WO2016142427A1 - Procédé et kit pour la reprogrammation de cellules somatiques - Google Patents

Procédé et kit pour la reprogrammation de cellules somatiques Download PDF

Info

Publication number
WO2016142427A1
WO2016142427A1 PCT/EP2016/055029 EP2016055029W WO2016142427A1 WO 2016142427 A1 WO2016142427 A1 WO 2016142427A1 EP 2016055029 W EP2016055029 W EP 2016055029W WO 2016142427 A1 WO2016142427 A1 WO 2016142427A1
Authority
WO
WIPO (PCT)
Prior art keywords
inhibitor
cell
cells
aza
parp
Prior art date
Application number
PCT/EP2016/055029
Other languages
English (en)
Inventor
Dmitry Bulavin
Doria FILIPPONI
Original Assignee
INSERM (Institut National de la Santé et de la Recherche Médicale)
Universite Nice Sophia Antipolis
Centre National De La Recherche Scientifique (Cnrs)
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 INSERM (Institut National de la Santé et de la Recherche Médicale), Universite Nice Sophia Antipolis, Centre National De La Recherche Scientifique (Cnrs) filed Critical INSERM (Institut National de la Santé et de la Recherche Médicale)
Publication of WO2016142427A1 publication Critical patent/WO2016142427A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0696Artificially 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/41Hedgehog proteins; Cyclopamine (inhibitor)
    • 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
    • C12N2501/65MicroRNA
    • 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
    • C12N2501/70Enzymes
    • C12N2501/72Transferases [EC 2.]
    • C12N2501/727Kinases (EC 2.7.)
    • 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
    • C12N2501/999Small molecules not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/09Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from epidermal cells, from skin cells, from oral mucosa cells
    • C12N2506/095Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from epidermal cells, from skin cells, from oral mucosa cells from mammary cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/30Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from cancer cells, e.g. reversion of tumour cells

Definitions

  • the invention relates to method and kit for reprogramming somatic cells.
  • the invention relates to a method for reprogramming somatic cells into pluripotent cells by using a combination of small-molecule compounds and kits comprising such compounds.
  • Yamanaka's lab reported that mouse embryonic fibroblast cells could be reprogrammed in induced pluripotent stem cells also called iPS cells, through retro-viral introduction of four transcriptional factors, Oct4, Sox2, Klf and c-Myc (Takahashi and Yamanaka 2006). After that, Oct4, Sox2, Klf and c-Myc were so-called reprogramming factors.
  • Yamanaka's lab and Thomson's lab both reported the generation of human iPS cells from adult human fibroblasts by the combination "Oct4, Sox2, Klf and c- Myc" or "Oct4, Sox2, Nanog, Lin-28", respectively (Takahashi et al.
  • IPS cells were very similar to embryonic stem cells in gene profiling, differentiation potential and epigenetic modifications. They were able to self-renew and differentiate into all mature cell types, including neurons, hematopoietic cells, muscle cells and islet cells.
  • iPS cells can be created by over-expression of one or more genes, for example one or more of the following four genes: Oct4, Sox2, c-Myc and Klf4 through retroviral infection, but with low efficiencies. All of these four genes are known to be or considered to be DNA binding proteins, transcription factors. Notably, the oncogene c-Myc used in this approach causes tumor formation in cells derived from the iPS cells.
  • iPS cells can be generated with only Oct4, Sox2 and Klf , the efficiency is even lower; fewer than iPS colonies from out of 100,000 cells. These issues pose significant barriers for creation of iPS cells for therapeutic applications.
  • One obvious concerns is the use of retroviruses, which integrate into chromosomal DNA and can cause ancillary problems (mutations).
  • European patent application N° EP2537920 discloses a method for producing an iPS cell, which comprises the steps of: introducing c-Myc into a somatic cell; and culturing said somatic cell in the presence of cell in the presence of a sirtuin inhibitor and/or a poly-ADP ribose polymerase (PARP) inhibitor.
  • PARP poly-ADP ribose polymerase
  • the invention relates to a method for reprogramming a human somatic cell into a human induced pluripotent stem cell (iPS) comprising a step of culturing said somatic cell into the presence of at least a combination of a DNA methyltransferase (DNMT) inhibitor, a Sirtuin inhibitor and a poly(ADP-ribose)polymerase (PARP) inhibitor.
  • iPS human induced pluripotent stem cell
  • the invention relates to an human iPS obtained by the method of the invention.
  • the invention in a third aspect, relates to a kit for reprogramming a somatic cell comprising at least a DNMT inhibitor, a Sirtuin inhibitor and a PARP inhibitor.
  • the invention is based on the discovery that a specific chemical combination enables the reprogramming of human somatic cells into a human induced pluripotent stem cell (iPS).
  • iPS human induced pluripotent stem cell
  • pluripotent refers to cells with the ability to give rise to progeny that can undergo differentiation, under appropriate conditions, into cell types that collectively exhibit characteristics associated with cell lineages from the three germ layers (endoderm, mesoderm, and ectoderm). Pluripotent stem cells can contribute to tissues of a prenatal, postnatal or adult organism. A standard art-accepted test, such as the ability to form a teratoma in 8-12 week old SCID mice, can be used to establish the pluripotency of a cell population. However, identification of various pluripotent stem cell characteristics can also be used to identify pluripotent cells.
  • human pluripotent stem cells may express at least some, and optionally all, of the markers from the following non- limiting list: SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, TRA-2-49/6E, Alkaline phosphatase (ALP), Sox2, E- cadherin, UTF-I, Oct4, Lin28, Rexl, and Nanog.
  • markers from the following non- limiting list: SSEA-3, SSEA-4, TRA-1-60, TRA-1-81, TRA-2-49/6E, Alkaline phosphatase (ALP), Sox2, E- cadherin, UTF-I, Oct4, Lin28, Rexl, and Nanog.
  • induced pluripotent stem cell refers to a pluripotent stem cell artificially derived from a non-pluripotent cell.
  • a non-pluripotent cell can be a cell of lesser potency to self-renew and differentiate than a pluripotent stem cell.
  • Cells of lesser potency can be, but are not limited to, somatic stem cells, tissue specific progenitor cells, primary or secondary cells.
  • reprogramming refers to the process of changing the fate of a target cell into that of a different cell type, caused by the expression of a small set of factors (or reprogramming factors) in the target cells.
  • factors or reprogramming factors
  • a "reprogramming factor” may be a transcription factor, which can be used to reprogram a target cell but further includes any analogue molecule that mimics the function of the factor with respect to reprogramming capacity.
  • germline cells sperm and egg
  • the somatic cell is an epithelial cell (such as a mammary gland epithelial cell), a fibroblast, a muscle cell, a cumulus cell, a neural cell, a liver cell, a GI tract cell, a mammary cell, a kidney cell, a blood cell, a vascular cell, a skin cell, an immune system cell, a lung cell, a bone cell, keratinocyte, or a pancreatic islet cell.
  • the term "DNMT inhibitor” refers to an inhibitor or antagonist of DNA methyltransferases (DNMTl, DNMT3A and/or DNMT3B) activity.
  • a DNMT inhibitor or antagonist is a compound that selectively inhibits the activity of DNMT.
  • a drug also able to decrease DNMTs expression is also considered as DNMT inhibitor.
  • DNMT inhibitors are well known from the skilled person. Examples of DNMT inhibitors are described in Fahy et al, Expert Opin. Ther. Patents (2012) 22(12): 1427-1442. A person skilled in the art can easily determine whether a compound is capable of inhibiting DNMT activity. Assays for evaluating DNMT activity are for example, described in Kim B Y et al, (Anal Biochem. 2004 Mar. 1; 326(1): 21-4) or in Yan L et al, (Cancer Biol Ther. 2003 September-October; 2(5):552-6).
  • DNMT inhibition may be determined using conventional methods, including for example assays that measure the methylation state of cellular DNA by incubation with 4 U of Sssl CpG methylase in the presence of 1.5 ⁇ S-adenosyl-L-[methyl- 3H]methionine, as described in Fang J et al, J Virol, 75(20): 9753-9761, 2001.
  • Non-limiting examples of DNMT inhibitors include 5-azacytidine (5-aza-CR; vidaza), 5-aza-2'-deoxycytidine (5-aza-dC; decitabine), l-[beta]-D-arabinofuranosyl-5-azacytosine, dihydro-5-azacytidine, zebularine, sinefungin, 5-fluoro-2'-deoxycyticine (FdCyd).
  • the DNMT inhibitor is 5-aza-2'- deoxycytidine (5-aza-dC; decitabine).
  • NU1025 is provided at a concentration range of between about from about 0.1 to about 20 ⁇ , e.g., between about 0.2-20 ⁇ , e.g., between about 0.5-15 ⁇ , e.g., between about 1-10 ⁇ , e.g., between about 2-8 ⁇ , e.g., between about 3-6 ⁇ , e.g., about 5 ⁇ .
  • Sirtuin inhibitor refers to an inhibitor or antagonist of sirtuin family of deacetylase enzymes (SIRT1-7) activity.
  • SIRT1-7 deacetylase enzymes
  • a Sirtuin inhibitor or antagonist is a compound that selectively inhibits the activity of Sirtuins.
  • a drug also able to decrease Sirtuin expression is also considered as DNMT inhibitor.
  • Sirtuins inhibitors are well known from the skilled person. Examples of Sirtuins inhibitors are described in Porcu et al, 2005 TRENDS in Pharmacological Sciences Vol.26 No.2 February 2005 and in Alcain et al., Expert Opinion on Therapeutic Patents, March 2009, Vol. 19, No. 3 : Pages 283-294.
  • Non-limiting examples of compounds which are known as Sirtuins inhibitors and which may be used include compounds and derivatives thereof from the class of Dihydrocoumarin derivatives, Naphthopyranone derivatives, NAD derivatives and 2- Hydro xy-naphtaldehyde derivatives.
  • the Sirtuin inhibitor is selected from the group consisting of Sirtuin inhibitors include Nicotinamide, Carbamido-NAD, NADH, Dihydrocoumarin, A3, Splitomicin, 2-OH-naphtaldehyde, Tenovin 1, Tenovin 6, AK-1, AK- 6, Selistat, Sirtinol and M15.
  • the Sirtuin inhibitor is Nicotinamide.
  • Nicotinamide is provided at a concentration range of between about 0.05 to about 50 ⁇ , e.g., from about 0.1 to about 40 ⁇ , e.g., between about 0.5-30 ⁇ , e.g., between about 1-25 ⁇ , e.g., between about 5-20 ⁇ , e.g., about 15 ⁇ .
  • PARP inhibitor refers to an inhibitor or antagonist of Poly(ADP-ribose) polymerases (PARP 1 and/or PARP2) activity.
  • a PARP inhibitor or antagonist is a compound that selectively inhibits the activity of PARP.
  • a drug also able to decrease PARPs expression is also considered as PARP inhibitor.
  • PARP inhibitors are well known from the skilled person. Examples of PARP inhibitors are described in Penning, Current Opinion In Drug Discovery & Development 2010 13 (5): 577-586. A person skilled in the art can easily determine whether a compound is capable of inhibiting PARP activity. Assays for evaluating PARP activity are for example, described in Poly(ADP-ribose) (PAR) polymer is a death signal (Andrabi SA et al., 2006). PARP inhibition may be determined using conventional methods, including for example dot blots (Affar EB et al., Anal Biochem.
  • Non-limiting examples of compounds which are known as PARP inhibitors and which may be used include compounds and derivatives thereof from the class of Nicotinamides, Benzamides, Isoquinolinones, Dihydroisoquinolinones, Benzimidazoles, indoles, Phthalazin-1 (2H)-ones, quinazolinones, Isoindolinones, Phenanthridines, phenanthhdinones, Benzopyrones, Unsaturated hydroximic acid derivatives and Pyridazines.
  • the PARP inhibitor is selected from the group consisting of NU1025 (8-hydroxy-2-methylquinazolinone), AZD-2281 (olaparib), AG014699 (rucaparib), ABT-888 (veliparib), BSI-201 (iniparib), CEP-9722, MK-4827, BMN-673, AG 14361, Nicotinamide and 3-AB (3-aminobenzamide).
  • the PARP inhibitor is NU1025 (8- hydroxy-2-methylquinazolinone).
  • NU1025 is provided at a concentration range of between about 0.05 to about 30 ⁇ , e.g., from about 0.1 to about 30 ⁇ , e.g., between about 0.2-30 ⁇ , e.g., between about 0.2-25 ⁇ , e.g., between about 0.5-20 ⁇ , e.g., between about 0,8-15 ⁇ , e.g., between about 1-10 ⁇ , e.g., between about 2-5 ⁇ , e.g., about 3 ⁇ .
  • a compound which is both a sirtuin inhibitor and a PARP inhibitor may be used (such as for instance Nicotinamide).
  • the method for reprogramming a human somatic cell into a human iPS comprising a step of culturing said somatic cell into the presence of at least a combination of a DNMT inhibitor as above- described (such as decitabine) and Nicotinamide as a sirtuin inhibitor and a PARP inhibitor.
  • a DNMT inhibitor such as decitabine
  • Nicotinamide as a sirtuin inhibitor and a PARP inhibitor
  • the somatic cell is further cultured in the presence of at least one compound selected from the group consisting of a c-Jun N-terminal kinase (JNK) inhibitor, a p38 mitogen-activated protein (MAP) kinase inhibitor and a Sonic Hedgehog (SHH) signaling pathway agonist.
  • JNK c-Jun N-terminal kinase
  • MAP mitogen-activated protein
  • SHH Sonic Hedgehog
  • JNK inhibitor refers to an inhibitor or antagonist of c-Jun N-terminal kinase (JNKl, JNK2 and/or JNK3) activity.
  • a JNK inhibitor or antagonist is a compound that selectively inhibits the activity of JNK.
  • a drug also able to decrease JNKs expression is also considered as JNK inhibitor.
  • JNK inhibitors are well known from the skilled person. Examples of JNK inhibitors are described in Zhang et al., Chem Biol. 2012 Jan 27; 19(1): 140-154. A person skilled in the art can easily determine whether a compound is capable of inhibiting JNK activity. Assays for evaluating JNK activity are for example, described in Guenat et al, J Biomol Screen, 2006; 11 : pages 1015-1026). JNK inhibition may be determined using conventional methods, including for example kinase assays (e.g. Alpha screen test) that measure the inhibition of human JNK mediated phosphorylation of a c-Jun substrate such as c-Jun, ATF2 and/or Elk-1.
  • kinase assays e.g. Alpha screen test
  • Non-limiting examples of compounds which are known as JNK inhibitors and which may be used include compounds and derivatives thereof from the class of Aryl-oxindole derivatives, Pyrazoloanthrones derivatives, Tetrahydro-pyrimidine derivatives, Benzazoles derivatives, Sulfonyl amino acid derivatives and Sulfonyl hydrazide derivatives.
  • JNK inhibitors include SP600125, AEG3482 and AS602801 (bentamapimo d) .
  • the JNK inhibitor is AS602801
  • AS602801 is provided at a concentration range of between about 0.05 to about 30 ⁇ , e.g., from about 0.1 to about 30 ⁇ , e.g., between about 0.2-30 ⁇ , e.g., between about 0.2-25 ⁇ , e.g., between about 0.2-20 ⁇ , e.g., between about 0.2-15 ⁇ , e.g., between about 0.2-10 ⁇ , e.g., between about 0.2-8 ⁇ , e.g., between about 0.2-6 ⁇ , e.g., between about 0.5-5 ⁇ , e.g., between about 1-4 ⁇ , e.g., about 3 ⁇ .
  • p38 inhibitor refers to an inhibitor or antagonist of p38 mitogen-activated kinase (MAPK) ( ⁇ 38 ⁇ , ⁇ 38 ⁇ , ⁇ 38 ⁇ , and ⁇ 38 ⁇ ) activity.
  • a p38 inhibitor or antagonist is a compound that selectively inhibits the activity of p38.
  • a drug also able to decrease p38 expression is also considered as p38 inhibitor.
  • p38 inhibitors are well known from the skilled person. Examples of p38 inhibitors are described in Genovese, Arthritis & Rheumatism Vol. 60, No. 2, Februrary 2009 pp 317-320 and in Buhler & Laufer, Expert Opinion on Theraputic Patents, May 2014, Vol. 24, No.
  • Assays for evaluating p38 activity are available such as the CycLex® p38 Kinase Assay/Inbibitor Screening kit purchased from MBL Inc.
  • Non- limiting examples of such p38 MAPK inhibitors include BIRB796
  • the p38 inhibitor is BIRB 796 (doramapimod).
  • BIRB 796 is provided at a concentration range of between about 0.1 to about 10 ⁇ , e.g., between about 0,5 ⁇ , e.g., between about 5 ⁇ , e.g., between about 2 ⁇ , e.g., between about 6 ⁇ , e.g., about 1 ⁇ .
  • SHH signaling pathway agonist refers to a compound
  • Hedgehog Hedgehog
  • Preferred hedgehog agonists can be used to overcome a ptc gain-of- function and/or a smoothened loss-of-function, the latter also being refered to as smoothened agonists.
  • Such agonist encompasses any compound that may act by directly activating the normal function of the hedgehog protein, but also to any compound that activates the Hh signalling pathway, and thus inhibits the function of ptc.
  • Hh signaling was first identified in Drosophila as an important regulatory mechanism for embryonic pattern formation.
  • the vertebrate family of Hedgehog genes includes three members that exist in mammals, known as Desert (Dhh), Sonic (Shh) and Indian (Ihh) Hedgehogs.
  • SHH signaling pathway agonists are well known from the skilled person. Examples of SHH signaling pathway agonists are described in the international patent applications N° WO0174344 and WO03027234 as well as in Carney & Ingham BMC Biology 2013, 11 :37.
  • Non-limiting examples of such SHH signaling pathway agonist include SAG (Hh- Agl .3) and purmorphamine (9-cyclohexyl-N-(4-morpholinophenyl)-2-(naphthalen-l-yloxy)- 9H-purin-6-amine) which activates the Hedgehog pathway by targeting Smoothened.
  • the SHH signaling pathway agonist is purmorphamine .
  • purmorphamine is provided at a concentration range of between about 0.1-10 ⁇ , e.g., between about 0.2-8 ⁇ , e.g., between about 0.5-5 ⁇ , e.g., about 1 ⁇ .
  • a compound which is simultaneously a sirtuin inhibitor and to lesser extend a PARP inhibitor may be used such as Nicotinamide.
  • the method for reprogramming a human somatic cell into a human induced pluripotent stem cell comprising a step of culturing said somatic cell into the presence of at least a combination of a DNA methyltransferase (DNMT) inhibitor as above described, Nicotinamide and at least a JNK inhibitor and/or a p38 inhibitor and/or a SHH signaling pathway agonist.
  • iPS human induced pluripotent stem cell
  • the method for reprogramming a human somatic cell into a human iPS comprising a step of culturing said somatic cell into the presence of at least a combination of a DNMT inhibitor, Nicotinamide, a JNK inhibitor, a p38 inhibitor and a SHH signaling pathway agonist (as described in EXAMPLE 4).
  • the method for reprogramming a human somatic cell into a human iPS comprising a step of culturing said somatic cell into the presence of at least a combination of a DNA methyltransferase (DNMT) inhibitor, a Sirtuin inhibitor and a poly(ADP-ribose)polymerase (PARP) inhibitor and macroH2A inhibitor.
  • DNMT DNA methyltransferase
  • Sirtuin inhibitor a poly(ADP-ribose)polymerase (PARP) inhibitor
  • macroH2A inhibitor macroH2A inhibitor
  • macroH2A refers to H2A histone variants due to the presence of a 30-kDa non-histone domain (macro domain) at their C-termini.
  • macroH2A variants are generally considered transcriptionally repressive in nature.
  • the level of macroH2A is regulated significantly at transcriptional level, several signalling pathways could be involved, typically, Protein kinase A-dependent signalling or Epithelial Mesenchymal Transition could play an important role.
  • the chemical modulation of either of these pathways could result in reduction of macroH2A levels and thus to improve reprogramming, as described in Example 6.
  • macroH2A inhibitor refers to an inhibitor or antagonist of the activity or expression of macroH2A.
  • the inhibitor of macroH2A expression is a small inhibitory RNAs (siRNAs).
  • macroH2A expression can be reduced by contacting the subject or cell with a small double stranded RNA (dsRNA), or a vector or construct causing the production of a small double stranded RNA, such that macroH2A expression is specifically inhibited (i.e. RNA interference or RNAi).
  • dsRNA small double stranded RNA
  • RNAi RNA interference
  • the macroH2A inhibitor expression is an endonuclease.
  • the mechanism behind endonuclease-based genome inactivating generally requires a first step of DNA single or double strand break, which can then trigger two distinct cellular mechanisms for DNA repair, which can be exploited for DNA inactivating: the errorprone nonhomologous end-joining (NHEJ) and the high-fidelity homo logy-directed repair (HDR).
  • Endonucleases for gene inactivation have come in various forms, which includes CRISPR)/CRISPR associated (Cas) systems, mega nucleases (MN), zinc finger nucleases (ZFN), and transcription activator- like effector nucleases (TALEN).
  • Endonucleases for use in the present invention are disclosed in WO 2010/079430, WO2011072246, WO2013045480, Mussolino C, et al (Curr Opin Biotechnol. 2012 Oct;23(5):644-50) and Papaioannou I. et al (Expert Opinion on Biological Therapy, March 2012, Vol. 12, No. 3 : 329-342).
  • the endonuclease is CRISPR-cas.
  • CRISPR-cas has its general meaning in the art and refers to clustered regularly interspaced short palindromic repeats associated which are the segments of prokaryotic DNA containing short repetitions of base sequences.
  • the endonuclease is CRISPR-cas9 which is from Streptococcus pyogenes.
  • the CRISPR/Cas9 system has been described in US 8697359 Bl and US 2014/0068797. Originally an adaptive immune system in prokaryotes (Barrangou and Marraffmi, 2014), CRISPR has been recently engineered into a new powerful tool for genome editing. It has already been successfully used to target important genes in many cell lines and organisms, including human (Mali et al, 2013, Science, Vol. 339 : 823-826), bacteria (Fabre et al, 2014, PLoS Negl. Trap. Dis., Vol.
  • the endonuclease is CRISPR-Cpfl which is the more recently characterized CRISPR from Provotella and Francisella 1 (Cpfl) in Zetsche et al. ("Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System (2015); Cell; 163, 1-13).
  • the macroH2A inhibitor is a Protein kinase C activator.
  • PKC Protein kinase C
  • PKC activator is Phorbol 12-myristate 13-acetate (PMA), such as described in Tahara et al 2009.
  • the macroH2A inhibitor is an EMT inducer.
  • the epithelial-mesenchymal transition is a process by which epithelial cells lose their cell polarity and cell-cell adhesion, and gain migratory and invasive properties to become mesenchymal stem cells; these are multipotent stromal cells that can differentiate into a variety of cell types.
  • EMT inducers could be TGF-beta and Wnt 5 a.
  • the step of culturing human somatic cells with the combination of compounds of the invention contained in a culture medium shall be carried out for the necessary time required for the production of iPS.
  • the duration of this culture step may be determined easily by one of skill in the art. For instance, during the culture the person skilled in the art can monitor the cultured cells for the expression of markers specifically expressed by iPS (e.g. pluripotency genes such as OCT4, SOX2, NANOG and KLF4) or activity of said cells such as Alkaline Phosphatase.
  • markers specifically expressed by iPS e.g. pluripotency genes such as OCT4, SOX2, NANOG and KLF4
  • activity of said cells such as Alkaline Phosphatase.
  • Monitoring of these markers can be performed using for instance RT-PCR analysis of RNA extracted from cultured cells with specific primers, immunofluorescence analysis with antibodies specific of the markers, ELISA and FACS or any method to detect the R A/protein/activity corresponding to the specific marker or activity.
  • the step of culturing somatic cells may be carried out for 1 to 20 days, preferably 10 days.
  • the culture medium of the invention can be renewed, partly or totally, at regular intervals.
  • the culture medium of the invention can be replaced with fresh culture medium of the invention every other day, for 10 days.
  • culture medium refers to any medium capable of supporting the growth and the differentiation of definitive endoderm cells into hepatic progenitor cells.
  • Preferred media formulations that will support the growth and reprogramming of a human somatic cell into a human iPS include chemically defined medium (CDM).
  • CDM chemically defined medium
  • the term “chemically defined medium” (CDM) refers to a nutritive solution for culturing cells which contains only specified components, preferably components of known chemical structure.
  • a chemically defined medium is a serum- free and feeder- free medium.
  • serum-free refers to a culture medium containing no added serum.
  • feeder-free refers to culture medium containing no added feeder cells.
  • the culture medium used by the invention may be a water-based medium that includes a combination of substances such as salts, nutrients, minerals, vitamins, amino acids, nucleic acids, proteins such as cytokines, growth factors (such as LIF, EGF and/or FGF) and hormones, all of which are needed for cell survival.
  • a culture medium according to the invention may be a synthetic tissue culture medium such as the DMEM (Dulbecco's Modified Eagle Medium) such as DMEM/F12, the RPMI (Roswell Park Memorial Institute medium) such as RPMI 1640 or the CMRL-1066 (Connaught Medical Research Laboratory) for human use, supplemented with additives as is further described below (Section Examples) such as B27.
  • the B-27 supplement contains SOD, catalase and other anti- oxidants, and unique fatty acids, such as linoleic acid and linolenic acid.
  • the invention in a second aspect, relates to an human iPS obtained by the method of the invention.
  • the invention in a third aspect, relates to a kit for reprogramming a somatic cell comprising at least a DNMT inhibitor, a Sirtuin inhibitor and a PARP inhibitor.
  • the kit according to the invention wherein the DNMT inhibitor is 5-aza-2'- deoxycytidine (5-aza-dC; decitabine), the Sirtuin inhibitor is nicotinamide and the PARP inhibitor is NU 1025 (8-hydroxy-2-methylquinazolinone).
  • the DNMT inhibitor is 5-aza-2'- deoxycytidine (5-aza-dC; decitabine)
  • the Sirtuin inhibitor is nicotinamide
  • the PARP inhibitor is NU 1025 (8-hydroxy-2-methylquinazolinone).
  • kit according to the invention further comprising at least a JNK inhibitor, a p38 inhibitor or a SHH signaling pathway agonist.
  • FIGURES are a diagrammatic representation of FIGURES.
  • Figure 1 Levels of expression of pluripotency genes in human cells treated with one compound (A) or with a combination of three compounds (B).
  • Figure 2 Levels of expression of different genes relative to the level for non- treated hMep cells.
  • FIG. 3 siRNA knockdown of the expression of mH2Al.l strongly de-repress pluripotency genes in HCT116 isogenic cell line after 3i treatment.
  • HCT116 wt cells were pre-treated with 5uM 5-AZA, 5uM 5-AZA + PARPi or 3i treatment for 3 days and then transfected with siRNA specific for mH2A.l or with scramble siRNA. Forty-eight hours post- transfection the cells were starved and RNA isolated for RT-PCR analysis.
  • the siRNA treatment efficiently suppresses the expression of mH2Al .
  • the level of mH2A was checked after siRNA knock-down by RT-PCR and normalized to both scramble and housekeeping gene.
  • FIG. 4 (B) Real-time PCR quantification of the relative expression of Oct4 in Hctl l6 pre- treated with 5uM 5-AZA, 5uM 5-AZA + PARPi or 3i transfected with siRNA specific for mH2A.1 or with scramble siRNA.
  • Figure 4 ETM induction cooperates with 3i in activation of endogenous Oct4 locus.
  • HCT116 cells with Crispr/Cas integrated Cre recombinase into endogenous Oct4 locus were pre-treated with StemXVivo EMT Inducing Media Supplement (R&D Systems) (left panel) or PMA (right panel) and 3i cocktail was added 3 days later. The analysis of endogenous Oct4 locus was carried out 24h later. Bright green cells represent cells expressing high level of endogenous Oct4.
  • EXAMPLE 1 Identifying the combination of chemical drugs to improve the re- activation of pluripotent genes.
  • the human colon cancer cell lines HCT116 were maintained in DMEM containing 10% heat-inactivated FCS, 100 U/ml penicillin and lmM glutamine.
  • the human breast cancer cell lines MCF7 were maintained in RPMI1640 containing 10% of heat- inactivated FCS, 100 U/ml penicillin and lmM glutamine and lmM AA. Both cell lines were incubated at 37°C, 5%> C0 2 -95%> air using standard tissue culture incubators.
  • Drugs treatment Both MCF7 and HCT116 cells were sited at 30% confluence on 6 well plate 1 day before treatment.
  • the mRNA transcription levels of pluripotency genes including OCT4, SOX2, NANOG and KLF4 were determined using a real-time quantitative PCR system.
  • Relative quantitation was performed using the comparative Ct method with data from the ABI PRISM 7000 Detection System following the manufacturer's protocol. The Ct values were measured, and the average Ct of triplicate samples was calculated. Alteration of mRNA expression was defined as a 3-fold difference in the expression level after treatment, relative to that before treatment and to Gapdh or 18S.
  • pluripotent genes are silent in adult tissues with only exception for germ cells. Based on our previous analysis of Wipl deficient cells, we speculated that chemical treatment with a specific set of inhibitors could result in re-activation of pluripotency genes". To access this further, we analyzed the epigenetic regulation of pluripotent-associated genes NANOG, OCT4, c-MYC, KLF4, and SOX2, and their correlation with gene expression in different cancer cell lines using a combination of epigenetic and DNA damage modulators with known chemical compounds.
  • EXAMPLE 2 Induction of pluripotent genes in primary human cells and improvement of the protocol with additional cocktail of the chemical compounds.
  • Human primary mammary gland epithelial cells (hMep) were plated on 6-well plates in DMEM/F12 medium containing 20ng/ml human LIF and B27. The drug treatment started on day 2 after plating and continued for 10 days. The drugs were changed every second day.
  • 1% DMSO as a control
  • 3i containing 5-aza-dC 5 ⁇
  • Nicotinamide 15mM PARPi NU 1025 ⁇
  • 6i (3i + .INK. inhibitor Bentamapimod 10 iiM; p38 inhibitor BIRB 796 3 ⁇ , ⁇ and SHH agonist Purmorphamine 1 ⁇ ).
  • the cells were harvested and mRNA was purified for analysis of pluripotent genes.
  • RNA from cells was isolated using RNeasy mini kit (Qiagen) according the standard protocol, ⁇ g of RNA was reverse transcribed into oligo-dT primed cDNA with PowerScript II Reverse transcriptase (Life Technology) and 40-50 ng of cDNA were used for individual PCR.
  • the quantitative PCR were performed using KAPA SYBR Fast qPCR kit (KAPA Biosystems) according to manufacturer protocol in Applied Biosystems 7300 Real- Time PCR System.
  • KAPA Biosystems KAPA SYBR Fast qPCR kit according to manufacturer protocol in Applied Biosystems 7300 Real- Time PCR System.
  • the collected Ct data were normalised to GAPDH Ct and quantified to obtain relative fold changes using Microsoft Excel program. Results
  • EXAMPLE 3 Acquisition of pluripotent state after 10 days of treatment.
  • Material & Methods Human primary mammary gland epithelial cells (hMep) were plated on 6-well plates in DMEM/F12 medium containing lOng/ml EGF, 10 ng/ml FGF and 1% B27. The drug treatment started second day after plating and continued for 10 days. The drugs were changed every day for the first 4 days and subsequently every second day for the rest of the treatment.
  • hMep Human primary mammary gland epithelial cells
  • DMEM/F12 medium containing lOng/ml EGF, lOng/ml FGF and 1% B27.
  • the drug treatment started second day after plating and continued for 10 days.
  • the drugs were changed every day for the first 4 days and subsequently every second day for the rest of the treatment.
  • Alkaline phosphatase activity is considered to be a marker of pluripotency in embryonic stem cells and is detected in iPS after the reprogramming process using a standard protocol with overexpression of 4 factors, Oct4, Klf4, Sox2 and Myc.
  • the second condition is the pre-treatment with Protein kinase C activator, PMA. Similar to EMT induction, this pre-treatment has a strong cooperative effect with 3i treatment on induction of endogenous Oct4 locus (Figure 2, right panel).

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Microbiology (AREA)
  • Transplantation (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne un procédé permettant la reprogrammation d'une cellule somatique humaine en une cellule souche pluripotente induite humaine (iPS), le procédé comprenant une étape de mise en culture de ladite cellule somatique en présence d'au moins une combinaison d'un inhibiteur d'ADN méthyltransférase (DNMT), d'un inhibiteur de la Sirtuine et d'un inhibiteur de poly (ADP-ribose) polymérase (PARP).
PCT/EP2016/055029 2015-03-10 2016-03-09 Procédé et kit pour la reprogrammation de cellules somatiques WO2016142427A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15305361 2015-03-10
EP15305361.6 2015-03-10

Publications (1)

Publication Number Publication Date
WO2016142427A1 true WO2016142427A1 (fr) 2016-09-15

Family

ID=52686300

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/055029 WO2016142427A1 (fr) 2015-03-10 2016-03-09 Procédé et kit pour la reprogrammation de cellules somatiques

Country Status (1)

Country Link
WO (1) WO2016142427A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3102199A4 (fr) * 2014-01-21 2018-01-17 The Johns Hopkins University Schéma thérapeutique et procédés pour sensibiliser des cellules cancéreuses traitées avec une thérapie épigénétique à des inhibiteurs parp dans des cancers multiples
WO2019127697A1 (fr) * 2017-12-29 2019-07-04 诺未科技(北京)有限公司 Système de culture permettant d'amplifier une cellule souche hématopoïétique et/ou une cellule progénitrice hématopoïétique et procédé s'y rapportant, cellule souche hématopoïétique et cellule progénitrice hématopoïétique
CN113462638A (zh) * 2021-06-30 2021-10-01 呈诺再生医学科技(珠海横琴新区)有限公司 一种高效无遗传修饰的iPSC诱导、产业化单克隆挑取平台及应用
CN118048296A (zh) * 2024-04-16 2024-05-17 成都赛济元生物医药有限公司 一种用于细胞重编程的培养体系、试剂盒及方法

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
WO2001068836A2 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
WO2001074344A2 (fr) 2000-03-30 2001-10-11 Curis, Inc. Petits regulateurs de molecules organiques de proliferation de cellules
WO2003027234A2 (fr) 2001-09-26 2003-04-03 Curis, Inc. Regulateurs de proliferation cellulaire se presentant sous la forme de molecules organiques de petites dimensions
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
WO2009061442A1 (fr) * 2007-11-06 2009-05-14 Children's Medical Center Corporation Procédé de production de cellules souches pluripotentes induites (ips) à partir de cellules humaines non embryonnaires
WO2009102983A2 (fr) * 2008-02-15 2009-08-20 President And Fellows Of Harvard College Induction efficace de cellules souches multipotentes à l’aide de composés à petites molécules
WO2010079430A1 (fr) 2009-01-12 2010-07-15 Ulla Bonas Domaines modulaires de liaison à l'adn et procédés d'utilisation
WO2011072246A2 (fr) 2009-12-10 2011-06-16 Regents Of The University Of Minnesota Modification de l'adn induite par l'effecteur tal
EP2537920A1 (fr) 2010-02-16 2012-12-26 Saitama Medical University Procédé de production de cellule souche pluripotente artificielle
WO2013045480A1 (fr) 2011-09-26 2013-04-04 Justus-Liebig-Universität Giessen Nucléases chimériques pour ciblage de gènes
US20140068797A1 (en) 2012-05-25 2014-03-06 University Of Vienna Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999032619A1 (fr) 1997-12-23 1999-07-01 The Carnegie Institution Of Washington Inhibition genetique par de l'arn double brin
US6506559B1 (en) 1997-12-23 2003-01-14 Carnegie Institute Of Washington Genetic inhibition by double-stranded RNA
US6573099B2 (en) 1998-03-20 2003-06-03 Benitec Australia, Ltd. Genetic constructs for delaying or repressing the expression of a target gene
WO2001036646A1 (fr) 1999-11-19 2001-05-25 Cancer Research Ventures Limited Inhibition d"expression genique a l"aide d"arn bicatenaire
WO2001068836A2 (fr) 2000-03-16 2001-09-20 Genetica, Inc. Procedes et compositions d'interference d'arn
WO2001074344A2 (fr) 2000-03-30 2001-10-11 Curis, Inc. Petits regulateurs de molecules organiques de proliferation de cellules
WO2003027234A2 (fr) 2001-09-26 2003-04-03 Curis, Inc. Regulateurs de proliferation cellulaire se presentant sous la forme de molecules organiques de petites dimensions
WO2009061442A1 (fr) * 2007-11-06 2009-05-14 Children's Medical Center Corporation Procédé de production de cellules souches pluripotentes induites (ips) à partir de cellules humaines non embryonnaires
WO2009102983A2 (fr) * 2008-02-15 2009-08-20 President And Fellows Of Harvard College Induction efficace de cellules souches multipotentes à l’aide de composés à petites molécules
WO2010079430A1 (fr) 2009-01-12 2010-07-15 Ulla Bonas Domaines modulaires de liaison à l'adn et procédés d'utilisation
WO2011072246A2 (fr) 2009-12-10 2011-06-16 Regents Of The University Of Minnesota Modification de l'adn induite par l'effecteur tal
EP2537920A1 (fr) 2010-02-16 2012-12-26 Saitama Medical University Procédé de production de cellule souche pluripotente artificielle
WO2013045480A1 (fr) 2011-09-26 2013-04-04 Justus-Liebig-Universität Giessen Nucléases chimériques pour ciblage de gènes
US20140068797A1 (en) 2012-05-25 2014-03-06 University Of Vienna Methods and compositions for rna-directed target dna modification and for rna-directed modulation of transcription
US8697359B1 (en) 2012-12-12 2014-04-15 The Broad Institute, Inc. CRISPR-Cas systems and methods for altering expression of gene products

Non-Patent Citations (41)

* Cited by examiner, † Cited by third party
Title
AFFAR EB ET AL., ANAL BIOCHEM., vol. 259, no. 2, 1998, pages 280 - 3
AGNÈS I. LUKASZEWICZ ET AL: "Small Molecules and Stem Cells. Potency and Lineage Commitment: The New Quest for the Fountain of Youth", JOURNAL OF MEDICINAL CHEMISTRY, vol. 53, no. 9, 13 May 2010 (2010-05-13), pages 3439 - 3453, XP055191381, ISSN: 0022-2623, DOI: 10.1021/jm901361d *
ALCAIN ET AL., EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 19, no. 3, March 2009 (2009-03-01), pages 283 - 294
ALEXANDRE GASPAR-MAIA ET AL: "MacroH2A histone variants act as a barrier upon reprogramming towards pluripotency", NATURE COMMUNICATIONS, vol. 4, 5 March 2013 (2013-03-05), pages 1565, XP055263465, DOI: 10.1038/ncomms2582 *
BUHLER; LAUFER, EXPERT OPINION ON THERAPUTIC PATENTS, vol. 24, no. 5, May 2014 (2014-05-01), pages 535 - 554
CARNEY; INGHAM, BMC BIOLOGY, vol. 11, 2013, pages 37
DE CARVALHO D D ET AL: "DNA methylation and cellular reprogramming", TRENDS IN CELL BIOLOGY, ELSEVIER SCIENCE LTD, XX, vol. 20, no. 10, 1 October 2010 (2010-10-01), pages 609 - 617, XP027331701, ISSN: 0962-8924, [retrieved on 20100924] *
DICARLO ET AL., NUCLEIC ACIDS RES., vol. 41, 2013, pages 4336 - 4343
FABRE ET AL., PLOS NEGL. TROP. DIS., vol. 8, 2014, pages E2671
FAHY ET AL., EXPERT OPIN. THER. PATENTS, vol. 22, no. 12, 2012, pages 1427 - 1442
FANG J ET AL., J VIROL, vol. 75, no. 20, 2001, pages 9753 - 9761
GENOVESE, ARTHRITIS & RHEUMATISM, vol. 60, no. 2, February 2009 (2009-02-01), pages 317 - 320
GRATZ ET AL., GENETICS, 2014
GUENAT ET AL., J BIOMOL SCREEN, vol. 11, 2006, pages 1015 - 1026
GUO ET AL., DEVELOPMENT, vol. 141, 2014, pages 707 - 714
HAI ET AL., CELL RES., 2014
HOU P; LI Y; ZHANG X; LIU C; GUAN J; LI H; ZHAO T; YE J; YANG W; LIU K: "Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds", SCIENCE, vol. 341, no. 6146, 9 August 2013 (2013-08-09), pages 651 - 4
HUANGFU DANWEI ET AL: "Induction of pluripotent stem cells by defined factors is greatly improved by small-molecule compounds", NATURE BIOTECHNOLOGY, NATURE PUBLISHING GROUP, US, vol. 26, no. 7, 1 July 2008 (2008-07-01), pages 795 - 797, XP002502536, ISSN: 1087-0156, [retrieved on 20080622], DOI: 10.1038/NBT1418 *
HWANG ET AL., PLOS ONE, vol. 8, 2013, pages E68708
K.J. DILLON ET AL., JOURNAL OFBIOMOLECULAR SCREENING, vol. 8, no. 3, 2003, pages 347 - 352
KIM B Y ET AL., ANAL BIOCHEM, vol. 326, no. 1, 1 March 2004 (2004-03-01), pages 21 - 4
LI Y; ZHANG Q; YIN X; YANG W; DU Y; HOU P; GE J; LIU C; ZHANG W; ZHANG X: "Generation of iPSCs from mouse fibroblasts with a single gene, Oct4, and small molecules", CELL RES., vol. 21, no. 1, January 2011 (2011-01-01), pages 196 - 204
LUKASZEWICZ AI; MCMILLAN MK; KAHN M.: "Small molecules and stem cells. Potency and lineage commitment: the new quest for the fountain of youth", J MED CHEM., vol. 53, no. 9, 13 May 2010 (2010-05-13), pages 3439 - 53
MA ET AL., CELL RES., vol. 24, 2014, pages 122 - 125
MALI ET AL., SCIENCE, vol. 339, 2013, pages 823 - 826
MASHIKO ET AL., DEV. GROWTH DIFFER., vol. 56, 2014, pages 122 - 129
MIKKELSEN TARJEI S ET AL: "Dissecting direct reprogramming through integrative genomic analysis", NATURE, NATURE PUBLISHING GROUP, UNITED KINGDOM, vol. 454, no. 7200, 3 July 2008 (2008-07-03), pages 49 - 55, XP002564354, ISSN: 0028-0836, [retrieved on 20080528], DOI: 10.1038/NATURE07056 *
MUSSOLINO C ET AL., CURR OPIN BIOTECHNOL., vol. 23, no. 5, October 2012 (2012-10-01), pages 644 - 50
NIU ET AL., CELL, vol. 156, 2014, pages 836 - 843
P. HOU ET AL: "Pluripotent Stem Cells Induced from Mouse Somatic Cells by Small-Molecule Compounds", SCIENCE, vol. 341, no. 6146, 18 July 2013 (2013-07-18), pages 651 - 654, XP055079363, ISSN: 0036-8075, DOI: 10.1126/science.1239278 *
PAPAIOANNOU I. ET AL., EXPERT OPINION ON BIOLOGICAL THERAPY, vol. 12, no. 3, March 2012 (2012-03-01), pages 329 - 342
PENNING, CURRENT OPINION IN DRUG DISCOVERY & DEVELOPMENT, vol. 13, no. 5, 2010, pages 577 - 586
PORCU ET AL., TRENDS IN PHARMACOLOGICAL SCIENCES, vol. 26, no. 2, 2005
SMITH ET AL., ANAL BIOCHEM., vol. 394, no. 1, 1 November 2009 (2009-11-01), pages 101 - 109
TAKAHASHI KL; TANABE K; OHNUKI M; NARITA M; ICHISAKA T; TOMODA K; YAMANAKA S: "Induction of pluripotent stem cells from adult human fibroblasts by defined factors;", CELL, vol. 131, no. 5, 2007, pages 861 - 872
TAKAHASHI, K.; YAMANAKA, S.: "Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors", CELL, vol. 126, no. 4, 2006, pages 663 - 676
YAN L ET AL., CANCER BIOL THER., vol. 2, no. 5, September 2003 (2003-09-01), pages 552 - 6
YANG ET AL., J. MOL. CELL BIOL., vol. 6, 2014, pages 97 - 99
YU J; VODYANIK MA; SMUGA-OTTO K; ANTOSIEWICZ-BOURGET J; FRANE JL; TIAN S; NIE J; JONSDOTTIR GA; RUOTTI V; STEWART R: "Induced pluripotent stem cell lines derived from human somatic cells.", SCIENCE, vol. 318, no. 5858, 2007, pages 1917 - 1920
ZETSCHE ET AL.: "Cpfl is a Single RNA-guided Endonuclease of a Class 2 CRISPR-Cas System", CELL, vol. 163, 2015, pages 1 - 13
ZHANG ET AL., CHEM BIOL., vol. 19, no. 1, 27 January 2012 (2012-01-27), pages 140 - 154

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3102199A4 (fr) * 2014-01-21 2018-01-17 The Johns Hopkins University Schéma thérapeutique et procédés pour sensibiliser des cellules cancéreuses traitées avec une thérapie épigénétique à des inhibiteurs parp dans des cancers multiples
US10105383B2 (en) 2014-01-21 2018-10-23 University Of Maryland, Baltimore Therapy regimen and methods to sensitize cancer cells treated with epigenetic therapy to PARP inhibitors in lung cancer
US10105382B2 (en) 2014-01-21 2018-10-23 University Of Maryland, Baltimore Therapy regimen and methods to sensitize cancer cells treated with epigenetic therapy to PARP inhibitors in ovarian cancer
US10363264B2 (en) 2014-01-21 2019-07-30 University Of Maryland, Baltimore Therapy regimen and methods to sensitize cancer cells treated with epigenetic therapy to PARP inhibitors in multiple cancers
WO2019127697A1 (fr) * 2017-12-29 2019-07-04 诺未科技(北京)有限公司 Système de culture permettant d'amplifier une cellule souche hématopoïétique et/ou une cellule progénitrice hématopoïétique et procédé s'y rapportant, cellule souche hématopoïétique et cellule progénitrice hématopoïétique
CN113462638A (zh) * 2021-06-30 2021-10-01 呈诺再生医学科技(珠海横琴新区)有限公司 一种高效无遗传修饰的iPSC诱导、产业化单克隆挑取平台及应用
CN118048296A (zh) * 2024-04-16 2024-05-17 成都赛济元生物医药有限公司 一种用于细胞重编程的培养体系、试剂盒及方法

Similar Documents

Publication Publication Date Title
Collinson et al. Deletion of the polycomb-group protein EZH2 leads to compromised self-renewal and differentiation defects in human embryonic stem cells
Shan et al. PRC2 specifies ectoderm lineages and maintains pluripotency in primed but not naive ESCs
Hernandez et al. Dppa2/4 facilitate epigenetic remodeling during reprogramming to pluripotency
Luo et al. NuRD blocks reprogramming of mouse somatic cells into pluripotent stem cells
Chen et al. Molecular basis of the first cell fate determination in mouse embryogenesis
Betschinger et al. Exit from pluripotency is gated by intracellular redistribution of the bHLH transcription factor Tfe3
Guo et al. microRNA-29b is a novel mediator of Sox2 function in the regulation of somatic cell reprogramming
EP2766474B1 (fr) Inhibition et amélioration de la re-programmation par des enzymes de modification de la chromatine
D'aniello et al. A novel autoregulatory loop between the Gcn2-Atf4 pathway and L-Proline metabolism controls stem cell identity
Jiang et al. MicroRNA-137 represses Klf4 and Tbx3 during differentiation of mouse embryonic stem cells
Diaz Perez et al. Derivation of new human embryonic stem cell lines reveals rapid epigenetic progression in vitro that can be prevented by chemical modification of chromatin
Huang et al. Impairment of preimplantation porcine embryo development by histone demethylase KDM5B knockdown through disturbance of bivalent H3K4me3-H3K27me3 modifications
US11377636B2 (en) Endogenous retrovirus transcription as a marker for primate naive pluripotent stem cells
Lynch et al. Global hyperactivation of enhancers stabilizes human and mouse naive pluripotency through inhibition of CDK8/19 Mediator kinases
WO2016142427A1 (fr) Procédé et kit pour la reprogrammation de cellules somatiques
Deng et al. Aberrant DNA and histone methylation during zygotic genome activation in goat cloned embryos
Collins et al. HOX genes in normal, engineered and malignant hematopoiesis
US20210363495A1 (en) Culture media for pluripotent stem cells
Fu et al. Involvement of histone acetylation of Sox17 and Foxa2 promoters during mouse definitive endoderm differentiation revealed by microRNA profiling
EP3452578A1 (fr) Procédés de fabrication in vitro de tissu de fundus d'estomac et compositions associées à celui-ci
Jessop et al. Developmental functions of the dynamic DNA methylome and hydroxymethylome in the mouse and zebrafish: similarities and differences
Huang et al. Geminin is essential to prevent DNA re-replication-dependent apoptosis in pluripotent cells, but not in differentiated cells
Rejano-Gordillo et al. Aryl hydrocarbon receptor: From homeostasis to tumor progression
Zeevaert et al. YAP1 is essential for self-organized differentiation of pluripotent stem cells
Shukla et al. Activation of transcription factor circuity in 2i-induced ground state pluripotency is independent of repressive global epigenetic landscapes

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16708699

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16708699

Country of ref document: EP

Kind code of ref document: A1