SMN protein levels and SMN2E7 transcript levels under various experimental conditions (e.g., in the presence or absence of a candidate therapeutic compound), informative SNP genotypes, genomic sequence, and tissue/cell-type specific expression profiles.
  • Biopsy samples are stored for up to 5-7 days at 4° C. in a “biopsy medium” containing KO-DMEM and supplemented with 10% fetal bovine serum (FBS), Earl's Salts, nucleosides, beta-mercaptoethanol (BME), non-essential amino acids, glutamine, and penicillin/streptomycin. Biopsies are minced into 4-5 pieces, and the pieces are then transferred to a 60 mm dish. The pieces are then “sandwiched” under an acid-washed coverslip and cultured in biopsy medium for five days. Subsequently, the sandwiched biopsy explants are cultured in human fibroblast (“hFib”) medium containing KO-DMEM, Earl's Salts, 10% FBS, glutamine, penicillin/streptomycin, and medium is replaced every 3-4 days until the coverslip is confluent. SMA iPSCs are generated, as described in Example 1, from fibroblasts obtained from each biopsy.
  • An SMN2 splicing minigene reporter construct is generated that incorporates exons 6, 7, and 8, and utilizing the SMN2 promoter is generated essentially as described in Zhang et al (2001), Gene Ther., 8:1532-1538 and Wilson et al, Stem Cells and Development, 16:1027-1041. The SMN2 reporter construct will incorporate the DD-AmCyan1 fluorescent protein reporter to maximize the signal to noise ratio in a compound screening reporter gene assay. The DD-AmCyan1 protein contains a degradation (“DD”) domain that conditionally destabilizes the protein thereby keeping “background” levels of the reporter protein prior to a test compound screening assay very low. However, upon addition of the cell-permeable “Shield1” ligand (Invitrogen), which selectively binds to the DD domain, the reporter protein is stabilized and can therefore accumulate. Thus, potential differences in DD-AmCyan1 reporter levels in the presence or absence of test compounds are maximized by measuring almost exclusively reporter protein produced after the beginning of the screening assay, i.e., after the addition of the Shield1 ligand and test compound. Additional reporter constructs will include AmCyan1 or luciferase as the reporter. Other constructs will include the CMV promoter to drive SMN2 minigene expression. The SMN2 reporter construct is then stably transfected into type I, type II, and type III SMA-iPSCs and healthy control (SMN1WT/WT) iPSCs to generate SMN2-reporter SMA-iPSC lines of varying disease severity backgrounds, and SMN2 reporter control iPSC lines, respectively. Primary screening of test compounds for the ability to increase properly spliced SMN2 transcript levels is conducted initially in motor neurons derived from Type I SMA reporter iPSCs.
  • On day 0, confluent 10 cm plates of SMN2 reporter SMA-iPSCs are trypsinized and then washed/resuspended in embryoid body (EB) medium containing KO DMEM (Invitrogen, catalog #10829-018), Knockout Serum Replacement (Invitrogen, catalog# A1099202), Plasmanate (Talecris), Glutamax (Invitrogen, catalog# 35050079), non-essential amino acids (Invitrogen, cat#11140050). After washing and resuspension, the cells are plated in ultra-low attachment (ULF) 6-well plates and grown into EBs over the next 4-5 days. On day 5, EBs are washed, gently resuspended in EB medium, and replated in a new ULA E-well plate, and the wash/replate procedure is repeated on day 8 or 9. On day 11, EBs are collected and resuspended in N2 base medium (DMEM/F12, Glutamax (Invitrogen, catalog#10565), N-2 Supplement (Invitrogen, catalog#17502-048), D-Glucose (Sigma, catalog # G8769), Ascorbic Acid (Sigma, catalog# A4403-100mG)) supplemented with 1 μM Retinoic Acid (RA) and 100 nM Purmorphamine. (PM). On day 14, EBs are transferred to in N2 Base medium+1 μM RA+1 μM Purmorphamine and replated (3 ml of EB suspension/well) on ULA 6 well plates. N2 base
  • The RA (1 μM)/PM (1 μM)-supplemented EB medium is replaced every 3-5 days, as needed, until approximately day 28. Afterwards, EBs are dissociated by dilute papain treatment and gentle trituration, and then replated on new ULF 6-well plates followed by gentle trituration every 10 minutes over a period of 45 minutes. After dissociation, the resulting cell suspension is collected and transferred to a 50 ml conical tube containing motor neuron maturation medium (DMEM/F12, Glutamax, N-2 Supplement (Invitrogen, catalog#17502-048), B-27 Supplement (Invitrogen, catalog# 17504-044), D-Glucose, Ascorbic Acid (Sigma, catalog# A4403-100mG), 2 ng/mL each GDNF (R&D, catalog#212-GD), BDNF (R&D, catalog#248-BD), and CNTF (R&D, catalog#257-NT/CF). The cell suspension is pelleted by centrifugation at 1000 RPM for five minutes, and is then resuspended in motor neuron maturation medium at a cell concentration of approximately 1.6×106 cells/ml. Aliquots (50 μl) of cell suspension are then plated on laminin-coated wells of optical grade 96 well plates. Beginning on day 31, half-medium changes are conducted every other day or every day depending on how quickly the medium becomes spent. The differentiated cultures are maintained in motor neuron differentiation medium for another four weeks prior to beginning SMN2-reporter assays to allow expansion and maturation of the motor neuron population.
  • At the beginning of the screening assay, all wells of 96-well plate mature motor neuron (MMN) cultures are incubated in the presence of Shield1 is at a final concentration of 1 μM. Test wells are incubated in the presence of test compounds from the NIH Clinical Collection Library (available from BioFocus DPI) at a final concentration of 50 μM. Negative control wells receive no addition or are incubated with a vehicle compound (e.g., DMSO) at a concentration equivalent to that present in some of the test compound solutions. Positive control wells are incubated in the presence of sodium vanadate (50 μM), which has previously been shown to significantly increase levels of SMN2E7 transcript (Zhang et al (2001), Gene Therapy, 8, 1532-1538). After incubation for 24 hours, cultures are fixed and processed for immunofluorescence detection of Islet ½ (mature motor neurons) and Olig2 (motor neuron progenitors) and DD-AmCyan1 fluorescence levels are imaged and quantified in Islet ½+ and Olig2+ cells. Compounds that increase SMN2 reporter levels (“candidate therapeutic” compounds) are screened in secondary assays for their ability to increase SMN2E7 transcript levels and for their ability to promote SMA motor neuron survival over a period of about two weeks. Candidate therapeutic compounds are then tested on motor neurons derived from additional type I SMA SMN2-reporter iPSC lines, and from type II and type III iPSC lines to validate the effect of the therapeutic candidate compounds on motor neurons from diverse genetic backgrounds extant in the SMA patient population.
  • It is expected that identification of compounds that increase the net level SMN2E7 transcript in patient-derived motor neurons is likely to be more relevant for identification of therapeutic drug candidates for SMA than a similar assay in cell types relatively unaffected or less affected by loss of SMN1 (e.g., fibroblasts) or heterologous cell lines.
  • Example 3 Generation of iPSC Lines iPSCs from Patients with Idiopathic Parkinson's Disease and Defined Mutations in Genes Associated with Parkinson's Disease
  • Parkinson's Disease (PD) is one of the most common neurodegenerative diseases of aging, affecting 1-2% of the population over 65 years of age. Clinical symptoms include rest tremor, bradykinesia, and rigidity. We seek to generate a PD patient iPSC model to identify candidate therapeutic agents that slow, halt, or reverse PD progression.
  • iPSC lines are generated from skin biopsies obtained from 10 healthy control subjects with no known family history of PD, 10 patients with sporadic PD, patients each with mutations in the genes that encode α-synuclein (PARK1), parkin (PARK2), PINK 1 (PARK6), or LRRK2 (PARKS) for a total of 10 patients for each mutation. iPSCs are generated as described in Example 1. Afterwards, dopaminergic neurons are derived by differentiating each of the patient iPSC lines and control subject iPSCs. A dopaminergic phenotype is established by immunocytochemical staining for tyrosine hydroxylase positivity, and assaying the differentiated cells for the ability to synthesize and release dopamine. Dopaminergic neurons are obtained by differentiating the iPSCs according to the method of Perrier et al (2004), Proc Natl Acad Sci USA 101, 12543-12548. After validating the dopaminergic phenotype of neurons differentiated from each of the above iPSC lines, cultures of the patient iPSC-derived dopaminergic neurons are tested in a battery of cellular phenotype assays and compared to control subject dopaminergic neurons. These are: assays for aggregation of α-synuclein, dopaminergic neuron apoptosis (TUNEL, caspase activation) and necrosis (CytoTox-Glo), oxidative stress indicators (glutathione levels, ROS, and 4-HNE), and mitochondrial dysfunction (ATP content, membrane potential, morphology, and calcium buffering). It is expected that sporadic forms of PD and PD caused by the above-mentioned mutations will exhibit very similar dopaminergic cellular phenotypes in at least some of these assays. Once this is established, one of more of the PD-associated cellular phenotypes is used as the basis of a screen for candidate therapeutic agents that can reverse or ameliorate these cellular phenotypes. Further, it is expected that as the PD cellular phenotypes are identified in disease relevant cells (dopaminergic neurons) from human PD patients, their predictive value and reliability for the development of therapeutic agents will be more robust than those based on heterologous assay models.
  • While preferred embodiments of the present invention have been shown and described herein, it will be apparent to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (6)

1. A method for identifying an agent that corrects a phenotype associated with a health condition or a predisposition for the health condition, comprising:
(i) contacting a first population of cells from a human induced pluripotent stem cell line, or cells differentiated from the human induced pluripotent stem cell line, with a candidate agent;
(ii) contacting a second population of cells from the human induced pluripotent stem cell line, or cells differentiated from the human induced pluripotent stem cell line, with a negative control agent;
(iii) assaying the phenotype in the first population and second population after the contacting steps; and
(iv) identifying the candidate agent as correcting the phenotype if the assayed phenotype of the first population after the contacting step is closer to a normal phenotype than the phenotype of the second population after the contacting step;
wherein the cells in the first and second populations of human induced pluripotent stem cells:
(a) comprise at least one endogenous allele associated with the health condition or the predisposition for the health condition; or
(b) are generated from a subject suffering from the health condition or the predisposition for the health condition.
2-46. (canceled)
47. A method for evaluating a physiological function of a compound comprising treating cells obtained by inducing differentiation of an induced pluripotent stem (iPS) cell with the compound, wherein the iPS cell is obtained by nuclear reprogramming of a somatic cell, which comprises contacting a nuclear reprogramming factor with the somatic cell to obtain an induced pluripotent stem cell.
48. A method for evaluating the toxicity of a compound comprising treating cells obtained by inducing differentiation of an induced pluripotent stem (iPS) cell with the compound, wherein the iPS cell is obtained by nuclear reprogramming of a somatic cell, which comprises contacting a nuclear reprogramming factor with the somatic cell to obtain an induced pluripotent stem cell.
49. A method for evaluating the effect of a compound on a cellular phenotype comprising treating cells obtained by inducing differentiation of an induced pluripotent stem (iPS) cell with the compound, wherein the iPS cell is obtained by inducing pluripotency of a somatic cell, which comprises contacting an induction factor with the somatic cell to obtain an induced pluripotent stem cell.
50. A method for evaluating the toxicity of a compound comprising treating cells obtained by inducing differentiation of an induced pluripotent stem (iPS) cell with the compound, wherein the iPS cell is obtained by inducing pluripotency of a somatic cell, which comprises contacting an induction factor with the somatic cell to obtain an induced pluripotent stem cell.
US12/709,379 2007-06-15 2010-02-19 Methods and platforms for drug discovery Abandoned US20100240090A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/709,379 US20100240090A1 (en) 2007-06-15 2010-02-19 Methods and platforms for drug discovery

Applications Claiming Priority (14)

Application Number Priority Date Filing Date Title
JPJPO-2007-159382 2007-06-15
JP2007159382A JP2008307007A (en) 2007-06-15 2007-06-15 Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
PCT/EP2007/010019 WO2009006930A1 (en) 2007-06-15 2007-11-20 Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
EPPCT/EP2007/010019 2007-11-20
US4064608P 2008-03-28 2008-03-28
US6159208P 2008-06-13 2008-06-13
US6159408P 2008-06-13 2008-06-13
EPPCT/EP2008/005047 2008-06-13
PCT/IB2008/002540 WO2009007852A2 (en) 2007-06-15 2008-06-13 Multipotent/pluripotent cells and methods
IBPCT/IB2008/002540 2008-06-13
PCT/EP2008/005047 WO2009006997A1 (en) 2007-06-15 2008-06-13 Human pluripotent stem cells and their medical use
US12/157,967 US8211697B2 (en) 2007-06-15 2008-06-13 Induced pluripotent stem cells produced using reprogramming factors and a rho kinase inhibitor or a histone deacetylase inhibitor
US12/484,163 US8257941B2 (en) 2007-06-15 2009-06-12 Methods and platforms for drug discovery using induced pluripotent stem cells
US12/709,379 US20100240090A1 (en) 2007-06-15 2010-02-19 Methods and platforms for drug discovery

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US12/484,163 Continuation US8257941B2 (en) 2007-06-15 2009-06-12 Methods and platforms for drug discovery using induced pluripotent stem cells

Publications (1)

Publication Number Publication Date
US20100240090A1 true US20100240090A1 (en) 2010-09-23

Family

ID=39253880

Family Applications (9)

Application Number Title Priority Date Filing Date
US12/663,840 Abandoned US20110039332A1 (en) 2007-06-15 2007-11-20 Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US12/157,967 Active 2029-09-20 US8211697B2 (en) 2007-06-15 2008-06-13 Induced pluripotent stem cells produced using reprogramming factors and a rho kinase inhibitor or a histone deacetylase inhibitor
US12/465,633 Abandoned US20090304646A1 (en) 2007-06-15 2009-05-13 Multipotent/Pluripotent Cells and Methods
US12/484,163 Active 2028-11-11 US8257941B2 (en) 2007-06-15 2009-06-12 Methods and platforms for drug discovery using induced pluripotent stem cells
US12/564,836 Abandoned US20100120069A1 (en) 2007-06-15 2009-09-22 Multipotent/pluripotent cells and methods
US12/580,216 Abandoned US20100105100A1 (en) 2007-06-15 2009-10-15 Multipotent/pluripotent cells and methods
US12/685,569 Abandoned US20100267135A1 (en) 2007-06-15 2010-01-11 Multipotent/pluripotent cells and methods
US12/709,379 Abandoned US20100240090A1 (en) 2007-06-15 2010-02-19 Methods and platforms for drug discovery
US14/195,698 Active 2028-06-24 US9714433B2 (en) 2007-06-15 2014-03-03 Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue

Family Applications Before (7)

Application Number Title Priority Date Filing Date
US12/663,840 Abandoned US20110039332A1 (en) 2007-06-15 2007-11-20 Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US12/157,967 Active 2029-09-20 US8211697B2 (en) 2007-06-15 2008-06-13 Induced pluripotent stem cells produced using reprogramming factors and a rho kinase inhibitor or a histone deacetylase inhibitor
US12/465,633 Abandoned US20090304646A1 (en) 2007-06-15 2009-05-13 Multipotent/Pluripotent Cells and Methods
US12/484,163 Active 2028-11-11 US8257941B2 (en) 2007-06-15 2009-06-12 Methods and platforms for drug discovery using induced pluripotent stem cells
US12/564,836 Abandoned US20100120069A1 (en) 2007-06-15 2009-09-22 Multipotent/pluripotent cells and methods
US12/580,216 Abandoned US20100105100A1 (en) 2007-06-15 2009-10-15 Multipotent/pluripotent cells and methods
US12/685,569 Abandoned US20100267135A1 (en) 2007-06-15 2010-01-11 Multipotent/pluripotent cells and methods

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/195,698 Active 2028-06-24 US9714433B2 (en) 2007-06-15 2014-03-03 Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue

Country Status (16)

Country Link
US (9) US20110039332A1 (en)
EP (4) EP2213727A1 (en)
JP (2) JP2008307007A (en)
KR (1) KR101657318B1 (en)
CN (1) CN102317448B (en)
AP (1) AP2010005102A0 (en)
AU (1) AU2008273817B2 (en)
CA (1) CA2690629A1 (en)
GB (2) GB2465291B (en)
HK (1) HK1143831A1 (en)
IL (1) IL202678A (en)
MX (1) MX2009013706A (en)
MY (1) MY159971A (en)
NZ (1) NZ582018A (en)
WO (3) WO2009006930A1 (en)
ZA (1) ZA200908904B (en)

Cited By (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090047263A1 (en) * 2005-12-13 2009-02-19 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090068742A1 (en) * 2005-12-13 2009-03-12 Shinya Yamanaka Nuclear Reprogramming Factor
US20090191159A1 (en) * 2007-06-15 2009-07-30 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20090227032A1 (en) * 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090299763A1 (en) * 2007-06-15 2009-12-03 Izumi Bio, Inc. Methods of cell-based technologies
US20100279404A1 (en) * 2008-05-02 2010-11-04 Shinya Yamanaka Method of nuclear reprogramming
US20110016546A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Porcine genome editing with zinc finger nucleases
US20110016539A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genome editing of neurotransmission-related genes in animals
US20110016541A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genome editing of sensory-related genes in animals
US20110016540A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genome editing of genes associated with trinucleotide repeat expansion disorders in animals
US20110016543A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genomic editing of genes involved in inflammation
US20110023141A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved with parkinson's disease
US20110023158A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Bovine genome editing with zinc finger nucleases
US20110023139A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in cardiovascular disease
US20110023150A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of genes associated with schizophrenia in animals
US20110023140A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Rabbit genome editing with zinc finger nucleases
US20110023156A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Feline genome editing with zinc finger nucleases
US20110023151A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of abc transporters
US20110023148A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of addiction-related genes in animals
US20110023145A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in autism spectrum disorders
US20110023154A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Silkworm genome editing with zinc finger nucleases
US20110023149A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in tumor suppression in animals
US20110023143A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of neurodevelopmental genes in animals
US20110023146A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in secretase-associated disorders
US20110023153A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in alzheimer's disease
US20110023144A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in amyotrophyic lateral sclerosis disease
US20110023147A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of prion disorder-related genes in animals
US20110023152A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of cognition related genes in animals
US20110030072A1 (en) * 2008-12-04 2011-02-03 Sigma-Aldrich Co. Genome editing of immunodeficiency genes in animals
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
WO2012092379A2 (en) 2010-12-29 2012-07-05 Sigma-Aldrich Co. Llc Cells having disrupted expression of proteins involved in adme and toxicology processes
WO2014172616A3 (en) * 2013-04-18 2014-12-11 President And Fellows Of Harvard College Methods, compositions and kits for promoting motor neuron survival and treating and diagnosing neurodegenerative disorders
US20150328247A1 (en) * 2012-12-24 2015-11-19 Ramot At Tel-Aviv University Ltd. Agents for treating genetic diseases resulting from nonsense mutations, and methods for identifying the same
WO2017120443A1 (en) * 2016-01-07 2017-07-13 Cedars-Sinai Medical Center Method to identify key markers of human pluripotent cell-derived somatic cells that predict molecular similarity to in vivo target cells
CN108291237A (en) * 2015-10-16 2018-07-17 菲特治疗公司 Platform for inducing and safeguarding ground state versatility
WO2018140657A1 (en) * 2017-01-25 2018-08-02 Whitehead Institute For Biomedical Research Methods for building genomic networks and uses thereof
US10731134B2 (en) 2011-05-27 2020-08-04 Public University Corporation Yokohama City University Production method for artificial cancer stem cell and induced differentiation method therefor
WO2021127594A1 (en) * 2019-12-18 2021-06-24 Editas Medicine, Inc. Engineered cells for therapy
US20220076839A1 (en) * 2018-12-21 2022-03-10 I Peace, Inc. Health risk information management device, health risk information management method, and program

Families Citing this family (337)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7794704B2 (en) 2004-01-23 2010-09-14 Advanced Cell Technology, Inc. Methods for producing enriched populations of human retinal pigment epithelium cells for treatment of retinal degeneration
SG10201606441SA (en) 2004-01-23 2016-09-29 Astellas Inst For Regenerative Medicine Improved modalities for the treatment of degenerative diseases of the retina
EP3354723B1 (en) 2005-08-29 2023-12-13 Technion Research & Development Foundation Ltd. Media for culturing stem cells
WO2007120811A2 (en) 2006-04-14 2007-10-25 Advanced Cell Technology, Inc. Hemangio-colony forming cells
EP3441459B1 (en) 2006-08-02 2021-03-17 Technion Research & Development Foundation Limited Methods of expanding embryonic stem cells in a suspension culture
US8440461B2 (en) 2007-03-23 2013-05-14 Wisconsin Alumni Research Foundation Reprogramming somatic cells using retroviral vectors comprising Oct-4 and Sox2 genes
WO2008156708A2 (en) 2007-06-15 2008-12-24 Massachusetts Institute Of Technology Methods and compositions for enhanced differentiation from embryonic stem cells
JP2011500024A (en) 2007-10-12 2011-01-06 アドバンスド セル テクノロジー, インコーポレイテッド Improved method for generating RPE cells and compositions of RPE cells
AU2008286249B2 (en) 2007-12-10 2013-10-10 Kyoto University Efficient method for nuclear reprogramming
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
US8986696B2 (en) 2007-12-21 2015-03-24 Depuy Mitek, Inc. Trans-capsular administration of p38 map kinase inhibitors into orthopedic joints
US20090226912A1 (en) * 2007-12-21 2009-09-10 Wake Forest University Health Sciences Methods and compositions for correlating genetic markers with prostate cancer risk
WO2009086425A1 (en) * 2007-12-28 2009-07-09 Fate Therapeutics, Inc. Methods for reprogramming cells to a pluripotent state and therapeutic applications related thereto
US20110014164A1 (en) * 2008-02-15 2011-01-20 President And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
SG10202103401QA (en) * 2008-03-17 2021-05-28 Scripps Research Inst Combined chemical and genetic approaches for generation of induced pluripotent stem cells
MX2010012089A (en) 2008-05-06 2011-07-28 Advanced Cell Tech Inc Methods for producing enucleated erythroid cells derived from pluripotent stem cells.
AU2009244231B2 (en) * 2008-05-06 2015-04-09 Astellas Institute For Regenerative Medicine Hemangio colony forming cells and non-engrafting hemangio cells
EP2288726B1 (en) * 2008-05-13 2012-06-13 Gen-Probe Incorporated Inactivatable target capture oligomers for use in the selective hybridization and capture of target nucleic acid sequences
AU2009256202B2 (en) 2008-06-04 2014-07-03 FUJIFILM Cellular Dynamics, Inc. Methods for the production of IPS cells using non-viral approach
WO2010008486A2 (en) 2008-06-24 2010-01-21 Parkinsons Institute Pluripotent cell lines and methods of use thereof
WO2010004989A1 (en) * 2008-07-07 2010-01-14 タカラバイオ株式会社 Method for production of pluripotent stem cell
US20110201110A1 (en) * 2008-07-31 2011-08-18 Gifu University Efficient method for establishing induced pluripotent stem cells
CN107988261A (en) 2008-08-12 2018-05-04 细胞动力国际有限公司 The method for producing IPS cells
WO2010036923A1 (en) * 2008-09-25 2010-04-01 Salk Institute For Biological Studies Induced pluripotent stem cells and methods of use
EP2334804B1 (en) * 2008-10-07 2013-12-04 True North Therapeutics, Inc. Co-culture compositions and methods
EP3450545B1 (en) * 2008-10-24 2023-08-23 Wisconsin Alumni Research Foundation Pluripotent stem cells obtained by non-viral reprogramming
KR101723144B1 (en) 2008-12-17 2017-04-06 더 스크립스 리서치 인스티튜트 Generation and maintenance of stem cells
CN101792776B (en) * 2009-02-01 2013-12-11 中国人民解放军第二军医大学东方肝胆外科医院 Recombinant adenovirus vector for efficiently inducing pluripotent stem cell (PS cell), method for inducing PS cell by using recombinant adenovirus vector and usage of recombinant adenovirus vector
WO2010096746A1 (en) * 2009-02-20 2010-08-26 Cellular Dynamics International, Inc. Methods and compositions for the differentiation of stem cells
EP2401364B1 (en) 2009-02-27 2015-04-22 Cellular Dynamics International, Inc. Differentiation of pluripotent cells
JP5553289B2 (en) * 2009-02-27 2014-07-16 国立大学法人京都大学 New nuclear initialization material
US20120128655A1 (en) * 2009-04-03 2012-05-24 The Mclean Hospital Corporation Induced pluripotent stem cells
RU2399667C1 (en) * 2009-04-10 2010-09-20 Общество С Ограниченной Ответственностью "Лаборатория Клеточных Технологий" Method for preparing pluripotent cells
US20120094304A1 (en) 2009-04-17 2012-04-19 Tohoku University Method of preparing human lung tissue stem cells and method of inducing differentiation into human alveolar epithelial cells
CN101580816B (en) * 2009-04-23 2012-02-29 中国科学院广州生物医药与健康研究院 Novel serum-free culture medium for inducing fast and efficient production of pluripotent stem cells and use method thereof
WO2010131747A1 (en) * 2009-05-15 2010-11-18 国立大学法人 東京大学 Virus-producing cell
EP3150701B1 (en) 2009-06-05 2018-10-03 FUJIFILM Cellular Dynamics, Inc. Reprogramming t cells and hematopoietic cells
US20110044961A1 (en) * 2009-06-19 2011-02-24 Salk Institute For Biological Studies Generation of Induced Pluripotent Stem Cells from Cord Blood
US20120178158A1 (en) * 2009-06-23 2012-07-12 The Scripps Research Institute Adult animals generated from induced pluripotent cells
JP5645197B2 (en) * 2009-06-23 2014-12-24 学校法人日本大学 A novel method for maintaining the undifferentiated state of stem cells
JP2011004674A (en) * 2009-06-26 2011-01-13 Fujitsu Ltd METHOD FOR PRODUCING INDUCED PLURIPOTENT STEM CELL (iPS CELL)
WO2011005326A1 (en) 2009-07-09 2011-01-13 Massachusetts Institute Of Technology Methods and compositions for increased safety of stem cell-derived populations
US9550975B2 (en) 2009-07-15 2017-01-24 Mari Dezawa SSEA-3 pluripotent stem cell isolated from body tissue
WO2011019957A1 (en) * 2009-08-12 2011-02-17 University Of Southern California Method for formation of induced pluripotent stem cells
WO2011021706A1 (en) 2009-08-19 2011-02-24 国立大学法人東北大学 Sheet for corneal transplants
JP2013502220A (en) * 2009-08-21 2013-01-24 ザ ボード オブ トラスティーズ オブ ザ リーランド スタンフォード ジュニア ユニバーシティ Method for improving generation efficiency of induced pluripotent stem cells from human somatic cells
CN102612562A (en) * 2009-08-27 2012-07-25 色奈普提科研究有限公司 A novel protein delivery system to generate induced pluripotent stem (ips) cells or tissue-specific cells
US20120263689A1 (en) * 2009-09-10 2012-10-18 The Salk Institute For Biological Studies Adipose-derived induced pluripotent stem cells
WO2011032166A2 (en) * 2009-09-14 2011-03-17 The Johns Hopkins University Reprogramming blood cells to pluripotent and multipotent stem cells
WO2011038002A1 (en) * 2009-09-22 2011-03-31 Yale University Immunogenic epitopes as targets for universal cancer vaccines
US9422525B2 (en) * 2009-09-30 2016-08-23 Agency For Science, Technology And Research Nuclear receptor and mutant thereof and the use of the same in the reprogramming of cells
EP4206319A1 (en) 2009-10-16 2023-07-05 The Scripps Research Institute Induction of pluripotent cells
AU2010312291A1 (en) 2009-10-29 2012-06-21 Mcmaster University Generating induced pluripotent stem cells and progenitor cells from fibroblasts
SG10201913006XA (en) * 2009-10-30 2020-02-27 Univ North Carolina Chapel Hill Multipotent stem cells from the extrahepatic billary tree and methods of isolating same
US9295697B2 (en) 2009-11-04 2016-03-29 Cellular Dynamics International, Inc. Episomal reprogramming with chemicals
WO2011059920A2 (en) 2009-11-10 2011-05-19 The J. David Gladstone Institutes Methods of generating neural stem cells
ES2932664T3 (en) * 2009-11-12 2023-01-23 Technion Res & Dev Foundation Culture media, cell cultures and methods of culturing pluripotent stem cells in the undifferentiated state
IL301479A (en) 2009-11-17 2023-05-01 Astellas Inst For Regenerative Medicine Methods of producing human rpe cells and pharmaceutical preparations of human rpe cells
CA3115233A1 (en) 2009-12-04 2011-06-09 Astellas Institute For Regenerative Medicine Large scale generation of functional megakaryocytes and platelets from human embryonic stem cells under stromal-free conditions
WO2011071118A1 (en) 2009-12-09 2011-06-16 国立大学法人京都大学 Agent for promoting differentiation of pluripotent stem cells into cardiac muscle cells which includes nitrovin
WO2011096223A1 (en) * 2010-02-03 2011-08-11 独立行政法人国立がん研究センター Induced hepatic stem cell and process for production thereof, and applications of the cell
WO2011096825A1 (en) * 2010-02-05 2011-08-11 Auckland Uniservices Limited Cell programming
EP2363502B1 (en) * 2010-03-04 2017-02-15 miacom Diagnostics GmbH Enhanced multiplex FISH
CN102190731B (en) * 2010-03-09 2016-01-27 中国科学院上海生命科学研究院 Multipotential stem cell is produced with manual transcription factor induction
EP3936608A1 (en) 2010-03-31 2022-01-12 The Scripps Research Institute Reprogramming cells
WO2011130624A2 (en) 2010-04-16 2011-10-20 Immune Disease Institute, Inc. Sustained polypeptide expression from synthetic, modified rnas and uses thereof
US8048675B1 (en) 2010-05-12 2011-11-01 Ipierian, Inc. Integration-free human induced pluripotent stem cells from blood
WO2011143415A1 (en) 2010-05-12 2011-11-17 Abt Holding Company Modulation of splenocytes in cell therapy
US9352003B1 (en) 2010-05-14 2016-05-31 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US8883210B1 (en) 2010-05-14 2014-11-11 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
US10130736B1 (en) 2010-05-14 2018-11-20 Musculoskeletal Transplant Foundation Tissue-derived tissuegenic implants, and methods of fabricating and using same
WO2011146862A1 (en) * 2010-05-21 2011-11-24 Bellicum Pharmaceuticals, Inc. Methods for inducing selective apoptosis
KR20130080444A (en) 2010-05-25 2013-07-12 도쿠리츠교세이호진 고쿠리츠간켄큐센터 Induced malignant stem cells or pre-induction cancer stem cells capable of self-replication outside of an organism, production method for same, and practical application for same
EP2576766A4 (en) * 2010-06-02 2015-03-18 Agency Science Tech & Res Method for inducing pluripotency in human somatic cells with prdm14 or nfrkb
EP2580320B1 (en) 2010-06-14 2018-08-01 The Scripps Research Institute Reprogramming of cells to a new fate
EP2397851B1 (en) 2010-06-21 2015-01-07 Centre d'Etude des Cellules Souches Method for selecting mevalonate synthesis modulators using cells derived from pluripotent human cells
US20120021445A1 (en) * 2010-06-29 2012-01-26 Hogan Kirk J Pluripotent Stem Cells for Drug Induced Myopathy and Malignant Hyperthermia
JP2012029684A (en) * 2010-06-30 2012-02-16 Cell Aid Kenkyusho:Kk Method for producing cell
WO2012012708A1 (en) * 2010-07-22 2012-01-26 Regents Of The University Of Minnesota Induced pluripotent stem cells
PL2596119T3 (en) * 2010-07-23 2021-12-06 Astellas Institute For Regenerative Medicine Methods for detection of rare subpopulations of cells and highly purified compositions of cells
SG187830A1 (en) 2010-08-22 2013-03-28 Univ Ramot Induced pluripotent stem cells derived from human pancreatic beta cells
US9499790B2 (en) 2010-08-26 2016-11-22 Kyoto University Method for promoting differentiation of pluripotent stem cells into cardiac muscle cells
WO2012026491A1 (en) 2010-08-26 2012-03-01 国立大学法人京都大学 Pluripotent stem cell cardiomyocyte differentiation-promoting agent
WO2012027486A2 (en) * 2010-08-26 2012-03-01 The Regents Of The University Of California Methods and compositions for generation of induced pluripotent stem cells by rnaa
US8068011B1 (en) 2010-08-27 2011-11-29 Q Street, LLC System and method for interactive user-directed interfacing between handheld devices and RFID media
EP2611943B1 (en) 2010-09-03 2017-01-04 Wake Forest University Health Sciences Methods and compositions for correlating genetic markers with prostate cancer risk
JP2013545439A (en) 2010-09-17 2013-12-26 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Functional genomics assay to characterize the usefulness and safety of pluripotent stem cells
US9732128B2 (en) 2010-10-22 2017-08-15 Biotime, Inc. Methods of modifying transcriptional regulatory networks in stem cells
US9732319B2 (en) 2010-12-22 2017-08-15 Fate Therapeutics, Inc. Cell culture platform for single cell sorting and enhanced reprogramming of iPSCs
US20120252122A1 (en) * 2011-01-05 2012-10-04 Sangamo Biosciences, Inc. Methods and compositions for increasing production of induced pluripotent stem cells (ipscs)
US9534256B2 (en) 2011-01-06 2017-01-03 Wake Forest University Health Sciences Methods and compositions for correlating genetic markers with risk of aggressive prostate cancer
JP2014506453A (en) 2011-01-19 2014-03-17 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Innate pluripotent somatic cells
WO2012112458A2 (en) * 2011-02-14 2012-08-23 The Regents Of The University Of California Compositions and methods for increasing reprogramming efficiency
US9228204B2 (en) 2011-02-14 2016-01-05 University Of Utah Research Foundation Constructs for making induced pluripotent stem cells
TWI445301B (en) * 2011-03-03 2014-07-11 Realtek Semiconductor Corp Transceiving circuit and transceiving circuit resistance calibration method
US8834928B1 (en) 2011-05-16 2014-09-16 Musculoskeletal Transplant Foundation Tissue-derived tissugenic implants, and methods of fabricating and using same
WO2012159079A1 (en) 2011-05-18 2012-11-22 The Parkinson's Institute Assay to determine lrrk2 activity in parkinson's disease
WO2012174467A2 (en) * 2011-06-15 2012-12-20 Salk Institute For Biological Studies Cord blood-derived neurons by expression of sox2
WO2013013206A1 (en) 2011-07-21 2013-01-24 The Board Of Trustees Of The Leland Stanford Junior University Cardiomyocytes from induced pluripotent stem cells from patients and methods of use
EP2737064B1 (en) 2011-07-25 2017-12-06 Kyoto University Method for screening induced pluripotent stem cells
WO2013018851A1 (en) * 2011-08-02 2013-02-07 独立行政法人国立がん研究センター Method for inducing hepatic differentiation from induced hepatic stem cell, and induced hepatic progenitor cell
EP2756092A4 (en) * 2011-09-16 2015-10-28 Giovanni Amabile Methods of making cells, tissues, and antibodies
CN102329769B (en) * 2011-09-28 2013-07-31 浙江大学 Method for obtaining hematopoietic stem cells
EP2766474B1 (en) 2011-10-14 2020-10-07 Children's Medical Center Corporation Inhibition and enhancement of reprogramming by chromatin modifying enzymes
JP6162604B2 (en) 2011-10-21 2017-07-12 国立大学法人京都大学 Multi-dispersed monodisperse cell culture method by laminar flow
GB2496375A (en) 2011-10-28 2013-05-15 Kymab Ltd A non-human assay vertebrate comprising human antibody loci and human epitope knock-in, and uses thereof
JP5999658B2 (en) 2011-11-25 2016-09-28 国立大学法人京都大学 Method for culturing pluripotent stem cells
WO2013081188A1 (en) 2011-11-30 2013-06-06 独立行政法人国立がん研究センター Induced malignant stem cells
US10428309B2 (en) 2011-12-01 2019-10-01 New York Stem Cell Foundation, Inc. Systems and methods for producing stem cells and differentiated cells
KR20240019378A (en) 2011-12-01 2024-02-14 뉴욕 스템 셀 파운데이션, 인코포레이티드 Automated system for producing induced pluripotent stem cells or differentiated cells
GB201122047D0 (en) 2011-12-21 2012-02-01 Kymab Ltd Transgenic animals
AU2012347919B2 (en) * 2011-12-05 2017-02-02 Factor Bioscience Inc. Methods and products for transfecting cells
EP2808383B1 (en) 2012-01-27 2018-07-25 Kyoto University Method for inducing cardiac differentiation of pluripotent stem cell
IN2014DN08862A (en) * 2012-03-28 2015-05-22 Quarrymen Corp
US9447378B2 (en) 2012-04-27 2016-09-20 Massachusetts Institute Of Technology Method for differentiating human embryonic stem cells into β-cells for the treatment of type I diabetes
US10119150B2 (en) * 2012-05-13 2018-11-06 Allele Biotechnology & Pharmaceuticals, Inc. Feeder-free Derivation of human-induced pluripotent stem cells with synthetic messenger RNA
US10155929B2 (en) * 2012-05-13 2018-12-18 Allele Biotechnology & Pharmaceuticals, Inc. Feeder-free derivation of human-induced pluripotent stem cells with synthetic messenger RNA
PL2800811T3 (en) 2012-05-25 2017-11-30 Emmanuelle Charpentier Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
EP2872181B1 (en) 2012-07-11 2020-11-18 Tissuetech, Inc. Compositions containing hc-ha/ptx3 complexes and methods of use thereof
US9175263B2 (en) * 2012-08-22 2015-11-03 Biotime, Inc. Methods and compositions for targeting progenitor cell lines
WO2014031085A1 (en) * 2012-08-24 2014-02-27 Agency For Science, Technology And Research The use of pdgf-bb in a method of enhancing the efficiency of reprogramming of a somatic cell to a pluripotent stem cell
US20150268254A1 (en) * 2012-10-05 2015-09-24 Children's Hospital Medical Center Sebocyte cell culturing and methods of use
US20150313947A1 (en) * 2012-12-03 2015-11-05 Anthrogenesis Corporation Treatment of schizophrenia using amnion derived adherent cells
CN105101979B (en) 2012-12-21 2021-10-08 安斯泰来再生医药协会 Method for preparing platelets from pluripotent stem cells and composition thereof
JP6433902B2 (en) * 2013-01-04 2018-12-05 国立大学法人京都大学 Reprogrammed stem cells
US9709554B2 (en) 2013-01-31 2017-07-18 Rutgers, The State University Of New Jersey In vitro model of macrosteatotic (fatty) liver
JP6541577B2 (en) 2013-02-06 2019-07-10 ユニバーシティー オブ ロチェスター Induced pluripotent cell-derived oligodendrocyte precursor cells for the treatment of myelin disorders
BR112015018923B1 (en) * 2013-02-06 2022-03-03 Nc Medical Research Inc In vitro methods of producing heterogeneous subpopulation of bone marrow cells and to optimize experimental protocol to isolate cell population that treats neurodegeneration caused by ischemic stroke
US20160002599A1 (en) 2013-02-08 2016-01-07 Kyoto University Production methods for megakaryocytes and platelets
US10160950B2 (en) 2013-03-01 2018-12-25 Wisconsin Alumni Research Foundation Methods of maintaining, expanding and differentiating neuronal subtype specific progenitors
US20140248696A1 (en) * 2013-03-01 2014-09-04 Wisconsin Alumni Research Foundation Methods of maintaining, expanding, and diffrentiating neuronal subtype specific progenitors
EP2966163B1 (en) 2013-03-06 2018-01-17 Kyoto University Culture system for pluripotent stem cells and method for subculturing pluripotent stem cells
WO2014136519A1 (en) 2013-03-08 2014-09-12 国立大学法人京都大学 Promoter of differentiation of pluripotent stem cell into myocardium, which comprises egf receptor inhibitor
US9434935B2 (en) 2013-03-10 2016-09-06 Bellicum Pharmaceuticals, Inc. Modified caspase polypeptides and uses thereof
WO2014143383A1 (en) 2013-03-13 2014-09-18 Agilent Technologies, Inc. Transposome tethered to a gene delivery vehicle
JP2016520291A (en) 2013-03-14 2016-07-14 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Production of medial ganglion progenitor cells in vitro
US20160237400A1 (en) * 2013-03-15 2016-08-18 The Jackson Laboratory Isolation of non-embryonic stem cells and uses thereof
SG11201507515YA (en) * 2013-03-15 2015-10-29 Marcus Kare Torleif Larsson Cells, methods and apparatuses for umbilical cord blood collection and isolation of cells
US10519421B2 (en) 2013-03-21 2019-12-31 Kyoto University Induction of motor neurons from pluripotent stem cells
US10072242B2 (en) 2013-03-25 2018-09-11 Foundation For Biomedical Research And Innovation At Kobe Cell sorting method
JP6461787B2 (en) 2013-04-12 2019-01-30 国立大学法人京都大学 Method for inducing alveolar epithelial progenitor cells
WO2014185358A1 (en) 2013-05-14 2014-11-20 国立大学法人京都大学 Efficient myocardial cell induction method
EP3006559B1 (en) 2013-05-31 2019-11-06 iHeart Japan Corporation Layered cell sheet incorporating hydrogel
US9913882B2 (en) 2013-06-05 2018-03-13 Bellicum Pharmaceuticals, Inc. Methods for inducing partial apoptosis using caspase polypeptides
CA2914615C (en) 2013-06-05 2023-10-17 Biotime, Inc. Compositions and methods for induced tissue regeneration in mammalian species
JP6697380B2 (en) 2013-06-10 2020-05-20 プレジデント・アンド・フェロウズ・オブ・ハーバード・カレッジ Early developmental genomic assay to characterize the utility and safety of pluripotent stem cells
SG11201510201PA (en) 2013-06-11 2016-01-28 Univ Kyoto Method for producing renal progenitor cells and drug comprising the same
EP3009503B1 (en) * 2013-06-12 2019-08-07 Shiseido Company Ltd. Serum-free medium containing pdgf for ds cells
US9816070B2 (en) 2013-06-14 2017-11-14 Massachusetts Institute Of Technology Articles and methods for stem cell differentiation
WO2014210533A1 (en) * 2013-06-27 2014-12-31 The New York Stem Cell Foundation Improved systems and methods for producing stem cells and differentiated cells
WO2015006725A2 (en) 2013-07-12 2015-01-15 Cedars-Sinai Medical Center Generation of induced pluripotent stem cells from normal human mammary epithelial cells
US20150024406A1 (en) * 2013-07-17 2015-01-22 Robin N. Coger-Simmons Increased aquaporin expression on cellular membrane to improve cryopreservation efficiency
CA2919374C (en) 2013-07-30 2019-12-03 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US9796962B2 (en) 2013-08-07 2017-10-24 Kyoto University Method for generating pancreatic hormone-producing cells
DK2840132T3 (en) 2013-08-22 2017-02-20 Georg-August-Universität Göttingen Stiftung Öffenlichen Rechts Universitätsmedizin Method of Manufacturing Constructed Heart Muscle (EHM)
MY184219A (en) 2013-09-05 2021-03-26 Univ Kyoto New method for inducing dopamine-producing neural precursor cells
US20160222347A1 (en) 2013-09-12 2016-08-04 Kaneka Corporation Method for inducing differentiation of induced pluripotent stem cells and method for selecting the same
EP3045451B1 (en) 2013-09-13 2018-03-28 Kyoto University Compound promoting differentiation of pluripotent stem cells into cardiomyocytes
EP3047019B1 (en) 2013-09-20 2019-07-03 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts Universitätsmedizin A method to direct differentiation of pluripotent stem cells into functional heart muscle
CN104450609B (en) * 2013-09-25 2017-07-14 深圳华大基因科技有限公司 A kind of method for separating and cultivating umbilical cord mesenchymal stem cells
WO2015058117A1 (en) * 2013-10-18 2015-04-23 Icahn School Of Medicine At Mount Sinai Directed cardiomyocyte differentiation and ventricular specification of stem cells
EP3060649A4 (en) * 2013-10-23 2017-07-12 The J. David Gladstone Institutes, A Testamentary Trust Established under The Will of J. David Gladstone Reprogramming cardiomyocytes with one transcription factor
WO2015064754A1 (en) 2013-11-01 2015-05-07 国立大学法人京都大学 Novel chondrocyte induction method
WO2015069736A1 (en) 2013-11-08 2015-05-14 The Mclean Hospital Corporation METHODS FOR EFFICIENT GENERATION OF GABAergic INTERNEURONS FROM PLURIPOTENT STEM CELLS
US9932607B2 (en) 2013-11-15 2018-04-03 The Board Of Trustees Of The Leland Stanford Junior University Site-specific integration of transgenes into human cells
US20160298096A1 (en) 2013-11-18 2016-10-13 Crispr Therapeutics Ag Crispr-cas system materials and methods
AU2014363032A1 (en) 2013-12-11 2016-06-09 Pfizer Limited Method for producing retinal pigment epithelial cells
EP3080266B1 (en) 2013-12-12 2021-02-03 The Regents of The University of California Methods and compositions for modifying a single stranded target nucleic acid
US9512406B2 (en) 2013-12-20 2016-12-06 The J. David Gladstone Institute, a testamentary trust established under the Will of J. David Gladstone Generating hepatocytes
US11078462B2 (en) 2014-02-18 2021-08-03 ReCyte Therapeutics, Inc. Perivascular stromal cells from primate pluripotent stem cells
CN106414721A (en) 2014-03-04 2017-02-15 菲特治疗公司 Improved reprogramming methods and cell culture platforms
WO2015143125A1 (en) * 2014-03-19 2015-09-24 The Brigham And Women's Hospital, Inc. Methods relating to pluripotent cells
WO2015173652A1 (en) 2014-05-14 2015-11-19 Mark Davies Method for testing compounds on living cells
US10233426B2 (en) 2014-05-30 2019-03-19 Kyoto University Method for inducing cardiac differentiation of pluripotent stem cell with low-molecular compounds
US20170107498A1 (en) * 2014-06-05 2017-04-20 Cedars-Sinai Medical Center Novel and efficient method for reprogramming immortalized lymphoblastoid cell lines to induced pluripotent stem cells
US10240127B2 (en) 2014-07-03 2019-03-26 ReCyte Therapeutics, Inc. Exosomes from clonal progenitor cells
EP3170901B1 (en) 2014-07-14 2021-06-02 Chugai Seiyaku Kabushiki Kaisha Method for producing dendritic cells from stem cells
US10570418B2 (en) 2014-09-02 2020-02-25 The Regents Of The University Of California Methods and compositions for RNA-directed target DNA modification
WO2016061298A1 (en) * 2014-10-15 2016-04-21 Coyne Ip Holdings, Llc Methods for conducting stimulus-response studies with induced pluripotent stem cells derived from perinatal cells or tissues
EP3215522B1 (en) 2014-11-03 2021-12-01 Academisch Ziekenhuis Leiden H.O.D.N. Leids Universitair Medisch Centrum T cell receptors directed against bob1 and uses thereof
WO2016085765A1 (en) 2014-11-25 2016-06-02 President And Fellows Of Harvard College Methods for generation of podocytes from pluripotent stem cells and cells produced by the same
WO2016088243A1 (en) * 2014-12-05 2016-06-09 株式会社ニコン Determination device, observation system, observation method, program for same, method for manufacturing cell, and cell
JP7253692B2 (en) 2014-12-26 2023-04-07 国立大学法人京都大学 Hepatocyte induction method
EP3256585A4 (en) 2015-02-13 2018-08-15 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
JP2018510649A (en) 2015-02-17 2018-04-19 ユニバーシティー ヘルス ネットワーク Methods for making and using sinoatrial node-like pacemaker cardiomyocytes and ventricular cardiomyocytes
JP2016202172A (en) 2015-04-16 2016-12-08 国立大学法人京都大学 Production method of pseudoislet
US9724432B2 (en) 2015-04-30 2017-08-08 University Of Rochester Non-human mammal model of human degenerative disorder, uses thereof, and method of treating human degenerative disorder
CA3177726A1 (en) 2015-05-21 2016-11-24 Musculoskeletal Transplant Foundation Modified demineralized cortical bone fibers
RU2725737C2 (en) * 2015-05-29 2020-07-03 Регенерон Фармасьютикалс, Инк. Non-human animals with disturbance in locus c9orf72
EP3319632A4 (en) * 2015-07-06 2019-05-29 University of Georgia Research Foundation Inc. Methods and compositions related to increased influenza virus production
US10912864B2 (en) 2015-07-24 2021-02-09 Musculoskeletal Transplant Foundation Acellular soft tissue-derived matrices and methods for preparing same
US11020463B2 (en) 2015-08-07 2021-06-01 The J. David Gladstone Institutes Method of determining cellular protein homeostasis
US11052175B2 (en) 2015-08-19 2021-07-06 Musculoskeletal Transplant Foundation Cartilage-derived implants and methods of making and using same
JP7370529B2 (en) * 2015-08-31 2023-10-30 剛士 田邊 Pluripotent stem cell production system, method for inducing stem cells, suspension culture method for stem cells, suspension culture vessel for stem cells, method for producing induced pluripotent stem cells, and method for producing specific somatic cells from animal cells
HUE057135T2 (en) 2015-09-01 2022-04-28 Ncardia B V An in vitro method of differentiating a human pluripotent stem cell population into a cardiomyocyte cell population
AU2016339053A1 (en) 2015-09-24 2018-04-12 Crispr Therapeutics Ag Novel family of RNA-programmable endonucleases and their uses in genome editing and other applications
JP6781752B2 (en) * 2015-10-08 2020-11-04 高雄醫學大學Kaohsiung Medical University Composition that rapidly separates fatty stromal cells
WO2017079029A1 (en) * 2015-11-02 2017-05-11 ORIG3N Inc. Cell cycle block improves efficiency in generating induced pluripotent stem cells
WO2017100683A1 (en) * 2015-12-09 2017-06-15 Stemgenics, Inc. Differential drug screening using pluripotent stem cells induced with functionalized nanoparticles
JOP20200228A1 (en) 2015-12-21 2017-06-16 Novartis Ag Compositions and methods for decreasing tau expression
WO2017136479A1 (en) 2016-02-01 2017-08-10 Cedars-Sinai Medical Center Systems and methods for growth of intestinal cells in microfluidic devices
US20190249172A1 (en) 2016-02-18 2019-08-15 The Regents Of The University Of California Methods and compositions for gene editing in stem cells
WO2017159862A1 (en) 2016-03-18 2017-09-21 国立大学法人京都大学 Freezing method for aggregates of pluripotent stem cell-derived myocardial cells
US11773375B2 (en) 2016-03-25 2023-10-03 Ncardia B.V. In vivo method for differentiating human pluripotent stem cells into atrial cardiomyocytes
ES2886631T3 (en) 2016-04-15 2021-12-20 Univ Kyoto Method for Inducing Antigen-Specific CD8-Positive T Cells
CN109072198B (en) 2016-04-22 2022-08-26 国立大学法人京都大学 Preparation method of dopamine-producing neural progenitor cells
WO2017196175A1 (en) 2016-05-12 2017-11-16 Erasmus University Medical Center Rotterdam A method for culturing myogenic cells, cultures obtained therefrom, screening methods, and cell culture medium.
US10221395B2 (en) 2016-06-16 2019-03-05 Cedars-Sinai Medical Center Efficient method for reprogramming blood to induced pluripotent stem cells
US11572545B2 (en) 2016-06-16 2023-02-07 Cedars-Sinai Medical Center Efficient method for reprogramming blood to induced pluripotent stem cells
WO2017216771A2 (en) 2016-06-17 2017-12-21 Genesis Technologies Limited Crispr-cas system, materials and methods
US11103537B2 (en) 2016-06-23 2021-08-31 Tithon Biotech Inc. Cells expressing parathyroid hormone 1 receptor and uses thereof
RU2771328C2 (en) * 2016-06-23 2022-04-29 Титон Биотек, Инк. Cells expressing parathyroid hormone 1 receptor, and their use
US10576167B2 (en) 2016-08-17 2020-03-03 Factor Bioscience Inc. Nucleic acid products and methods of administration thereof
WO2018073615A1 (en) 2016-10-21 2018-04-26 Longboat Explorers Ab Methods and compositions for generating hematopoietic cells
WO2018124118A1 (en) 2016-12-27 2018-07-05 住友化学株式会社 Evaluation method and selection method for induced pluripotent stem cells, and production method for induced pluripotent stem cells
US20190367877A1 (en) 2017-01-20 2019-12-05 Kyoto University METHOD FOR PRODUCING CD8a+b+ CYTOTOXIC T CELLS
WO2018140647A1 (en) 2017-01-25 2018-08-02 Cedars-Sinai Medical Center In vitro induction of mammary-like differentiation from human pluripotent stem cells
WO2018139548A1 (en) 2017-01-26 2018-08-02 国立大学法人大阪大学 Medium for inducing differentiation of stem cells into mesodermal cells and method for producing mesodermal cells
US10828330B2 (en) 2017-02-22 2020-11-10 IO Bioscience, Inc. Nucleic acid constructs comprising gene editing multi-sites and uses thereof
JPWO2018159805A1 (en) 2017-03-03 2020-01-09 国立大学法人京都大学 Method for producing pancreatic progenitor cells
WO2018170139A1 (en) * 2017-03-14 2018-09-20 EMULATE, Inc. Neuromuscular junction: nmj-on-chip
EP3597734A4 (en) 2017-03-14 2021-03-03 Kyoto University Method for producing helper t cells from pluripotent stem cells
US11767513B2 (en) 2017-03-14 2023-09-26 Cedars-Sinai Medical Center Neuromuscular junction
US11414648B2 (en) 2017-03-24 2022-08-16 Cedars-Sinai Medical Center Methods and compositions for production of fallopian tube epithelium
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US11821007B2 (en) 2017-05-25 2023-11-21 Kyoto University Method for inducing differentiation of intermediate mesodermal cell to renal progenitor cell, and method for inducing differentiation of pluripotent stem cell to renal progenitor cell
WO2018230588A1 (en) 2017-06-14 2018-12-20 武田薬品工業株式会社 Cell-sealing device
JP6758631B2 (en) 2017-06-19 2020-09-23 国立大学法人大阪大学 Corneal endothelial cell marker and its use
JP6736772B2 (en) 2017-06-19 2020-08-05 公益財団法人神戸医療産業都市推進機構 Method for predicting differentiation potential of pluripotent stem cells and reagent therefor
NL2019517B1 (en) 2017-09-08 2019-03-19 Univ Erasmus Med Ct Rotterdam New therapy for Pompe disease
WO2019078263A1 (en) 2017-10-17 2019-04-25 国立大学法人京都大学 Method for obtaining artificial neuromuscular junction from pluripotent stem cells
MA50579A (en) 2017-11-09 2020-09-16 Crispr Therapeutics Ag AUTO-INACTIVATION (INS) CRISPR / CAS OR CRISPR / CPF1 SYSTEMS AND THEIR USES
KR20200087201A (en) 2017-11-15 2020-07-20 셈마 테라퓨틱스, 인크. Islet cell production composition and method of use
CA3083253A1 (en) 2017-11-30 2019-06-06 Kyoto University Method for culture of cells
KR102137884B1 (en) * 2017-12-12 2020-07-24 순천향대학교 산학협력단 High-efficiency cell culture medium additive including tauroursodeoxycholic acid
KR102137883B1 (en) * 2017-12-12 2020-07-24 순천향대학교 산학협력단 High-efficiency cell culture medium additive including sodium phenylbutyrate
WO2019118935A1 (en) 2017-12-14 2019-06-20 Casebia Therapeutics Limited Liability Partnership Novel rna-programmable endonuclease systems and their use in genome editing and other applications
TWI821230B (en) 2017-12-22 2023-11-11 日商千紙鶴治療公司 Cell culture device, culture medium aspirator and cell culture method
WO2019165320A1 (en) * 2018-02-22 2019-08-29 Celularity, Inc. Post partum tissue-derived induced pluripotent stem cells and uses thereof
EP3768834A1 (en) 2018-03-19 2021-01-27 CRISPR Therapeutics AG Novel rna-programmable endonuclease systems and uses thereof
JPWO2019182157A1 (en) 2018-03-19 2021-03-18 国立大学法人京都大学 Hydrogel capsule
KR102209108B1 (en) * 2018-03-27 2021-01-28 국립암센터 A composition for inhibiting stemness comprising peptides for blocking the function of OCT4
JP7440868B2 (en) 2018-03-30 2024-02-29 国立大学法人京都大学 Cell manufacturing method
CN111918866B (en) 2018-03-30 2023-07-04 千纸鹤治疗公司 Heterocyclic compounds
WO2019189554A1 (en) 2018-03-30 2019-10-03 国立大学法人京都大学 Cardiomyocyte maturation promoter
WO2019208505A1 (en) 2018-04-23 2019-10-31 国立大学法人京都大学 Growth inhibitor
US20210254006A1 (en) 2018-06-06 2021-08-19 Ideaya Biosciences, Inc. Methods of culturing and/or expanding stem cells and/or lineage committed progenitor cells using lactam compounds
WO2019241620A1 (en) * 2018-06-14 2019-12-19 Academia Sinica Method to generate induced oligodendrocyte-lineage cells and treatment using such cells
CA3103663A1 (en) 2018-06-18 2019-12-26 University Of Rochester Inhibition of re1-silencing transcription factor in the treatment of schizophrenia and other neuropsychiatric disorders
US20220062378A1 (en) 2018-06-21 2022-03-03 University Of Rochester Methods of treating or inhibiting onset of huntington's disease
SG11202100260QA (en) 2018-07-13 2021-02-25 Univ Kyoto METHOD FOR PRODUCING γδ T CELLS
JP7285015B2 (en) 2018-07-19 2023-06-01 国立大学法人京都大学 Lamellar cartilage derived from pluripotent stem cells and method for producing the same
WO2020022261A1 (en) 2018-07-23 2020-01-30 国立大学法人京都大学 Novel renal progenitor cell marker and method for concentrating renal progenitor cells using same
SG11202101098YA (en) 2018-08-03 2021-03-30 Univ Kyoto Cell production method
TW202035697A (en) 2018-08-10 2020-10-01 國立大學法人京都大學 Transfection method to cardiomyocytes by using cationic lipids
CA3108275A1 (en) 2018-08-10 2020-02-13 Vertex Pharmaceuticals Incorporated Stem cell derived islet differentiation
CN112567025A (en) 2018-08-10 2021-03-26 国立大学法人京都大学 Method for producing CD3 positive cell
EA202190582A1 (en) 2018-08-22 2021-05-27 Киото Юниверсити METHOD FOR OBTAINING ENTERAL NEURAL PRECEDER CELLS
EP3845654A4 (en) 2018-08-31 2022-05-11 Noile-Immune Biotech, Inc. Car-expressing t cells and car expression vector
JP7295576B2 (en) 2018-09-19 2023-06-21 オリヅルセラピューティクス株式会社 insulin-producing cells
JPWO2020080270A1 (en) 2018-10-15 2021-09-16 公立大学法人横浜市立大学 Nutritional composition
US20220017867A1 (en) 2018-12-06 2022-01-20 Kirin Holdings Kabushiki Kaisha Production method for t cells or nk cells, medium for culturing t cells or nk cells, method for culturing t cells or nk cells, method for maintaining undifferentiated state of undifferentiated t cells, and growth-accelerating agent for t cells or nk cells
CN113728103A (en) 2018-12-11 2021-11-30 罗切斯特大学 Methods of treating schizophrenia and other neuropsychiatric disorders
IL301193A (en) 2018-12-20 2023-05-01 Regeneron Pharma Nuclease-mediated repeat expansion
WO2020130147A1 (en) 2018-12-21 2020-06-25 国立大学法人京都大学 Lubricin-localized cartilage-like tissue, method for producing same and composition comprising same for treating articular cartilage damage
JPWO2020138371A1 (en) 2018-12-26 2021-11-11 キリンホールディングス株式会社 Modified TCR and its manufacturing method
JPWO2020138256A1 (en) 2018-12-27 2021-11-04 国立大学法人京都大学 T cell receptor variants
JPWO2020158914A1 (en) 2019-02-01 2021-12-02 国立大学法人京都大学 Cell detection method
US20230057355A1 (en) 2019-02-13 2023-02-23 University Of Rochester Gene networks that mediate remyelination of the human brain
CN113811607A (en) 2019-03-07 2021-12-17 加利福尼亚大学董事会 CRISPR-Cas effector polypeptides and methods of use thereof
WO2020209959A1 (en) 2019-03-08 2020-10-15 Crispr Therapeutics Ag Nucleobase-editing fusion protein systems, compositions, and uses thereof
CA3132630A1 (en) 2019-03-12 2020-09-17 Crispr Therapeutics Ag Novel high fidelity rna-programmable endonuclease systems and uses thereof
EP3950953A4 (en) 2019-03-29 2023-04-19 Public University Corporation Yokohama City University Screening method and toxicity evaluation method
WO2020209389A1 (en) 2019-04-10 2020-10-15 国立大学法人京都大学 Method for producing biotissue-like structure
WO2020225606A1 (en) 2019-05-08 2020-11-12 Crispr Therapeutics Ag Crispr/cas all-in-two vector systems for treatment of dmd
CA3141455A1 (en) 2019-05-20 2020-11-26 Ajinomoto Co., Inc. Expansion culture method for cartilage or bone precursor cells
EP3985104A4 (en) 2019-06-11 2023-04-12 Kyoto University Method for producing renal interstitial cell
JP2022539169A (en) 2019-06-25 2022-09-07 バーテックス ファーマシューティカルズ インコーポレイテッド Enhanced beta cell differentiation
CN110257334A (en) * 2019-07-24 2019-09-20 安徽科门生物科技有限公司 A kind of nerve stem cell culture medium and cultural method
US10501404B1 (en) 2019-07-30 2019-12-10 Factor Bioscience Inc. Cationic lipids and transfection methods
WO2021030424A1 (en) 2019-08-13 2021-02-18 Semma Therapeutics, Inc. Pancreatic differentiation
WO2021033699A1 (en) 2019-08-20 2021-02-25 国立大学法人京都大学 Method for enriching cardiac myocytes
EP3886880B1 (en) 2019-10-18 2023-06-07 Amniotics AB Processes and apparatuses for obtaining amniotic mesenchymal stem cells from amniotic fluid and cells derived thereof
CN114929854A (en) 2019-10-21 2022-08-19 千纸鹤治疗公司 Proliferation inhibitor
US11760980B2 (en) 2019-10-28 2023-09-19 China Medical University Induced pluripotent stem cells (IPSCS) and applications thereof
TWI769410B (en) * 2019-10-28 2022-07-01 中國醫藥大學 New induced pluripotent stem cells (ipscs) and applications thereof
WO2021085576A1 (en) 2019-11-01 2021-05-06 国立大学法人京都大学 Method for producing t cell
WO2021106832A1 (en) 2019-11-25 2021-06-03 国立大学法人京都大学 T-cell master cell bank
EP4074321A4 (en) 2019-12-12 2024-01-03 Univ Chiba Nat Univ Corp Freeze-dried preparation containing megakaryocytes and platelets
WO2021142318A1 (en) 2020-01-09 2021-07-15 System1 Biosciences, Inc. Methods and compositions of pro-organ formation on particle substrates and uses thereof
US20230094922A1 (en) * 2020-02-16 2023-03-30 Genetikaplus Ltd. Methods of therapeutic prognostication
IL295964A (en) 2020-02-28 2022-10-01 Takeda Pharmaceuticals Co Method for producing natural killer cells from pluripotent stem cells
KR20220149592A (en) 2020-03-19 2022-11-08 오리즈루 세라퓨틱스 가부시키가이샤 Cardiomyocyte purification method
CN115427552A (en) 2020-03-19 2022-12-02 千纸鹤治疗公司 Method for purifying cardiomyocytes
CN115516084A (en) 2020-03-31 2022-12-23 国立大学法人京都大学 Method for producing T cell progenitor cell
CA3185067A1 (en) 2020-05-26 2021-12-02 Kouichi Tamura Hypoimmunogenic cells
JPWO2021241668A1 (en) 2020-05-28 2021-12-02
EP3922431A1 (en) 2020-06-08 2021-12-15 Erasmus University Medical Center Rotterdam Method of manufacturing microdevices for lab-on-chip applications
US20230235319A1 (en) 2020-06-12 2023-07-27 Bayer Aktiengesellschaft Crispr-cas12a directed random mutagenesis agents and methods
US20230257705A1 (en) 2020-06-17 2023-08-17 Kyoto University Chimeric antigen receptor-expressing immunocompetent cells
JP7429294B2 (en) 2020-07-13 2024-02-07 国立大学法人京都大学 Skeletal muscle progenitor cells and methods for purifying the same, compositions for treating myogenic diseases, and methods for producing cell groups containing skeletal muscle progenitor cells
CN116134130A (en) 2020-07-20 2023-05-16 学校法人爱知医科大学 Composition for maintaining and culturing pluripotent cells in undifferentiated state, culture medium for maintaining and culturing pluripotent cells in undifferentiated state, and method for producing pluripotent cells
CN116323917A (en) 2020-08-18 2023-06-23 国立大学法人京都大学 Method for maintaining and expanding human primordial germ cells/human primordial germ cell-like cells
JPWO2022107877A1 (en) 2020-11-20 2022-05-27
KR20230145101A (en) 2021-02-09 2023-10-17 오리즈루 세라퓨틱스 가부시키가이샤 maturation agent
USD969942S1 (en) * 2021-03-03 2022-11-15 Amber Puchlov Exercise device
WO2022191171A1 (en) 2021-03-09 2022-09-15 国立大学法人 東京医科歯科大学 Cell cluster production method
EP4310176A1 (en) 2021-03-17 2024-01-24 Astellas Pharma Inc. Pericyte having basic fibroblast growth factor (bfgf) gene introduced therein
CN113106059B (en) * 2021-04-07 2023-08-15 清华大学深圳国际研究生院 High-migration mesenchymal stem cells, and preparation method and application thereof
TW202305121A (en) 2021-04-08 2023-02-01 日商武田藥品工業股份有限公司 Method for activating t cell
JPWO2022230919A1 (en) 2021-04-28 2022-11-03
CA3218400A1 (en) 2021-04-30 2022-11-03 Riken Cord-like aggregates of retinal pigment epithelial cells, device and production method for producing same, and therapeutic agent comprising said cord-like aggregates
WO2022236187A1 (en) 2021-05-07 2022-11-10 Children's Hospital Los Angeles Methods for making stem cell-derived enteric neural crest cells and their use in enteric neuropathy treatment
WO2022250631A2 (en) * 2021-05-25 2022-12-01 T.C. Ankara Universitesi Rektorlugu Disease model of induced pluripotent stem cell (ipsc)-derived type 2 diabetes mellitus, associated with coronary artery disease
WO2022255489A1 (en) 2021-06-04 2022-12-08 キリンホールディングス株式会社 Cell composition, method for producing cell composition, and pharmaceutical composition containing cell composition
EP4353243A1 (en) 2021-06-10 2024-04-17 Ajinomoto Co., Inc. Method for producing mesenchymal stem cells
EP4352214A1 (en) 2021-06-11 2024-04-17 Bayer AG Type v rna programmable endonuclease systems
EP4101928A1 (en) 2021-06-11 2022-12-14 Bayer AG Type v rna programmable endonuclease systems
WO2022264033A1 (en) 2021-06-15 2022-12-22 Takeda Pharmaceutical Company Limited Method for producing natural killer cells from pluripotent stem cells
JPWO2023286834A1 (en) 2021-07-15 2023-01-19
JPWO2023286832A1 (en) 2021-07-15 2023-01-19
WO2023003025A1 (en) 2021-07-21 2023-01-26 国立大学法人京都大学 Method for producing retinal tissue
WO2023017848A1 (en) 2021-08-11 2023-02-16 国立大学法人京都大学 Method for producing renal interstitial progenitor cells, erythropoietin-producing cells, and method for producing renin-producing cells
EP4144841A1 (en) 2021-09-07 2023-03-08 Bayer AG Novel small rna programmable endonuclease systems with impoved pam specificity and uses thereof
US20230159890A1 (en) 2021-10-20 2023-05-25 University Of Rochester Treatment with genetically modified cells, and genetically modified cells per se, with increased competitive advantage and/or decreased competitive disadvantage
WO2023069979A1 (en) 2021-10-20 2023-04-27 University Of Rochester Isolated glial progenitor cells for use in the competition treatment of age-related white matter loss
US20230292719A1 (en) 2021-10-20 2023-09-21 University Of Rochester Humanized chimeras for the prospective assessment of cell addition and replacement therapies
CA3234231A1 (en) 2021-10-21 2023-04-27 Vertex Pharmaceuticals Incorporated Hypoimmune cells
CA3232971A1 (en) 2021-11-01 2023-05-04 George Harb Stem cell derived pancreatic islet differentiation
WO2023081633A1 (en) 2021-11-02 2023-05-11 University Of Rochester Tcf7l2 mediated remyelination in the brain
WO2023118068A1 (en) 2021-12-23 2023-06-29 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023150557A1 (en) 2022-02-01 2023-08-10 University Of Rochester Methods of generating a population of neurons from human glial progenitor cells and genetic constructs for carrying out such methods
WO2023167986A1 (en) 2022-03-02 2023-09-07 Lineage Cell Therapeutics, Inc. Methods and compositions for treating hearing loss
WO2023211857A1 (en) 2022-04-25 2023-11-02 Lineage Cell Therapeutics, Inc. Methods and compositions for treating vision loss
WO2023215455A1 (en) 2022-05-05 2023-11-09 University Of Rochester Dual macroglial-microglial approach towards therapeutic cell replacement in neurodegenerative and neuropsychiatric disease
WO2023237587A1 (en) 2022-06-10 2023-12-14 Bayer Aktiengesellschaft Novel small type v rna programmable endonuclease systems
WO2023247532A1 (en) 2022-06-21 2023-12-28 Institut National de la Santé et de la Recherche Médicale A method for producing a bioengineered mammal induced pluripotent stem cell-derived cardiac organoid

Citations (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4650764A (en) * 1983-04-12 1987-03-17 Wisconsin Alumni Research Foundation Helper cell
US4861719A (en) * 1986-04-25 1989-08-29 Fred Hutchinson Cancer Research Center DNA constructs for retrovirus packaging cell lines
US4937190A (en) * 1987-10-15 1990-06-26 Wisconsin Alumni Research Foundation Translation enhancer
US5225348A (en) * 1989-03-14 1993-07-06 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
US5266491A (en) * 1989-03-14 1993-11-30 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5324645A (en) * 1989-12-14 1994-06-28 Ajinomoto Co., Inc. Highly retrovirus-producing DNA construct and cell line
US5449614A (en) * 1988-09-01 1995-09-12 Whitehead Institue For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5525735A (en) * 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) * 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
US5591624A (en) * 1988-03-21 1997-01-07 Chiron Viagene, Inc. Retroviral packaging cell lines
US5637456A (en) * 1995-02-17 1997-06-10 The University Of Texas, Board Of Regents Rapid test for determining the amount of functionally inactive gene in a gene therapy vector preparation
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US5707618A (en) * 1995-03-24 1998-01-13 Genzyme Corporation Adenovirus vectors for gene therapy
US5744320A (en) * 1995-06-07 1998-04-28 Promega Corporation Quenching reagents and assays for enzyme-mediated luminescence
US5817491A (en) * 1990-09-21 1998-10-06 The Regents Of The University Of California VSV G pseusdotyped retroviral vectors
US5858740A (en) * 1993-06-11 1999-01-12 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US5910434A (en) * 1995-12-15 1999-06-08 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
US6013517A (en) * 1994-05-09 2000-01-11 Chiron Corporation Crossless retroviral vectors
US6017735A (en) * 1997-01-23 2000-01-25 Marie Curie Cancer Care Materials and methods for intracellular transport and their uses
US6025192A (en) * 1996-09-20 2000-02-15 Cold Spring Harbor Laboratory Modified retroviral vectors
US6140111A (en) * 1987-12-11 2000-10-31 Whitehead Institute For Biomedical Research Retroviral gene therapy vectors and therapeutic methods based thereon
US6203975B1 (en) * 1994-10-28 2001-03-20 The Trustees Of The University Of Pennsylvania Adenovirus and method of use thereof
US6255071B1 (en) * 1996-09-20 2001-07-03 Cold Spring Harbor Laboratory Mammalian viral vectors and their uses
US6365352B1 (en) * 1997-08-22 2002-04-02 Yale University Process to study changes in gene expression in granulocytic cells
US6395546B1 (en) * 2000-02-01 2002-05-28 Neurogeneration, Inc. Generation of dopaminergic neurons from human nervous system stem cells
US20020090722A1 (en) * 2000-06-15 2002-07-11 Tanja Dominko Pluripotent mammalian cells
US6451595B1 (en) * 1998-06-26 2002-09-17 Viromed Limited High efficiency retroviral vectors that contain none of viral coding sequences
US20030003574A1 (en) * 2000-01-24 2003-01-02 Jean Toma Multipotent stem cells from peripheral tissues and uses thereof
US6521455B2 (en) * 1995-07-28 2003-02-18 Marie Curie Cancer Care Nucleic acid molecule encoding a transport protein
US6521453B1 (en) * 1999-01-19 2003-02-18 Maxygen, Inc. Oligonucloetide mediated nucleic acid recombination
US20030044976A1 (en) * 2001-08-27 2003-03-06 Advanced Cell Technology De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US6605275B1 (en) * 1987-11-12 2003-08-12 Pharmastem Therapeutics, Inc. Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US20030161817A1 (en) * 2001-03-28 2003-08-28 Young Henry E. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US20040048297A1 (en) * 2002-07-30 2004-03-11 Gene Logic, Inc. Nucleic acid detection assay control genes
US20040137460A1 (en) * 2001-05-31 2004-07-15 Shinya Yamanaka Genes with es cell-specific expression
US6773920B1 (en) * 1999-03-31 2004-08-10 Invitrogen Corporation Delivery of functional protein sequences by translocating polypeptides
US6841535B2 (en) * 2000-07-31 2005-01-11 Active Motif Peptide-mediated transfection agents and methods of use
US20050019801A1 (en) * 2003-06-04 2005-01-27 Curis, Inc. Stem cell-based methods for identifying and characterizing agents
US20050026133A1 (en) * 2002-01-31 2005-02-03 Asahi Techno Glass Corporation Cryopreservation medium for primate embryo stem cells and cryopreservation method
US6872528B2 (en) * 1996-07-16 2005-03-29 Universite Pierre Et Marie Curie Highly productive packaging lines
US6875578B2 (en) * 1997-02-27 2005-04-05 Cellomics, Inc. System for cell-based screening
US20050079606A1 (en) * 2001-09-20 2005-04-14 Kyowa Hakko Kogyo Co., Ltd. Pluripotent stem cells originating in skeletal muscle intestinal tissue
US6881825B1 (en) * 1999-09-01 2005-04-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Identication of peptides that facilitate uptake and cytoplasmic and/or nuclear transport of proteins, DNA and virues
US20050130144A1 (en) * 2001-09-21 2005-06-16 Norio Nakatsuji Method of screening reprogramming factor, reprogramming factor screened by the method, method of using the reprogramming factor, method of differentiating undifferentiated fused cells and method of constructing cell, tissues and organs
US6910434B2 (en) * 2000-08-31 2005-06-28 Edwin Lundgren Control device for steering kite on a boat
US6995009B1 (en) * 1999-06-01 2006-02-07 Chugai Seiyaku Kabushiki Kaisha Packaging cell
US20060030041A1 (en) * 1999-08-05 2006-02-09 Regents Of The University Of Minnesota Multipotent adult stem cells and methods for isolation
US7029913B2 (en) * 1995-01-20 2006-04-18 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US7030292B2 (en) * 2001-01-02 2006-04-18 Stemron, Inc. Method for producing a population of homozygous stem cells having a pre-selected immunotype and/or genotype, cells suitable for transplant derived therefrom, and materials and methods using same
US20060084172A1 (en) * 2001-12-10 2006-04-20 Julius-Maximilians-Unversitat Wurzburg Method for producing stem cells with increased developmental potential
US20060088599A1 (en) * 2004-08-02 2006-04-27 Prasad Paras N Amino functionalized ORMOSIL nanoparticles as delivery vehicles
US20060095319A1 (en) * 2004-10-29 2006-05-04 Cardwell Carlzo B Marketing and compensation method
US7070994B2 (en) * 1988-03-21 2006-07-04 Oxford Biomedica (Uk) Ltd. Packaging cells
US20070033061A1 (en) * 2005-04-05 2007-02-08 Achaogen, Inc. Business methods for commercializing antimicrobial and cytotoxic compounds
US20070053884A1 (en) * 2003-05-16 2007-03-08 Kyowa Hakko Kogyo Co., Ltd Novel adult tissue-derived stem cell and use thereof
US20070155013A1 (en) * 2004-03-23 2007-07-05 Toshihiro Akaike Pluripotent stem cell growing method
US20070202592A1 (en) * 2004-07-08 2007-08-30 Yasuo Kitagawa Pluripotent Cells Distributed Ubiquitously In Animal Tissue, Which Proliferate Selectively In Lower-Serum Culture
US20080003560A1 (en) * 2001-09-21 2008-01-03 Reprocell Inc. Tailor-made pluripotent stem cell and use of the same
US20080085555A1 (en) * 2005-02-28 2008-04-10 Takayuki Asahara Method For In Vitro Amplification Of Adult Stem Cells
US20080132803A1 (en) * 2006-11-30 2008-06-05 Hyman Friedlander Method and system for doing business by mining the placental-chord complex
US20080171358A1 (en) * 2004-06-01 2008-07-17 Jacques Perrault Expression System
US20080171385A1 (en) * 2007-01-17 2008-07-17 Veit Bergendahl Culture of stem cells
US20080206865A1 (en) * 2001-10-03 2008-08-28 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20080233610A1 (en) * 2007-03-23 2008-09-25 Thomson James A Somatic cell reprogramming
US7439064B2 (en) * 2000-03-09 2008-10-21 Wicell Research Institute, Inc. Cultivation of human embryonic stem cells in the absence of feeder cells or without conditioned medium
US20090047263A1 (en) * 2005-12-13 2009-02-19 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090068742A1 (en) * 2005-12-13 2009-03-12 Shinya Yamanaka Nuclear Reprogramming Factor
US20090191171A1 (en) * 2008-01-18 2009-07-30 Yupo Ma Reprogramming of Differentiated Progenitor or Somatic Cells Using Homologous Recombination
US20090191159A1 (en) * 2007-06-15 2009-07-30 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20090227032A1 (en) * 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090246875A1 (en) * 2007-12-10 2009-10-01 Kyoto University Efficient method for nuclear reprogramming
US20100003757A1 (en) * 2008-06-04 2010-01-07 Amanda Mack Methods for the production of ips cells using non-viral approach
US20100021437A1 (en) * 2008-04-07 2010-01-28 The McLean Hospital Corporation Whitehead Institute for Biomedical Research Neural stem cells derived from induced pluripotent stem cells
US20100062534A1 (en) * 2008-09-09 2010-03-11 The General Hospital Corporation Inducible lentiviral vectors for reprogramming somatic cells
US20100075421A1 (en) * 2007-12-10 2010-03-25 Kyoto University Efficient method for nuclear reprogramming
US20100093090A1 (en) * 2008-04-03 2010-04-15 Peking University Method and kit for efficient reprogramming of somatic cells
US20100144031A1 (en) * 2003-11-26 2010-06-10 Rudolf Jaenisch Methods for reprogramming somatic cells
US20100184227A1 (en) * 2008-10-24 2010-07-22 James Thomson Pluripotent stem cells obtained by non-viral reprogramming
US20100184051A1 (en) * 2007-05-30 2010-07-22 The General Hospital Corporation Methods of generating pluripotent cells from somatic cells
US20100216236A1 (en) * 2005-12-13 2010-08-26 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20100233804A1 (en) * 2008-03-17 2010-09-16 The Scripps Research Institute Generation of pluripotent stem cells using recombinant proteins

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR901228A (en) 1943-01-16 1945-07-20 Deutsche Edelstahlwerke Ag Ring gap magnet system
US4864719A (en) * 1987-04-24 1989-09-12 Amp Incorporated Tool for removing electrical contacts
JP2886547B2 (en) 1988-07-26 1999-04-26 協和醗酵工業株式会社 Method for producing neuraminidase
JP2928287B2 (en) 1988-09-29 1999-08-03 協和醗酵工業株式会社 Novel polypeptide
FR2707091B1 (en) 1993-06-30 1997-04-04 Cohen Haguenauer Odile Retroviral vector for gene transfer and expression in eukaryotic cells.
US5534423A (en) 1993-10-08 1996-07-09 Regents Of The University Of Michigan Methods of increasing rates of infection by directing motion of vectors
JP4216350B2 (en) 1994-09-19 2009-01-28 大日本住友製薬株式会社 Recombinant DNA viral vector for animal cell infection
US5830725A (en) * 1995-04-28 1998-11-03 The Board Of Trustees For The Leland Stanford Junior University Rapid, stable high-titre production of recombing retrovirus
AU712801B2 (en) 1995-09-20 1999-11-18 Merck Sharp & Dohme Corp. Histone deacetylase as target for antiprotozoal agents
JP4053595B2 (en) 1995-09-22 2008-02-27 メディカル・リサーチ・カウンシル Improvements in or related to mutagenesis of nucleic acids
TR199902512T2 (en) 1997-04-16 2000-06-21 Amgen Inc. Osteoprotegerin binding proteins and receptors.
US6387673B1 (en) 1997-05-01 2002-05-14 The Salk Institute For Biological Studies Compounds useful for the modulation of processes mediated by nuclear hormone receptors, methods for the identification and use of such compounds
WO2000018885A1 (en) 1998-09-29 2000-04-06 Gamida Cell Ltd. Methods of controlling proliferation and differentiation of stem and progenitor cells
US6835567B1 (en) 1998-04-14 2004-12-28 Signal Pharmaceuticals, Inc. PNS cell lines and methods of use therefor
US20020174013A1 (en) 1998-04-17 2002-11-21 Viztec Inc., A Florida Corporation Chip card advertising method and system
GB9809178D0 (en) 1998-04-29 1998-07-01 Univ Edinburgh Nuclear reprogramming of somatic cells
EP1080218A1 (en) 1998-05-27 2001-03-07 University of Florida Method of preparing recombinant adeno-associated virus compositions by using an iodixanol gradient
FR2779445B1 (en) 1998-06-08 2000-08-11 Univ Nantes ENCAPSIDATION KIT
AUPP505798A0 (en) 1998-08-04 1998-08-27 Fujisawa Pharmaceutical Co., Ltd. Novel compound fr225497 substance
US6485959B1 (en) 1998-10-07 2002-11-26 Cedars Sinai Medical Center Cell preconditioning and cryopresevation medium
AU6339399A (en) * 1998-10-16 2000-05-08 Novartis Ag Promotion of self-renewal and improved gene transduction of hematopoietic stem cells by histone deacetylase inhibitors
US6667176B1 (en) 2000-01-11 2003-12-23 Geron Corporation cDNA libraries reflecting gene expression during growth and differentiation of human pluripotent stem cells
IL142748A0 (en) 1998-11-09 2002-03-10 Univ Monash Embryonic stem cells
US6153432A (en) 1999-01-29 2000-11-28 Zen-Bio, Inc Methods for the differentiation of human preadipocytes into adipocytes
US6312949B1 (en) 1999-03-26 2001-11-06 The Salk Institute For Biological Studies Regulation of tyrosine hydroxylase expression
EP1218489B1 (en) * 1999-09-24 2009-03-18 Cybios LLC Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US6280718B1 (en) 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
US6458589B1 (en) 2000-04-27 2002-10-01 Geron Corporation Hepatocyte lineage cells derived from pluripotent stem cells
AU6319901A (en) 2000-05-17 2001-11-26 Geron Corp Neural progenitor cell populations
DE10031179A1 (en) 2000-06-27 2002-01-31 Amaxa Gmbh Introducing active molecules into the nucleus of eukaryotic cells, useful for transfection and gene therapy, by applying controlled electrical pulse
JP2002065261A (en) 2000-08-30 2002-03-05 Mitsubishi Kasei Institute Of Life Sciences Method for obtaining reproductive cell
US20020127715A1 (en) 2000-11-27 2002-09-12 Nissim Benvenisty Transfection of human embryonic stem cells
US20080268054A1 (en) * 2000-12-04 2008-10-30 Eugene Bell Dermal derived human stem cells and compositions and methods thereof
US20020142397A1 (en) 2000-12-22 2002-10-03 Philippe Collas Methods for altering cell fate
JP2003009854A (en) 2001-04-09 2003-01-14 Kyowa Hakko Kogyo Co Ltd Method for embryoid body formation and use thereof
DE10119901A1 (en) 2001-04-23 2002-10-24 Amaxa Gmbh Apparatus for electrophoretic transfer of biologically active molecules into cells comprises capacitors connected to high voltage sources, which discharge via power transistor into cuvette holding sample
EP1390518B1 (en) 2001-04-23 2004-10-06 Amaxa GmbH Buffer solution for electroporation and a method comprising the use of the same
WO2003081536A1 (en) 2002-03-26 2003-10-02 So-Woon Kim System and method for 3-dimension simulation of glasses
US7422736B2 (en) * 2002-07-26 2008-09-09 Food Industry Research And Development Institute Somatic pluripotent cells
JP3736517B2 (en) 2002-11-13 2006-01-18 学校法人近畿大学 Somatic cell nuclear reprogramming factor
AU2003901099A0 (en) 2003-03-11 2003-03-27 Es Cell International Pte Ltd. Methods of inducing differentiation of stem cells
WO2004099372A2 (en) 2003-05-01 2004-11-18 University Of Florida Anti-scarring ribozymes and methods
US9567591B2 (en) 2003-05-15 2017-02-14 Mello Biotechnology, Inc. Generation of human embryonic stem-like cells using intronic RNA
CA2526120A1 (en) 2003-06-03 2005-02-24 Cell Genesys, Inc. Compositions and methods for enhanced expression of recombinant polypeptides from a single vector using a peptide cleavage site
WO2005033297A1 (en) 2003-09-19 2005-04-14 The Rockefeller University Compositions, methods and kits relating to reprogramming adult differentiated cells and production of embryonic stem cell-like cells
JP2005095027A (en) 2003-09-22 2005-04-14 Reprocell Inc Undifferentiated state marker promoter of cell and its utilization
CA2542130A1 (en) 2003-10-09 2005-04-21 Kyowa Hakko Kogyo Co., Ltd. Cell genomically modified to produce polypeptides with an altered glycosylation pattern
EP1682150B1 (en) 2003-11-10 2012-12-26 The Scripps Research Institute Compositions and methods for inducing cell dedifferentiation
JP2005155461A (en) * 2003-11-26 2005-06-16 Sanyo Electric Co Ltd Compressor
CA2549158A1 (en) 2003-12-01 2005-06-16 Technion Research And Development Foundation Ltd. Methods of generating stem cells and embryonic bodies carrying disease-causing mutations and methods of using same for studying genetic disorders
JP4340736B2 (en) 2004-01-15 2009-10-07 国立大学法人福井大学 Vector incorporating a reporter gene
WO2005080598A1 (en) 2004-02-19 2005-09-01 Dainippon Sumitomo Pharma Co., Ltd. Method of screening somatic cell nucleus initializer
WO2006084229A2 (en) 2004-07-15 2006-08-10 Primegen Biotech, Llc Use of nuclear material to therapeutically reprogram differentiated cells
FR2873175B1 (en) * 2004-07-16 2006-10-20 Poclain Hydraulics Ind Soc Par HYDRAULIC CIRCUIT COMPRISING A MULTIFUNCTION SELECTOR
JPWO2006035741A1 (en) 2004-09-29 2008-05-15 伸弥 山中 ES cell specific expression gene and use thereof
JP5053850B2 (en) 2004-10-08 2012-10-24 バークシス コーポレーション Use of RNA trans-splicing for antibody gene transfer and antibody polypeptide production
WO2006088867A2 (en) * 2005-02-15 2006-08-24 Medistem Laboratories, Incorporated Method for expansion of stem cells
FR2885536B1 (en) * 2005-05-12 2007-07-27 Roquette Freres COMPOSITION BASED ON DIANHYDROHEXITOL ETHERS FOR THE TREATMENT OF MATTER OTHER THAN THE HUMAN BODY
WO2007026255A2 (en) 2005-06-22 2007-03-08 Universitetet I Oslo Dedifferentiated cells and methods of making and using dedifferentiated cells
EP1910550A4 (en) 2005-07-21 2009-11-04 Abbott Lab Multiple gene expression including sorf constructs and methods with polyproteins, pro-proteins, and proteolysis
EP2302034B1 (en) 2005-08-03 2020-10-14 Astellas Institute for Regenerative Medicine Improved methods of reprogramming animal somatic cells
JP2007117081A (en) * 2005-09-30 2007-05-17 Institute Of Physical & Chemical Research Method for producing nuclear-transplanted egg
EP1957643A2 (en) 2005-11-11 2008-08-20 The University Court Of The University of Edinburgh Reprogramming and genetic modification of cells
WO2007097494A1 (en) 2006-02-27 2007-08-30 Imgen Co., Ltd. De-differentiation of astrocytes into neural stem cell using bmi-1
US20090252711A1 (en) * 2006-05-11 2009-10-08 Andrew Craig Boquest Stem Cells And Methods Of Making And Using Stem Cells
WO2008030610A2 (en) 2006-09-08 2008-03-13 Michigan State University Human transcriptome corresponding to human oocytes and use of said genes or the corresponding polypeptides to trans-differentiate somatic cells
JP2008099662A (en) * 2006-09-22 2008-05-01 Institute Of Physical & Chemical Research Method for culturing stem cell
US8158415B2 (en) 2007-02-27 2012-04-17 Procell Therapeutics Inc. Combined use of cell permeable Nanog and Oct4 for increasing self-renewal and suppressing differentiation of stem cells
WO2008105566A1 (en) 2007-02-27 2008-09-04 Korea Stem Cell Bank System for providing stem cell services using internet and method thereof
WO2008124133A1 (en) 2007-04-07 2008-10-16 Whitehead Institute For Biomedical Research Reprogramming of somatic cells
AU2008260187A1 (en) 2007-05-29 2008-12-11 Christopher B. Reid Methods for production and uses of multipotent cell populations
US9213999B2 (en) * 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
US20120282229A1 (en) * 2007-08-01 2012-11-08 Christian Kannemeier Non-viral delivery of transcription factors that reprogram human somatic cells into a stem cell-like state
US20090191160A1 (en) 2007-08-10 2009-07-30 University Of Dayton Methods of producing pluripotent stem-like cells
MX2010002242A (en) * 2007-08-31 2010-06-01 Whitehead Biomedical Inst Wnt pathway stimulation in reprogramming somatic cells.
WO2009057831A1 (en) * 2007-10-31 2009-05-07 Kyoto University Nuclear reprogramming method
WO2009061442A1 (en) * 2007-11-06 2009-05-14 Children's Medical Center Corporation Method to produce induced pluripotent stem (ips) cells form non-embryonic human cells
US9005966B2 (en) 2007-11-19 2015-04-14 The Regents Of The University Of California Generation of pluripotent cells from fibroblasts
KR101481164B1 (en) 2008-01-30 2015-01-09 주식회사 미래셀바이오 Method of manufacturing induced pluripotent stem cell originated from somatic cell
US20110014164A1 (en) 2008-02-15 2011-01-20 President And Fellows Of Harvard College Efficient induction of pluripotent stem cells using small molecule compounds
JP2011516042A (en) 2008-03-17 2011-05-26 へルムホルツ・ツェントルム・ミュンヘン−ドイチェス・フォルシュングスツェントルム・ヒューア・ゲズントハイト・ウント・ウムヴェルト(ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング) Vector and method for producing vector-free induced pluripotent stem (iPS) cells using site-specific recombination
CN101250502A (en) 2008-04-01 2008-08-27 中国科学院上海生命科学研究院 Method for preparing evoked pluripotent stem cell
WO2009133971A1 (en) 2008-05-02 2009-11-05 Kyoto University Method of nuclear reprogramming
EP2128245A1 (en) 2008-05-27 2009-12-02 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Generation of induced pluripotent stem (iPS) cells
US20110201110A1 (en) 2008-07-31 2011-08-18 Gifu University Efficient method for establishing induced pluripotent stem cells
US8298825B1 (en) 2008-08-25 2012-10-30 The General Hospital Corporation TGF-beta receptor inhibitors to enhance direct reprogramming
JP4992952B2 (en) 2009-11-02 2012-08-08 住友電気工業株式会社 Traffic signal control analyzer

Patent Citations (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4650764A (en) * 1983-04-12 1987-03-17 Wisconsin Alumni Research Foundation Helper cell
US4861719A (en) * 1986-04-25 1989-08-29 Fred Hutchinson Cancer Research Center DNA constructs for retrovirus packaging cell lines
US4937190A (en) * 1987-10-15 1990-06-26 Wisconsin Alumni Research Foundation Translation enhancer
US6605275B1 (en) * 1987-11-12 2003-08-12 Pharmastem Therapeutics, Inc. Isolation and preservation of fetal and neonatal hematopoietic stem and progenitor cells of the blood
US6140111A (en) * 1987-12-11 2000-10-31 Whitehead Institute For Biomedical Research Retroviral gene therapy vectors and therapeutic methods based thereon
US5591624A (en) * 1988-03-21 1997-01-07 Chiron Viagene, Inc. Retroviral packaging cell lines
US7070994B2 (en) * 1988-03-21 2006-07-04 Oxford Biomedica (Uk) Ltd. Packaging cells
US5716832A (en) * 1988-03-21 1998-02-10 Chiron Viagene, Inc. Packaging cells
US5955331A (en) * 1988-09-01 1999-09-21 Whitehead Institute For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5449614A (en) * 1988-09-01 1995-09-12 Whitehead Institue For Biomedical Research Recombinant retroviruses with amphotropic and ecotropic host ranges
US5225348A (en) * 1989-03-14 1993-07-06 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
US5266491A (en) * 1989-03-14 1993-11-30 Mochida Pharmaceutical Co., Ltd. DNA fragment and expression plasmid containing the DNA fragment
US5324645A (en) * 1989-12-14 1994-06-28 Ajinomoto Co., Inc. Highly retrovirus-producing DNA construct and cell line
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US5674980A (en) * 1989-12-21 1997-10-07 Biogen Inc Fusion protein comprising tat-derived transport moiety
US5817491A (en) * 1990-09-21 1998-10-06 The Regents Of The University Of California VSV G pseusdotyped retroviral vectors
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US5858740A (en) * 1993-06-11 1999-01-12 Cell Genesys, Inc. Method for production of high titer virus and high efficiency retroviral mediated transduction of mammalian cells
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US6013517A (en) * 1994-05-09 2000-01-11 Chiron Corporation Crossless retroviral vectors
US5525735A (en) * 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) * 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
US6203975B1 (en) * 1994-10-28 2001-03-20 The Trustees Of The University Of Pennsylvania Adenovirus and method of use thereof
US7029913B2 (en) * 1995-01-20 2006-04-18 Wisconsin Alumni Research Foundation Primate embryonic stem cells
US5637456A (en) * 1995-02-17 1997-06-10 The University Of Texas, Board Of Regents Rapid test for determining the amount of functionally inactive gene in a gene therapy vector preparation
US5707618A (en) * 1995-03-24 1998-01-13 Genzyme Corporation Adenovirus vectors for gene therapy
US5744320A (en) * 1995-06-07 1998-04-28 Promega Corporation Quenching reagents and assays for enzyme-mediated luminescence
US6521455B2 (en) * 1995-07-28 2003-02-18 Marie Curie Cancer Care Nucleic acid molecule encoding a transport protein
US5910434A (en) * 1995-12-15 1999-06-08 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
US6017761A (en) * 1995-12-15 2000-01-25 Systemix, Inc. Method for obtaining retroviral packaging cell lines producing high transducing efficiency retroviral supernatant
US6872528B2 (en) * 1996-07-16 2005-03-29 Universite Pierre Et Marie Curie Highly productive packaging lines
US6025192A (en) * 1996-09-20 2000-02-15 Cold Spring Harbor Laboratory Modified retroviral vectors
US6255071B1 (en) * 1996-09-20 2001-07-03 Cold Spring Harbor Laboratory Mammalian viral vectors and their uses
US6017735A (en) * 1997-01-23 2000-01-25 Marie Curie Cancer Care Materials and methods for intracellular transport and their uses
US6251398B1 (en) * 1997-01-23 2001-06-26 Marie Curie Cancer Care Materials and methods for intracellular transport and their uses
US6875578B2 (en) * 1997-02-27 2005-04-05 Cellomics, Inc. System for cell-based screening
US6365352B1 (en) * 1997-08-22 2002-04-02 Yale University Process to study changes in gene expression in granulocytic cells
US6451595B1 (en) * 1998-06-26 2002-09-17 Viromed Limited High efficiency retroviral vectors that contain none of viral coding sequences
US6521453B1 (en) * 1999-01-19 2003-02-18 Maxygen, Inc. Oligonucloetide mediated nucleic acid recombination
US6773920B1 (en) * 1999-03-31 2004-08-10 Invitrogen Corporation Delivery of functional protein sequences by translocating polypeptides
US6995009B1 (en) * 1999-06-01 2006-02-07 Chugai Seiyaku Kabushiki Kaisha Packaging cell
US20060030041A1 (en) * 1999-08-05 2006-02-09 Regents Of The University Of Minnesota Multipotent adult stem cells and methods for isolation
US6881825B1 (en) * 1999-09-01 2005-04-19 University Of Pittsburgh Of The Commonwealth System Of Higher Education Identication of peptides that facilitate uptake and cytoplasmic and/or nuclear transport of proteins, DNA and virues
US20030003574A1 (en) * 2000-01-24 2003-01-02 Jean Toma Multipotent stem cells from peripheral tissues and uses thereof
US6395546B1 (en) * 2000-02-01 2002-05-28 Neurogeneration, Inc. Generation of dopaminergic neurons from human nervous system stem cells
US7439064B2 (en) * 2000-03-09 2008-10-21 Wicell Research Institute, Inc. Cultivation of human embryonic stem cells in the absence of feeder cells or without conditioned medium
US20020090722A1 (en) * 2000-06-15 2002-07-11 Tanja Dominko Pluripotent mammalian cells
US6841535B2 (en) * 2000-07-31 2005-01-11 Active Motif Peptide-mediated transfection agents and methods of use
US6910434B2 (en) * 2000-08-31 2005-06-28 Edwin Lundgren Control device for steering kite on a boat
US7030292B2 (en) * 2001-01-02 2006-04-18 Stemron, Inc. Method for producing a population of homozygous stem cells having a pre-selected immunotype and/or genotype, cells suitable for transplant derived therefrom, and materials and methods using same
US20030161817A1 (en) * 2001-03-28 2003-08-28 Young Henry E. Pluripotent embryonic-like stem cells, compositions, methods and uses thereof
US20040137460A1 (en) * 2001-05-31 2004-07-15 Shinya Yamanaka Genes with es cell-specific expression
US7250255B2 (en) * 2001-05-31 2007-07-31 Shinya Yamanaka Genes with ES cell-specific expression
US20030044976A1 (en) * 2001-08-27 2003-03-06 Advanced Cell Technology De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20080076176A1 (en) * 2001-08-27 2008-03-27 Advanced Cell Technology, Inc. De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20060110830A1 (en) * 2001-08-27 2006-05-25 Advanced Cell Technology, Inc. De-differentiation and re-differentiation of somatic cells and production of cells for cell therapies
US20050079606A1 (en) * 2001-09-20 2005-04-14 Kyowa Hakko Kogyo Co., Ltd. Pluripotent stem cells originating in skeletal muscle intestinal tissue
US20050130144A1 (en) * 2001-09-21 2005-06-16 Norio Nakatsuji Method of screening reprogramming factor, reprogramming factor screened by the method, method of using the reprogramming factor, method of differentiating undifferentiated fused cells and method of constructing cell, tissues and organs
US20080003560A1 (en) * 2001-09-21 2008-01-03 Reprocell Inc. Tailor-made pluripotent stem cell and use of the same
US20080206865A1 (en) * 2001-10-03 2008-08-28 Su-Chun Zhang Method of in vitro differentiation of neural stem cells, motor neurons and dopamine neurons from primate embryonic stem cells
US20060084172A1 (en) * 2001-12-10 2006-04-20 Julius-Maximilians-Unversitat Wurzburg Method for producing stem cells with increased developmental potential
US20050026133A1 (en) * 2002-01-31 2005-02-03 Asahi Techno Glass Corporation Cryopreservation medium for primate embryo stem cells and cryopreservation method
US20040048297A1 (en) * 2002-07-30 2004-03-11 Gene Logic, Inc. Nucleic acid detection assay control genes
US20070053884A1 (en) * 2003-05-16 2007-03-08 Kyowa Hakko Kogyo Co., Ltd Novel adult tissue-derived stem cell and use thereof
US20050019801A1 (en) * 2003-06-04 2005-01-27 Curis, Inc. Stem cell-based methods for identifying and characterizing agents
US20100221827A1 (en) * 2003-11-26 2010-09-02 Rudolf Jaenisch Methods for reprogramming somatic cells
US20100144031A1 (en) * 2003-11-26 2010-06-10 Rudolf Jaenisch Methods for reprogramming somatic cells
US20070155013A1 (en) * 2004-03-23 2007-07-05 Toshihiro Akaike Pluripotent stem cell growing method
US20080171358A1 (en) * 2004-06-01 2008-07-17 Jacques Perrault Expression System
US20070202592A1 (en) * 2004-07-08 2007-08-30 Yasuo Kitagawa Pluripotent Cells Distributed Ubiquitously In Animal Tissue, Which Proliferate Selectively In Lower-Serum Culture
US20060088599A1 (en) * 2004-08-02 2006-04-27 Prasad Paras N Amino functionalized ORMOSIL nanoparticles as delivery vehicles
US20060095319A1 (en) * 2004-10-29 2006-05-04 Cardwell Carlzo B Marketing and compensation method
US20080085555A1 (en) * 2005-02-28 2008-04-10 Takayuki Asahara Method For In Vitro Amplification Of Adult Stem Cells
US20070033061A1 (en) * 2005-04-05 2007-02-08 Achaogen, Inc. Business methods for commercializing antimicrobial and cytotoxic compounds
US20100062533A1 (en) * 2005-12-13 2010-03-11 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090047263A1 (en) * 2005-12-13 2009-02-19 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090068742A1 (en) * 2005-12-13 2009-03-12 Shinya Yamanaka Nuclear Reprogramming Factor
US20100216236A1 (en) * 2005-12-13 2010-08-26 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090227032A1 (en) * 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20100210014A1 (en) * 2005-12-13 2010-08-19 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20080132803A1 (en) * 2006-11-30 2008-06-05 Hyman Friedlander Method and system for doing business by mining the placental-chord complex
US20080171385A1 (en) * 2007-01-17 2008-07-17 Veit Bergendahl Culture of stem cells
US20080233610A1 (en) * 2007-03-23 2008-09-25 Thomson James A Somatic cell reprogramming
US20100184051A1 (en) * 2007-05-30 2010-07-22 The General Hospital Corporation Methods of generating pluripotent cells from somatic cells
US20090191159A1 (en) * 2007-06-15 2009-07-30 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20100267135A1 (en) * 2007-06-15 2010-10-21 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20110039332A1 (en) * 2007-06-15 2011-02-17 Kazuhiro Sakurada Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US20100105100A1 (en) * 2007-06-15 2010-04-29 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20100120069A1 (en) * 2007-06-15 2010-05-13 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20090246875A1 (en) * 2007-12-10 2009-10-01 Kyoto University Efficient method for nuclear reprogramming
US20100075421A1 (en) * 2007-12-10 2010-03-25 Kyoto University Efficient method for nuclear reprogramming
US20090191171A1 (en) * 2008-01-18 2009-07-30 Yupo Ma Reprogramming of Differentiated Progenitor or Somatic Cells Using Homologous Recombination
US20100233804A1 (en) * 2008-03-17 2010-09-16 The Scripps Research Institute Generation of pluripotent stem cells using recombinant proteins
US20100093090A1 (en) * 2008-04-03 2010-04-15 Peking University Method and kit for efficient reprogramming of somatic cells
US20100021437A1 (en) * 2008-04-07 2010-01-28 The McLean Hospital Corporation Whitehead Institute for Biomedical Research Neural stem cells derived from induced pluripotent stem cells
US20100003757A1 (en) * 2008-06-04 2010-01-07 Amanda Mack Methods for the production of ips cells using non-viral approach
US20100062534A1 (en) * 2008-09-09 2010-03-11 The General Hospital Corporation Inducible lentiviral vectors for reprogramming somatic cells
US20100184227A1 (en) * 2008-10-24 2010-07-22 James Thomson Pluripotent stem cells obtained by non-viral reprogramming

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090047263A1 (en) * 2005-12-13 2009-02-19 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US20090068742A1 (en) * 2005-12-13 2009-03-12 Shinya Yamanaka Nuclear Reprogramming Factor
US8048999B2 (en) 2005-12-13 2011-11-01 Kyoto University Nuclear reprogramming factor
US20090227032A1 (en) * 2005-12-13 2009-09-10 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US8058065B2 (en) 2005-12-13 2011-11-15 Kyoto University Oct3/4, Klf4, c-Myc and Sox2 produce induced pluripotent stem cells
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
US20100210014A1 (en) * 2005-12-13 2010-08-19 Kyoto University Nuclear reprogramming factor and induced pluripotent stem cells
US9714433B2 (en) 2007-06-15 2017-07-25 Kyoto University Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US20110039332A1 (en) * 2007-06-15 2011-02-17 Kazuhiro Sakurada Human pluripotent stem cells induced from undifferentiated stem cells derived from a human postnatal tissue
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
US20100267135A1 (en) * 2007-06-15 2010-10-21 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20090324559A1 (en) * 2007-06-15 2009-12-31 Izumi Bio, Inc. Methods and platforms for drug discovery
US8257941B2 (en) 2007-06-15 2012-09-04 Kyoto University Methods and platforms for drug discovery using induced pluripotent stem cells
US8211697B2 (en) 2007-06-15 2012-07-03 Kyoto University Induced pluripotent stem cells produced using reprogramming factors and a rho kinase inhibitor or a histone deacetylase inhibitor
US20090304646A1 (en) * 2007-06-15 2009-12-10 Kazuhiro Sakurada Multipotent/Pluripotent Cells and Methods
US20090299763A1 (en) * 2007-06-15 2009-12-03 Izumi Bio, Inc. Methods of cell-based technologies
US20090191159A1 (en) * 2007-06-15 2009-07-30 Kazuhiro Sakurada Multipotent/pluripotent cells and methods
US20100279404A1 (en) * 2008-05-02 2010-11-04 Shinya Yamanaka Method of nuclear reprogramming
US9499797B2 (en) 2008-05-02 2016-11-22 Kyoto University Method of making induced pluripotent stem cells
US20110016540A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genome editing of genes associated with trinucleotide repeat expansion disorders in animals
US20110023158A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Bovine genome editing with zinc finger nucleases
US20110023148A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of addiction-related genes in animals
US20110023145A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in autism spectrum disorders
US20110023154A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Silkworm genome editing with zinc finger nucleases
US20110023149A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in tumor suppression in animals
US20110023143A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of neurodevelopmental genes in animals
US20110023146A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in secretase-associated disorders
US20110023153A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in alzheimer's disease
US20110023144A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in amyotrophyic lateral sclerosis disease
US20110023147A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of prion disorder-related genes in animals
US20110023152A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of cognition related genes in animals
US20110030072A1 (en) * 2008-12-04 2011-02-03 Sigma-Aldrich Co. Genome editing of immunodeficiency genes in animals
US20110023156A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Feline genome editing with zinc finger nucleases
US20110023140A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Rabbit genome editing with zinc finger nucleases
US20110023150A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of genes associated with schizophrenia in animals
US20110023139A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved in cardiovascular disease
US20110023151A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genome editing of abc transporters
US20110016546A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Porcine genome editing with zinc finger nucleases
US20110023141A1 (en) * 2008-12-04 2011-01-27 Sigma-Aldrich Co. Genomic editing of genes involved with parkinson's disease
US20110016543A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genomic editing of genes involved in inflammation
US20110016539A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genome editing of neurotransmission-related genes in animals
US20110016541A1 (en) * 2008-12-04 2011-01-20 Sigma-Aldrich Co. Genome editing of sensory-related genes in animals
WO2012092379A3 (en) * 2010-12-29 2014-02-27 Sigma-Aldrich Co. Llc Cells having disrupted expression of proteins involved in adme and toxicology processes
WO2012092379A2 (en) 2010-12-29 2012-07-05 Sigma-Aldrich Co. Llc Cells having disrupted expression of proteins involved in adme and toxicology processes
US10731134B2 (en) 2011-05-27 2020-08-04 Public University Corporation Yokohama City University Production method for artificial cancer stem cell and induced differentiation method therefor
US20150328247A1 (en) * 2012-12-24 2015-11-19 Ramot At Tel-Aviv University Ltd. Agents for treating genetic diseases resulting from nonsense mutations, and methods for identifying the same
US10987370B2 (en) * 2012-12-24 2021-04-27 Ramot At Tel-Aviv University Ltd. Methods of inducing read-through of a nonsense mutation associated with ataxia telangiectasia, Rett syndrome or spinal muscular atrophy by erythromycin or azithromycin
WO2014172616A3 (en) * 2013-04-18 2014-12-11 President And Fellows Of Harvard College Methods, compositions and kits for promoting motor neuron survival and treating and diagnosing neurodegenerative disorders
CN108291237A (en) * 2015-10-16 2018-07-17 菲特治疗公司 Platform for inducing and safeguarding ground state versatility
WO2017120443A1 (en) * 2016-01-07 2017-07-13 Cedars-Sinai Medical Center Method to identify key markers of human pluripotent cell-derived somatic cells that predict molecular similarity to in vivo target cells
US11359242B2 (en) 2016-01-07 2022-06-14 Cedars-Sinai Medical Center Method to identify key markers of human pluripotent cell-derived somatic cells that predict molecular similarity to in vivo target cells
WO2018140657A1 (en) * 2017-01-25 2018-08-02 Whitehead Institute For Biomedical Research Methods for building genomic networks and uses thereof
US20220076839A1 (en) * 2018-12-21 2022-03-10 I Peace, Inc. Health risk information management device, health risk information management method, and program
WO2021127594A1 (en) * 2019-12-18 2021-06-24 Editas Medicine, Inc. Engineered cells for therapy

Also Published As

Publication number Publication date
EP2213727A1 (en) 2010-08-04
CN102317448B (en) 2014-11-12
WO2009006997A1 (en) 2009-01-15
GB0810897D0 (en) 2008-07-23
MY159971A (en) 2017-02-15
US20100267135A1 (en) 2010-10-21
GB2465291B (en) 2012-01-18
EP2171045B1 (en) 2016-12-21
US20100105100A1 (en) 2010-04-29
WO2009007852A2 (en) 2009-01-15
EP2164952A1 (en) 2010-03-24
EP2171045A2 (en) 2010-04-07
EP2476749A1 (en) 2012-07-18
NZ582018A (en) 2012-05-25
GB2450603A (en) 2008-12-31
GB0922013D0 (en) 2010-02-03
US8211697B2 (en) 2012-07-03
IL202678A (en) 2015-09-24
HK1143831A1 (en) 2011-01-14
AU2008273817A1 (en) 2009-01-15
AU2008273817B2 (en) 2013-10-24
CA2690629A1 (en) 2009-01-15
WO2009006930A1 (en) 2009-01-15
US20090324559A1 (en) 2009-12-31
US20100120069A1 (en) 2010-05-13
WO2009007852A3 (en) 2009-08-20
US8257941B2 (en) 2012-09-04
US20140206083A1 (en) 2014-07-24
ZA200908904B (en) 2010-09-29
JP5547064B2 (en) 2014-07-09
AP2010005102A0 (en) 2010-02-28
CN102317448A (en) 2012-01-11
GB2450603B (en) 2010-02-10
GB2465291A (en) 2010-05-19
MX2009013706A (en) 2010-07-02
KR101657318B1 (en) 2016-09-13
IL202678A0 (en) 2011-08-01
US20090191159A1 (en) 2009-07-30
KR20100059781A (en) 2010-06-04
US20090304646A1 (en) 2009-12-10
EP2164952B1 (en) 2016-10-26
US20110039332A1 (en) 2011-02-17
JP2010529851A (en) 2010-09-02
JP2008307007A (en) 2008-12-25
US9714433B2 (en) 2017-07-25

Similar Documents

Publication Publication Date Title
US8257941B2 (en) Methods and platforms for drug discovery using induced pluripotent stem cells
WO2009152484A2 (en) Methods and platforms for drug discovery
US20120122212A1 (en) Tgf-beta pathway inhibitors for enhancement of cellular reprogramming of human cells
AU2014253960B2 (en) Age-modified cells and methods for making age-modified cells
US9523674B2 (en) Method of screening for substances capable of promoting induction of induced pluripotent stem cells
Shimamoto Generation and Characterization of Induced Pluripotent Stem
Shimamoto et al. Generation and Characterization of Induced Pluripotent Stem Cells from Aid

Legal Events

Date Code Title Description
AS Assignment

Owner name: KYOTO UNIVERSITY, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:IPIERIAN, INC.;REEL/FRAME:025914/0102

Effective date: 20110127

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION