WO2013030243A1 - Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use - Google Patents

Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use Download PDF

Info

Publication number
WO2013030243A1
WO2013030243A1 PCT/EP2012/066793 EP2012066793W WO2013030243A1 WO 2013030243 A1 WO2013030243 A1 WO 2013030243A1 EP 2012066793 W EP2012066793 W EP 2012066793W WO 2013030243 A1 WO2013030243 A1 WO 2013030243A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
ipam
paraxial mesoderm
spondin
wnt
Prior art date
Application number
PCT/EP2012/066793
Other languages
French (fr)
Inventor
Olivier Pourquie
Jérôme CHAL
Original Assignee
Inserm (Institut National De La Sante Et De La Recherche Medicale)
Cnrs (Centre National De La Recherche Scientifique)
Universite De Strasbourg
Association Francaise Contre Les Myopathies
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
Priority claimed from EP11306080A external-priority patent/EP2565264A1/en
Application filed by Inserm (Institut National De La Sante Et De La Recherche Medicale), Cnrs (Centre National De La Recherche Scientifique), Universite De Strasbourg, Association Francaise Contre Les Myopathies filed Critical Inserm (Institut National De La Sante Et De La Recherche Medicale)
Priority to EP12753961.7A priority Critical patent/EP2756075B1/en
Priority to DK12753961T priority patent/DK2756075T3/en
Priority to ES12753961T priority patent/ES2761859T3/en
Priority to EP19200815.9A priority patent/EP3611257A1/en
Priority to JP2014527647A priority patent/JP6570833B2/en
Priority to CA2847325A priority patent/CA2847325C/en
Priority to US14/342,251 priority patent/US10240123B2/en
Publication of WO2013030243A1 publication Critical patent/WO2013030243A1/en
Priority to IL231205A priority patent/IL231205B/en

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
    • C12N5/0603Embryonic 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
    • C12N5/0625Epidermal cells, skin cells; Cells of the oral mucosa
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0653Adipocytes; Adipose tissue
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0654Osteocytes, Osteoblasts, Odontocytes; Bones, Teeth
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0658Skeletal muscle cells, e.g. myocytes, myotubes, myoblasts
    • 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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem 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
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/60Buffer, e.g. pH regulation, osmotic pressure
    • C12N2500/62DMSO
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • 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
    • C12N2501/415Wnt; Frizzeled
    • 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
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells

Definitions

  • the present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
  • iPAM induced paraxial mesoderm progenitor
  • Embryonic stem (ES) cell research offers unprecedented potential for understanding fundamental developmental processes, such as lineage differentiation.
  • Embryonic stem cell lines are derived from early embryos and are characterized by their ability to self-renew, that is, to be maintained indefinitely in a proliferative and undifferentiated state in culture.
  • ES cells are also pluripotent, meaning they retain the capacity to differentiate into the three embryonic lineages: ectoderm, mesoderm and endoderm plus all of their derivatives (Chambers I., 2004).
  • ectoderm ectoderm
  • mesoderm and endoderm plus all of their derivatives
  • Such reprogrammed cell lines have already been generated from patients with a variety of diseases, such as Duchenne Muscular Dystrophy or Amyotrophic lateral sclerosis (ALS) and differentiation of the reprogrammed cells into the deficient tissue has been achieved for iPS cells from patients affected with several diseases such as ALS, thus, demonstrating the feasibility of the approach (Dimos et al, 2008; Park et al, 2008a).
  • diseases such as Duchenne Muscular Dystrophy or Amyotrophic lateral sclerosis (ALS)
  • T Brachyury
  • Msgnl Mesogeninl
  • Rspo3 also called Cristinl, Thsd2
  • Thsd2 is strongly expressed in the PSM and somites, as well as later in condensing mesenchymal cells, (Kazanskaya et al, 2004; Nam et al, 2007).
  • R-spondins (Rspo l to 4 genes) are secreted molecules containing a thrombospondin domain, that can activate canonical Wnt signaling and Beta-Catenin, via the Fzd/LRP/Lgr4/Lgr5 co-receptors complex (Carmon et al, 2011 ; de Lau et al, 2011 ; Kim et al, 2008; Nam et al, 2006), but they were also shown to bind Syndecan4 and induce Wnt/PCP signaling (Ohkawara et al, 2011). Interestingly, biochemical assays show that Rspo2 and 3 are more potent to activate Wnt signaling than Rspol and 4 (Kim et al, 2008).
  • R-spondins have also been shown to be implicated in bone formation and chondro genesis (Hankenson et al, 2010; Jin et al, 2011 ; Ohkawara et al, 2011), myogenesis (Han et al, 2011; Kazanskaya et al, 2004) and angiogenesis (Kazanskaya et al, 2008).
  • BMPs Bone Morpho genetic Proteins
  • TGFbeta superfamily BMP receptor type I and type II
  • BMPR-I and -II BMP receptor type I and type II
  • BMPR-I can consist of Activin receptor-like kinase (ALK)-2/3 and 6 (also known as ActR-IA , BMPR-IA and BMPR-IB respectively).
  • ALK Activin receptor-like kinase
  • BMPR-II can consist of BMPR-II, ActR- IIA and ActR-IIB.
  • the BMP receptor complex is formed by an heterotetrameric complex of two BMPR-I and two BMPR-II.
  • the BMP receptor contains an intracytoplasmic serine/threonine kinase domain which allows the phosphorylation of Smad 1/5/8 upon binding of the BMP dimer. Phosphorylated Smadl/5/8 then associate to Smad4 and shuttle to the nucleus to activate target genes, which include the inhibitor of DNA binding (Id) 1/2/3 genes - -
  • BMP/TGFP secreted agonists and antagonists have been described to regulate and fine-tune BMP signaling during development. Most notably noggin, chordin, follistatin and gremlin block BMP signaling by sequestrating secreted BMP, preventing its binding to the receptor.
  • BMP ligands prominently BMP2, 4 and 7
  • BMP receptors Smads, Co-Smads and BMP agonists/antagonists have been implicated in mesoderm specification and organogenesis during development [Derynck Rik, 2008; Reshef R. et al, Gen Dev 1998; Wijgerde M. et al, 2005; McMahon JA et al, 1998; Stafford DA et al, 2011; Pourquie O. et al, 1996 and Tonegawa A. et al, 1997].
  • the antibody against satellite cells SM/C- 2.6 was also used to isolate myogenic cells differentiated from mouse ES and iPS cells (Fukada et al, 2004; Mizuno et al, 2010).
  • mesoderm precursors differentiated from mouse ES cells were also isolated based on their expression of other surface markers such as the Platelet derived growth factor receptor alpha (PDGFRa) or Vascular endothelial growth factor receptor 2 (VEGFR2) ((Sakurai et al, 2009; Sakurai et al, 2008) Sakurai H. et al, 2006; Takebe A. et al, 2006). Whether this combination of markers is strictly specific for paraxial mesoderm precursors has however not been demonstrated.
  • PDGFRa Platelet derived growth factor receptor alpha
  • VEGFR2 Vascular endothelial growth factor receptor 2
  • the second strategy is based on forced expression of the transcription factors Pax3 or MyoD, or of the secreted factor Insulin Growth Factor 2 (IGF-2) in mouse ES cells (Darabi et al, 2008; Darabi et al, 2011; Dekel et al, 1992; Prelle et al, 2000; Shani et al, 1992).
  • these strategies show either limited efficiency or require introduction of exogenous DNA in the ES cells which is a major hurdle for the development of safe cell therapies and the differentiated cells often show limited proliferation and engraftment potential.
  • the present invention fulfils this need by providing a method for preparing multipotent progenitor cell lines expressing markers of the paraxial mesoderm progenitors and referred to as induced Paraxial Mesoderm progenitor cells or iPAM to distinguish them from the natural embryo Paraxial Mesoderm progenitor cells.
  • the iPAM cells are capable of giving rise to cell lineages of the muscular, skeletal (bone and cartilage), dermal tissue, and derivatives such as adipocytes and endothelium.
  • the inventors have shown that embryonic stem cells or pluripotent reprogrammed cells (iPS) can be differentiated into induced Paraxial Mesoderm progenitor (iPAM) cells using a limited number of factors.
  • the inventors have made the surprising finding that it is possible to efficiently obtain induced Paraxial Mesoderm progenitor (iPAM) cells by treatment with specific factors, without any genetic modification of the target cells. They have shown that the obtained induced Paraxial Mesoderm progenitor (iPAM) cells exhibit characteristics of endogenous Paraxial mesoderm progenitor cells.
  • the invention is the first description of a method for obtaining unlimited amounts of cells suitable for use as progenitor cells for regenerating either muscle, skeletal, adipose or dermal tissues and paraxial mesoderm derived endothelium. Therefore the invention is highly useful in particular in regenerative medicine.
  • the present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signalling pathway.
  • iPAM induced paraxial mesoderm progenitor
  • the present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
  • iPAM induced paraxial mesoderm progenitor
  • the invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of a - - member of the R-spondin family and an effective amount of an inhibitor of the Bone Morpho genetic Protein (BMP) signaling pathway.
  • iPAM induced paraxial mesoderm progenitor
  • the invention also relates to an alternate method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of a member of an inhibitor of the GSK-3B and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
  • BMP Bone Morphogenetic Protein
  • a first aspect of the invention relates to an ex vivo method for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention also relates to an ex vivo method for preparing a population of induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention also relates to an ex vivo method for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention relates to an ex vivo method for preparing a population of induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
  • iPAM induced Paraxial Mesoderm progenitor
  • Wnt signaling pathway denotes a signaling pathway which may be divided in two pathways: the “canonical Wnt/beta catenin signaling pathway” and the “Wnt/PCP signaling pathway”.
  • the term “canonical Wnt/beta catenin signaling pathway” or “Wnt/PCP signaling pathway” in its general meaning denotes a network of proteins and other bioactive molecules (lipids, ions, sugars%) best known for their roles in embryogenesis and cancer, but also involved in normal physiological processes in adult animals.
  • the "canonical Wnt/beta catenin signaling pathway” is characterized by a Wnt dependant inhibition of glycogen synthase kinase 3B (GSK-3B), leading to a subsequent stabilization of B-catenin, which then translocates to the nucleus to act as a transcription factor.
  • the "Wnt/PCP signaling pathway” does not involve GSK-3 B or B-catenin, and comprises several signaling branches including Calcium dependant signaling, Planar Cell Polarity (PCP) molecules, small GTPases and C-Jun N-terminal kinases (JNK) signaling. These pathways are well described in numerous reviews such as (Clevers, 2006; Montcouquiol et al, 2006; Schlessinger et al, 2009).
  • the Wnt signaling pathway is the canonical Wnt/B-catenin signaling pathway.
  • the Wnt signaling pathway is the Wnt/PCP signaling pathway.
  • the Wnt signaling pathway is the canonical Wnt/ B- catenin signaling pathway and Wnt/PCP signaling pathway.
  • activator denotes a substance that enhances Wnt signaling activity.
  • this activity can be measured by Wnt reporter activity using established multimers of LEF/TCF binding sites reporters, and/or inhibition of GSK-3B, and/or activation of canonical Wnt target genes such as T, Tbx6, Msgnl, or Axin2.
  • iPAM induced Paraxial Mesoderm progenitor cells
  • the iPAM cells are characterized by the following properties:
  • Msgnl gene as measured for example with a gene reporter assay comprising the Msgnl promoter, and;
  • they are multipotent cells, capable of differentiating into at least skeletal, dermis or muscle cell lineages;
  • iPAM induced Paraxial Mesoderm progenitor
  • multipotent refers to cells that can differentiate in more than one cell lineage depending on the environmental and culture conditions. Contrary to embryonic stem cells which are pluripotent and can differentiate into all types of somatic cell lineages, the induced paraxial mesoderm progenitor cells of the present invention have limited differentiation capacity.
  • pluripotent cells refers to mammalian undifferentiated cells which can give rise to a variety of different cell lineages. Typically, pluripotent cells may express the following markers Oct4, SOX2, Nanog, SSEA 3 and 4, TRA 1/81, see International Stem Cell Initiative recommendations, 2007.
  • the pluripotent cells are human pluripotent cells.
  • the pluripotent cells are non-human mammalian pluripotent cells.
  • the pluripotent cells are stem cells.
  • said stem cells are embryonic stem cells.
  • the pluripotent cells are human embryonic stem cells (hES cells).
  • hES cells human embryonic stem cells
  • hES cell lines Loser et al, 2010
  • hES cell lines such as the one described in the following table may be employed for the method of the invention: - - passage country of
  • the pluripotent cells are non-human embryonic stem cells, such as mouse stem cells, rodent stem cells or primate stem cells.
  • the pluripotent cells are induced pluripotent stem cells (iPS).
  • iPS induced pluripotent stem cells
  • iPS cells Induced pluripotent stem cells
  • iPS cells are a type of pluripotent stem cells artificially derived from a non-pluripotent, typically an adult somatic cell, by inducing a "forced" expression of certain genes.
  • iPS cells were first produced in 2006 from mouse cells (Takahashi and Yamanaka, 2006) and in 2007 from human cells (Takahashi et al., 2007; Yu et al., 2007).
  • the activator of the canonical Wnt/B-catenin signaling pathway or the Wnt/PCP signaling pathway according to the invention is a member of the R- spondin family, originating from a vertebrate species or modified.
  • the member of the R-spondin family is a member of the mammalian R-spondin family. - -
  • the member of the R-spondin family according to the invention is selected in the group consisting of R-spondin 1, R-spondin 2, R-spondin 3 and R- spondin 4.
  • the member of the R-spondin family according to the invention is R-spondin 3.
  • the member of the R-spondin family according to the invention is R-spondin 2.
  • R-spondin3 or “R-spondin2” refers to members of the family of secreted proteins in vertebrates that activate the Wnt signaling pathway.
  • An exemplary sequence for human R-spondin3 protein is deposited in the database under accession number NP l 16173.2 (SEQ ID NO: l).
  • An exemplary sequence for mouse R- spondin3 protein is deposited in the database under accession number NP 082627.3 (SEQ ID NO:2).
  • An exemplary sequence for human R-spondin2 protein is deposited in the database under accession number NP 848660.3 (SEQ ID NO:3).
  • An exemplary sequence for mouse R- spondin2 protein is deposited in the database under accession number NP 766403.1 (SEQ ID NO:4).
  • R-spondin3 also encompasses any functional variants of R- spondin3 wild type (naturally occurring) protein, provided that such functional variants retain the advantageous properties of differentiating factor for the purpose of the present invention.
  • said functional variants are functional homologues of R-spondin3 having at least 60%, 80%, 90% or at least 95% identity to the most closely related known natural R- spondin3 polypeptide sequence, for example, to human or mouse polypeptide R-spondin3 of SEQ ID NO: l or SEQ ID NO:2 respectively, and retaining substantially the same Wnt activation activity as the related wild type protein.
  • said functional variants are fragments of R-spondin3, for example, comprising at least 50, 100, or 200 consecutive amino acids of a wild type R-spondin3 protein, and retaining substantially the same Wnt activation activity.
  • such functional variant can consist in R-spondin3 gene product isoforms such as the isoform 2 of the human R-spondin3 as described under the ref. Q9BXY4-2 and CAI20142.1 (SEQ ID NO:5).
  • R-spondin2 also encompasses any functional variants of R- spondin2 wild type (naturally occurring) protein, provided that such functional variants retain the advantageous properties of differentiating factor for the purpose of the present invention. - -
  • said functional variants are functional homologues of R-spondin2 having at least 60%, 80%, 90% or at least 95% identity to the most closely related known natural R- spondin2 polypeptide sequence, for example, to human or mouse polypeptide R-spondin2 of SEQ ID NO:3 or SEQ ID NO:4 respectively, and retaining substantially the same Wnt activation activity as the related wild type protein.
  • said functional variants are fragments of R-spondin2, for example, comprising at least 50, 100, or 200 consecutive amino acids of a wild type R-spondin2 protein, and retaining substantially the same Wnt activation activity.
  • said functional variants can consist in R-spondin2 gene product isoforms such as the isoform 2 or the isoform 3 of the human R- spondin2 such as described respectively under the ref. Q6UXX9-2 (SEQ ID NO: 6) or under the ref. Q6UXX9-3 (SEQ ID NO:7).
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
  • the percent identity between two amino-acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4: 11-17, 1988) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
  • the activator according to the invention is a combination of the R-spondin 3 and R-spondin 2.
  • the activator according to the invention may be the human R- spondin-3 isoform 2 of sequence SEQ ID NO:5.
  • the activator according to the invention may be the human R- spondin-2 isoform 2 of sequence SEQ ID NO:6, or the human R-spondin-2 isoform 3 of sequence SEQ ID NO:7.
  • the concentration of R-spondin3 used for culture of pluripotent cells is between 0.1 ng/ml and 500 ng/ml, preferably between 1 ng/ml and 500 ng/ml and more preferably between 5 ng/ml and 30 ng/ml.
  • the concentration of R-spondin2 used for culture of pluripotent cells is between 1 ng/ml and 500 ng/ml, preferably between 5 ng/ml and 30 ng/ml.
  • the concentration of R-spondin3 or R-spondin2 is 10 ng/ml. With a concentration of 10 ng/ml, more than 50% up to 70% of pluripotent cells are differentiated in induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • pluripotent cells are cultured with R-spondin3 or R-spondin2 during 1 to 15 days, or for a shorter time period.
  • pluripotent cells are cultured with R-spondin3 or/and R-spondin2 during at least 10 days at a concentration of 10 ng/ml.
  • the term '"inhibitor of the BMP signaling pathway denotes any compound, natural or synthetic, which results in a decreased activation of the BMP (bone morphogenetic protein) signaling pathway, which is characterized by the binding of a dimer BMP proteins to an heterocomplex constituted of BMP type I and type II receptors, which results in a phosphorylation cascade leading to the phosphorylation of Smadl/5/8, and resulting in target genes activation, such as Id genes.
  • an inhibitor of the BMP signaling pathway provokes a decrease in the levels of phosphorylation of the proteins Smad 1, 5 and 8 (Gazzero and Minetti, 2007).
  • a compound is deemed to be an inhibitor of the BMP signaling pathway if, after culturing cells in the presence of said compound, the level of phosphorylated Smad 1, 5 or 8 is decreased compared to cells cultured in the absence of said compound.
  • Levels of phosphorylated Smad proteins can be measured by Western blot using antibodies specific for the phosphorylated form of said Smad proteins.
  • Target genes activation can typically be measured by direct Idl/2/3 transcripts (mRNA) production, via quantitative real-time PCR (qRT-PCR) and expression levels can be compared to control situation, in the absence of said compound.
  • the inhibitor of the BMP signaling pathway may be a BMP antagonist, a chemical compound that blocks BMP type I and/or type II receptors activity (BMP type I/II receptor inhibitor), an inhibitor of BMP type I and/or type II gene expression, or a molecule which inhibits any downstream step of the BMP signaling pathway.
  • the inhibitor of BMP signaling - - may be a natural or a synthetic compound.
  • the inhibitor of the BMP signaling pathway is a protein, it may be a purified protein or a recombinant protein or a synthetic protein.
  • the inhibitor of the BMP signaling pathway is a BMP type I receptors inhibitor.
  • the inhibitor of the BMP signaling pathway is selected from the group consisting of Noggin, Chordin and related proteins (Chordin-like 1/2/3), Follistatin and related proteins (Follistatin-like 1/2/3/4/5), proteins of the Dan family (including Cerberus 1, Gremlin 1 and 2, Cerl-2 (Coco), SOST (Sclerostin), SOSTDC1 (Wise)) and variants and fragments thereof which inhibit the BMP signaling pathway.
  • the inhibitor of the BMP signaling pathway is selected from the group consisting of BMP- 1/Tolloid- like proteins, TWSGl (twisted gastrulation), TMEFFs (Tomoregulins), Biglycan, TSK (Tsukushi), BMPER (Crossveinless 2), Ogon (Sizzled), AMN (Amnionless), CTGF (Connective Tissue Growth Factor), and HSPGs (including Glypican3 and Syndecan4).
  • BMP- 1/Tolloid- like proteins TWSGl (twisted gastrulation), TMEFFs (Tomoregulins), Biglycan, TSK (Tsukushi), BMPER (Crossveinless 2), Ogon (Sizzled), AMN (Amnionless), CTGF (Connective Tissue Growth Factor), and HSPGs (including Glypican3 and Syndecan4).
  • the inhibitor of the BMP signaling pathway is noggin.
  • Noggin can be murine (mouse noggin exemplified by GenPept accession number NP 032737, SEQ ID NO: 10) or human noggin (human noggin exemplified by GenPept accession number EAW94528, SEQ ID NO: l 1). It may be purified or recombinant. It may be in monomeric or dimeric form.
  • the inhibitor of the BMP signaling pathway is a compound that inhibits BMP signaling transduction cascade.
  • the compound that inhibits BMP signaling transduction cascade is a synthetic or a chemical compound.
  • the inhibitor of the BMP signaling pathway is an inhibitor of
  • BMP type I receptors for "Bone Morphogenetic Protein” denotes transmembrane proteins with serine/threonine protein kinase activity that mediates the addition of phosphate molecules on certain serine and threonine amino acids on particular - - cellular substrates. It is well known in the art that an inhibitor of BMP type I receptors may block the BMP signaling pathway, see for example Yu et al, Nat Chem Bio 1.2008.
  • the inhibitor of BMP type I receptors is Dorsomorphin, a chemical compound or any derivatives generated by structure-activity studies [Cuny GD et al, 2008].
  • Dorsomorphin (6-[4-(2-Piperidin-l-yl-ethoxy)phenyl]-3-pyridin-4-yl-pyrazolo[l,5- ajpyrimidine , also known as Compound C) is inhibiting specifically BMP type I receptors (ALK2, 3, and 6) [Yu PB et al, 2008].
  • Recombinant Noggin can be purchased from R&D Systems or Peprotech or can be produced using standard techniques as described above.
  • the inhibitor of the BMP signaling pathway is added to the culture medium of the invention in a concentration ranging from 1 to 10000 ng/ml, preferably from 5 to 1000 ng/ml, preferably from 5 to 500 ng/ml, preferably from 10 to 200 ng/ml, even more preferably at about 200 ng/ml.
  • noggin is added to the culture medium of the invention at a concentration ranging from 1 to 1000 ng/ml, preferably from 10 to 200 ng/ml, even more preferably at about 200 ng/ml.
  • Dorsomorphin is added to the culture medium of the invention in a concentration ranging from 0.1 to 2 ⁇ , preferably at 1 ⁇ .
  • pluripotent cells are cultured with the inhibitor of the BMP signaling pathway during 1 to 4 days.
  • the culture medium according to the invention comprises a Wnt activator and an inhibitor of BMP signalling pathway according to the invention to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the Wnt activator is R-spondin3 and the inhibitor of BMP signalling pathway is Noggin.
  • the culture medium according to the invention may further comprise DMSO (Dimethyl sulfoxide) or an equivalent of the DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • DMSO Dimethyl sulfoxide
  • iPAM Paraxial Mesoderm progenitor
  • the term "equivalent” means a substance exhibiting the same properties as DMSO which is a solvent that dissolves both polar and nonpolar compounds. - -
  • the culture medium according to the invention comprises R- spondin 3, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • the culture medium according to the invention comprises R- spondin 3 and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R- spondin 2, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R- spondin 2 and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R- spondin 3, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R- spondin 2, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R-spondin 3, R- spondin 2, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R-spondin 3, R-spondin 2 and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R-spondin 3, R-spondin 2, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • Vertebrate recombinant R-spondins can be purchased commercially, or produced as conditioned culture medium. This involves expressing a construct containing the coding sequence of an R-spondin protein into competent cells, such as COS cells. R-spondin protein is secreted in the culture medium. Conditioned medium can be applied directly to pluripotent cells or prediluted in basal medium. - -
  • the activator of the Wnt signaling pathway is an inhibitor of GSK-3 ⁇ .
  • GSK-3 ⁇ for "Glycogen synthase kinase 3 beta” denotes a serine/threonine protein kinase that mediates the addition of phosphate molecules on certain serine and threonine amino acids on particular cellular substrates. It is well known in the art that an inhibitor of GSK-3 ⁇ may activate the Wnt signaling pathway, see for example (Cohen and Goedert, 2004; Sato et al, 2004; Taelman et al, 2010; Wu and Pan, 2010).
  • the inhibitor of GSK-3 ⁇ is CHIR99021.
  • R-spondin factor 2. allowing ectopic expression of said R-spondin factor by introducing an expression vector comprising a coding sequence of R-spondin factor operably linked to control sequences into the pluripotent cells to be differentiated, or by introducing in the cells coding RNA for R-spondin factor
  • R-spondin factor for example as recombinant R-spondin factor (family of R-spondinl, 2 ,3 and 4) in the culture medium, or conditioned medium, or as substrate coating.
  • the culture medium according to the invention comprises CHIR99021 and an inhibitor of BMP signaling pathway according to the invention which is Dorsomorphin to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises CHIR99021, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises a Wnt activator which is a combination of R-spondin2, R-spondin3 and CHIR99021 ; and an - - inhibitor of BMP signaling according to the invention which is a combination of Noggin and Dorsomorphin to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • Wnt activator which is a combination of R-spondin2, R-spondin3 and CHIR99021
  • an - - inhibitor of BMP signaling according to the invention which is a combination of Noggin and Dorsomorphin to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R-spondin 3, R-spondin 2, CHIR99021 , Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the culture medium according to the invention comprises R-spondin 3, R-spondin 2, CHIR99021, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • introducing directly into the cells environment an appropriate amount of pharmacological GSK-3P inhibitor for example the chemical compound CHIR99021 is used as an alternative for increasing the activity of Wnt signaling pathway in the system, alone or in combination with R-spondin.
  • the invention relates to a composition for preparing induced Paraxial Mesoderm progenitor (iPAM) cells from pluripotent cells wherein said composition comprises an effective amount of an activator of the Wnt signaling pathway according to the invention and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention also relates to a composition for preparing induced Paraxial Mesoderm progenitor (iPAM) cells from pluripotent cells wherein said composition comprises an effective amount of an activator of the Wnt signaling pathway according to the invention.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention also relates to a kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said kit comprising:
  • BMP Bone Morphogenetic Protein
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention also relates to a kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said kit comprising:
  • iPAM induced Paraxial Mesoderm progenitor
  • the activator is a member of the R-spondin family.
  • the activator is selected from the group consisting of R- spondin 1, R-spondin 2, R-spondin 3 and R-spondin 4.
  • the activator is the R-spondin 2 or the R-spondin 3.
  • the activator is an inhibitor of GSK-3P such as CHIR99021.
  • the inhibitor according to the invention is a secreted antagonist of the BMP/TGFbeta family.
  • the inhibitor of BMP signaling pathway is selected from the group consisting of Noggin, Chordin, Chordin-like 1/2/3, Follistatin, Follistatin-like 1/2/3/4/5, a member of the Dan family, including Cerberus 1, Gremlin 1/2.
  • the inhibitor is Noggin or Follistatin.
  • the inhibitor is a chemical inhibitor of BMP signaling such as Dorsomorphin.
  • said kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells comprises,
  • composition comprising members of the R-spondin family
  • composition comprising an inhibitor of the BMP signaling pathway and c) DMSO or an equivalent.
  • said kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells comprises,
  • composition comprising members of the R-spondin family
  • said kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells comprises, - - a) a composition comprising a chemical compound inhibitor of GSK-3P; b) a composition comprising a chemical compound inhibitor of BMP type I receptors and
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention further relates to populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells obtainable from the method as described above.
  • populations typically may comprise other cell types in addition to induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the populations of the invention are characterized in that they comprise at least 10%, 20%>, 30%>, 40%>, 50%>, 60%>, 70%, 80% and preferably at least 90% of cells that exhibit high expression of at least one biomarker characteristic of paraxial mesoderm progenitor cells, for example Msgnl gene product.
  • biomarkers characteristic of paraxial mesoderm progenitor cells include, without limitation, one or more of the following proteins: Tbx6, EphrinAl, EphrinB2, EPHA4, PDGFRalpha, Salll, Sall4, Dill, D113, Papc (Pcdh8), Lfng, Hes7, Ripplyl, Ripply2, Brachyury (T), Cdx2, Cdx4, Evxl, Cxcr4, I117rd, Fgf8, Fgfl7, Gbx2, Wnt3a, Wnt5b, Rspo3, SP5, SP8, Has2, Dkkl, Dactl, Pax3, Pax7, Mespl, Mesp2.
  • any methods known in the art for measuring gene expression may be used, in particular, quantitative methods such as, real time quantitative PCR or microarrays, or methods using gene reporter expression, said gene reporter comprising Msgnl promoter as described in the Examples, or qualitative methods such as immuno staining or cell sorting methods identifying cells exhibiting specific biomarkers, including cell surface markers.
  • the Msgnl gene refers to the gene encoding Mesogeninl .
  • Examples of a nucleotide sequence of a gene encoding Mesogeninl in mouse and human are given in SEQ ID NO:8 (NM 019544.1) and SEQ ID NO:9 (NM 001105569.1) respectively.
  • SEQ ID NO:8 NM 019544.1
  • SEQ ID NO:9 NM 001105569.1
  • expression of Msgnl is considered high if expression is detectable in a quantitative assay for gene expression. In another embodiment, it is high if the expression level is significantly higher than the expression level observed in the original pluripotent cells, or in cells differentiating under non specific conditions such as Basal culture medium without LIF (Leukemia Inhibitory Factor) for mouse pluripotent cells or without FGF (Fibroblast Growth Factor) for human pluripotent cells. Expression levels between the control and the test cells may be normalized using constitutively expressed genes such as GAPDH or Beta Actin. Populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells may be cultured indefinitely under appropriate growth conditions.
  • iPAM induced Paraxial Mesoderm progenitor
  • growth conditions may be established by the skilled person in the art based on established growth conditions for embryonic stem cells or induced pluripotent stem cells (iPS cells) for example or as described in the Examples below. Growth conditions may advantageously comprise for example the use of serum replacement medium, KSR (Gibco), ESGRO (Chemicon/Millipore) supplemented with growth factors like FGFs, WNTs, or chemical compounds modulating the respective signaling pathways.
  • KSR Gibco
  • ESGRO Cemicon/Millipore
  • the induced Paraxial Mesoderm progenitor (iPAM) cells may be purified or the populations may be enriched in induced Paraxial Mesoderm progenitor (iPAM) cells by selecting cells expressing markers specific of induced Paraxial Mesoderm progenitor (iPAM) cells.
  • markers specific of induced Paraxial Mesoderm progenitor (iPAM) cells for purification or enrichment of a population of induced Paraxial Mesoderm progenitor (iPAM) cells may be selected among one or more of the following markers: Msgnl, Tbx6, EphrinAl , EphrinB2, EPHA4, PDGFRalpha, Salll , Sall4, Dill , D113, Papc (Pcdh8), Lfng, Hes7, Ripply 1, Ripply2, Brachyury (T), Cdx2, Cdx4, Evxl, Cxcr4, I117rd, Fgf8, Fgfl7, Gbx2, Wnt3a, Wnt5b, Rspo3, SP5, SP8, Has2, Dkkl, Dactl, Pax3, Pax7, Mespl, Mesp2, or selected negatively with markers of other lineages/cell type such as neural fate.
  • Purification or induced Paraxial Mesoderm progenitor (iPAM) cells enrichment may be achieved using cell sorting technologies, such as fluorescence activated cell sorting (FACS) or magnetic beads comprising specific binders of said cell surface markers of induced Paraxial Mesoderm progenitor (iPAM) cells, or fluorescent reporters for paraxial mesoderm progenitor markers.
  • FACS fluorescence activated cell sorting
  • iPAM induced Paraxial Mesoderm progenitor
  • Another method consists in taking advantage of the differential adhesion - - properties of induced Paraxial Mesoderm progenitor (iPAM) cells, by selective attachment on defined substrates.
  • the population may thus comprise more than 10%, 20%, 30%, 40%, 50%, 60%; 70%, 80%, 90% or more than 95% of cells having a high expression of a biomarker characteristic of induced Paraxial Mesoderm progenitor (iPAM) cells, for example, Msgnl gene product.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention relates to a composition
  • a composition comprising a population of induced Paraxial Mesoderm progenitor (iPAM) cells obtainable from the method as described above.
  • iPAM induced Paraxial Mesoderm progenitor
  • iPAM Mesoderm progenitor
  • the induced Paraxial Mesoderm progenitor (iPAM) cells may advantageously be cultured in vitro under differentiation conditions to generate skeletal muscle, bone, cartilage, dermal cells, as well as other derivatives of the paraxial mesoderm including but not restricted to adipocytes or endothelial cells.
  • the invention relates to a method for preparing populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages said method comprising the steps of
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention further relates to a composition for preparing populations of cell lineages comprising induced Paraxial Mesoderm progenitor (iPAM) cells according to the invention and appropriate conditions for their differentiation into the desired cell lineages.
  • iPAM induced Paraxial Mesoderm progenitor
  • the present invention provides a method for preparing a population comprising skeletal muscle cell lineages, said method comprising the steps of
  • iPAM induced Paraxial Mesoderm progenitor
  • step (c) optionally, culturing said population obtained from step (b) in a second differentiation medium comprising at least one or more compounds activating or inhibiting the Wnt, FGF, HGF (Hepatocyte growth factor), Activin, EGF (Epidermal growth factor), insulin, and IGF signaling pathways or compounds known to promote myogenic differentiation such as horse serum or transferrin,
  • a second differentiation medium comprising at least one or more compounds activating or inhibiting the Wnt, FGF, HGF (Hepatocyte growth factor), Activin, EGF (Epidermal growth factor), insulin, and IGF signaling pathways or compounds known to promote myogenic differentiation such as horse serum or transferrin,
  • skeletal muscle cell lineages that can be identified by markers such as Desmin, or Myosin Heavy Chain.
  • the extracellular matrix material is selected from the group consisting of Collagen I, Collagen IV, Fibronectin, Laminin, gelatin, poly-lysine, PDMS and Matrigel.
  • the invention further relates to a composition for preparing skeletal muscle cell lineages from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises:
  • composition further comprises at - - least another compound activating or inhibiting the Wnt, FGF, HGF, Activin, EGF, insulin, and IGF signaling pathways or compounds known to promote myogenic differentiation such as horse serum or transferrin.
  • the present invention provides a method for preparing a population comprising dermal cell lineages, said method comprising the steps of culturing a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of an efficient amount of at least one or more compounds activating or inhibiting BMP, TGFB, Wnt, FGF, EGF, retinoic acid, Notch and Hedgehog pathways.
  • Dermal cells can be identified using markers such as Dermo- 1.
  • the invention further relates to a composition for preparing dermal cell lineages from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one or more compounds activating or inhibiting BMP, TGFB, Wnt, FGF, EGF, retinoic acid, Notch and Hedgehog pathways.
  • iPAM induced Paraxial Mesoderm progenitor
  • the present invention provides a method for preparing a population comprising bone or cartilage cell lineages, comprising the step of culturing a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of an efficient amount of at least one or more compounds activating or inhibiting the retinoic acid, Wnt, Hedgehog, PTHrP, TGFB, BMP pathways, or compounds known to promote bone or cartilage differentiation such as dexamethasone, ascorbic acid, vitamin D3, and beta- glycerophosphate.
  • Cartilage cells can be identified by classical staining such as Alcian Blue and bone cells with alizarin red or Von Kossa stain.
  • the invention further relates to a composition for preparing bone or cartilage cell lineages from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one or more compounds activating or inhibiting retinoic acid, Wnt, Hedgehog, PTHrP, TGFB, BMP pathways, or compounds known to promote bone or cartilage differentiation such as dexamethasone, ascorbic acid, vitamin D3 and beta-glycerophosphate.
  • iPAM induced Paraxial Mesoderm progenitor
  • the present invention provides a method for preparing a population comprising adipocytes, said method comprising the steps of culturing the population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of - - an efficient amount of at least one or more compounds known to promote adipocyte differentiation including dexamethasone, isobutylxanthine and insulin.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention further relates to a composition for preparing adipocytes from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one compound known to promote adipocyte differentiation including dexamethasone, isobutylxanthine and insulin.
  • iPAM induced Paraxial Mesoderm progenitor
  • the present invention provides a method for preparing a population comprising endothelial cells, said method comprising the steps of culturing the population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of an efficient amount of at least one or more compounds activating or inhibiting the VEGF or FGF pathways. Endothelium can be detected by PECAM-1 (CD31) immuno staining.
  • the invention further relates to a composition for preparing endothelial cells from induced
  • Paraxial Mesoderm progenitor (iPAM) cells characterized in that it further comprises at least one or more compounds activating or inhibiting the VEGF or FGF pathways.
  • iPAM induced Paraxial Mesoderm progenitor
  • the present invention provides populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages as well as other derivatives derived from induced Paraxial Mesoderm progenitor (iPAM) cells.
  • populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages as well as other derivatives derived from induced Paraxial Mesoderm progenitor (iPAM) cells.
  • iPAM induced Paraxial Mesoderm progenitor
  • the present invention provides populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages as well as other derivatives derived from induced Paraxial Mesoderm progenitor (iPAM) cells obtained with a composition according to the invention.
  • populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages as well as other derivatives derived from induced Paraxial Mesoderm progenitor (iPAM) cells obtained with a composition according to the invention.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention relates to a composition comprising skeletal muscle, bone, cartilage, dermal cell, adipocyte or endothelial cell lineages obtainable by a method according to the invention.
  • - - iPAM cells population of cells derived from iPAM cells and uses thereof
  • Another aspect of the invention relates to the use of said populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells, or said populations comprising skeletal muscle, bone, cartilage or dermal cell lineages derived from differentiation of induced Paraxial Mesoderm progenitor (iPAM) cells, but also adipose tissue and endothelial paraxial mesoderm derivatives, hereafter referred as the Populations of the Invention.
  • iPAM induced Paraxial Mesoderm progenitor
  • the Populations of the Invention may be used in a variety of applications, in particular, in research or therapeutic field.
  • cells obtained from a patient suffering from a genetic defect may be cultured and genetically corrected according to methods known in the art, and subsequently reprogrammed into iPS cells and differentiated into induced Paraxial Mesoderm progenitor (iPAM) cells or its derivatives for re-administration into the patient.
  • iPS induced Paraxial Mesoderm progenitor
  • regenerative medicine can be used to potentially cure any disease that results from malfunctioning, damaged or failing tissue by either regenerating the damaged tissues in vivo by direct in vivo implantation of a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells or their derivatives comprising appropriate progenitors or cell lineages.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention relates to the induced Paraxial Mesoderm progenitor (iPAM) cells or their derivatives or the Populations of the Invention for use as a cell therapy product for implanting into a mammal, for example human patient.
  • iPAM induced Paraxial Mesoderm progenitor
  • the invention relates to a pharmaceutical composition comprising a population of induced Paraxial Mesoderm progenitor (iPAM) cells obtained according to the invention.
  • the invention relates to a pharmaceutical composition comprising a population of induced Paraxial Mesoderm progenitor (iPAM) cells including for example at least 10 2 , 10 3 , 10 4 , 10 5 , 10 6 , 10 7 , 10 8 , or at least 10 9 Msgnl expressing cells.
  • this composition comprises a pharmaceutically acceptable vehicle.
  • the Populations of the Invention are used for the treatment of a muscle genetic disorder, for example Duchenne muscular dystrophy, or any other genetic muscular dystrophy.
  • induced Paraxial Mesoderm progenitor (iPAM) cells are co- cultured with various cell types to induce their differentiation toward the desired lineage.
  • induced Paraxial Mesoderm progenitor (iPAM) cells are directly grafted into a recipient host.
  • iPAM induced Paraxial Mesoderm progenitor cells can be grafted after genetic correction by methods known in the art.
  • the Populations of the Invention are used in orthopaedic surgery for the treatment of joint or cartilage or bone damages caused by aging, disease, or by physical stress such as occurs through injury or repetitive strain.
  • the Populations of the Invention may also be used advantageously for the production of dermal tissues, for example, skin tissues, for use in regenerative medicine or in research, in particular in the cosmetic industry or for treatment of burns and plastic surgery.
  • the invention in another preferred embodiment, relates to a composition comprising the Populations of the Invention.
  • the composition comprising the Population of the invention may be used in cell therapy or regenerative medicine.
  • FIGURES and TABLES are identical to FIGURES and TABLES:
  • hRspondin NP-1 16173.2 SEQ ID MHLRLISWLF IILNFMEYIG SQNASRGRRQ
  • hRspondin CAI20142.1 SEQ ID MHLRLISWLF IILNFMEYIG SQNASRGRRQ
  • FIG. 1 R-spondin induces induced Paraxial Mesoderm progenitor (iPAM) cells fate.
  • iPAM Paraxial Mesoderm progenitor
  • A Comparison of fluorescent Msgnl Reporter activation (YFP positive cells (YFP+ cells)) after 4 days of differentiation of mES cells (Msgnl Rep V), under default culture conditions in 15% FBS or 15% KSR medium, with or without recombinant mouse Rspo3 (lOng/mL). YFP channel, 50X.
  • B Robustness of iPAM cells induction in response to mouse Rspo3 in 15% FBS medium. Triplicate wells measurements by flow cytometry. Error bar is s.e.m.
  • FIG. 1 Flow-cytometry analysis of the induction of the Msgnl-YFP+ (induced Paraxial Mesoderm progenitor (iPAM) cells) population upon treatment with Rspo3.
  • Msgnl-YFP+ induced Paraxial Mesoderm progenitor (iPAM) cells
  • (A) Flow-cytometry analysis on the MsgnlRepV mES cells at dayO of differentiation in 15% FBS medium. YFP+ population represents less than 1%.
  • (B) Flow-cytometry analysis at day 4 of differentiation in 15% FBS medium supplemented with R-spondin3 lOng/mL. YFP+ population represents more than 70% of the total population.
  • Figure 3 Paraxial mesoderm progenitors (induced Paraxial Mesoderm progenitor (iPAM) cells) characterization.
  • FIG. 4 R-spondin activity in induced Paraxial Mesoderm progenitor (iPAM) cells mediated by canonical Wnt signaling. - -
  • FIG. 5 R-spondin activity can be mimicked by the GSK3beta inhibitor CHIR99021.
  • DMSO has a positive effect on induced Paraxial Mesoderm progenitor (iPAM) cells induction.
  • iPAM Paraxial Mesoderm progenitor
  • iPAM induction after 4 days of differentiation in 15% FBS medium containing 0.5% DMSO.
  • Optimal iPAM induction is obtained by combining R-spondins and DMSO. Concentrations in ng/mL.
  • Figure 7 Rspo2 and 3 activity in defined medium.
  • Figure 8 Characterization of Populations of the Invention at day 18 of differentiation.
  • mice ES cells were differentiated in presence of Rspo 3, followed by 15% FBS medium until day 18.
  • Cell types were identified by tissue-specific antibody staining, namely Muscle (Desmin, green), Endothelium (PECAM1/CD31, green) and Cartilage (Alcian Blue).
  • Figure 9 Dermal and myogenic differentiation of the iPAM cells after 5 days of culture.
  • ES cells were cultured for 4 days in FBS 15%, DMSO 0.5% and 10 ng/ml Rspo3 and then switched to FBS 15% or FBS 1% or FBS1% plus Sonic Hedgehog (Shh) and Retinoic acid (FlShhRA) or plus Shh, Noggin and LiCl (FISNLi). Cells were harvested the next day and analyzed by qRT-PCR for the dermal marker Dermo 1 (A) and the muscle marker Myf5 (B). Graphs show fold enrichment.
  • Figure 10 R-spondin induces iPAM fate in human ES cells.
  • Figure 11 Noggin promotes iPAM fate by counteracting BMP4 activity.
  • Figure 12 Molecular characterization of early stages of paraxial mesoderm differentiation.
  • FIG. 1 Venn diagram comparing gene signature lists (GSL method) of posterior and anterior PSM domains and of the in vitro differentiated MsgnlRepV Venus positive ES cells harvested at day3 and 4, respectively, in presence of of 10 ng/ml Rspo3, DMSO 0.5%> and 200ng/ml Noggin.
  • Key signature genes shared between the PSM in vivo and in vitro the Venus positive ES cells are highlighted (black boxes). Red arrow shows the gene signature shift between day3 and day4 of ES cells differentiation.
  • iPAM induced Paraxial Mesoderm progenitor
  • FIG. 13 Pax3-positive PSM progenitors differentiated from ES cells in vitro can generate muscle fibers in vivo.
  • Tibialis anterior muscles were collected after 1 month post-transplantation with in vitro differentiated Pax3-positive cells (induced Paraxial Mesoderm progenitor (iPAM) cells) labeled with a CAG-GFP lentivirus (Transverse section).
  • Control Crtl, non-grafted area
  • engrafted area were stained (red) with antibodies against Dystrophin (DYS) and Laminin (LAMA1).
  • Engrafted progenitors produce large areas of muscle fibers expressing dystrophin and Laminin. Nuclei are counterstained with Hoechst. Scale bar, ⁇ .
  • Engrafted cells express the embryonic myosin MyHCemb, MyHCI (slow) and MyHCperi /MyHC II (fast) (Left panel), and overlay with corresponding GFP/Hoechst conterstaining is shown in Right panel. For each antibody, grafted tissue is shown on top and control tissue is shown below. Scale bar, 200 ⁇ .
  • Undifferentiated mouse ES cells MsgnlRepV (El 4 derived) were maintained on gelatin-coated dishes in DMEM supplemented with 15% fetal bovine serum (FBS; from PAA), penicillin, streptomycin, 2mM L-Glutamine, O. lmM non essential amino acids, ImM sodium pyruvate, 0.2%> ⁇ -mercaptoethanol and 1 ,500 U/mL LIF.
  • ES cells were co-cultured with mytomicin- inactivated MEFs (feeders).
  • Undifferentiated human ES cells were cultured on plates coated with matrigel (BD Biosciences) in mTeSR medium (STEMCELL Technologies). Cultures were maintained in an incubator in 5% C02 at 37°C.
  • ES cells were trypsinized and plated at various densities in gelatin coated, feeder-free, 24 well plates directly in serum-based (15% FBS) or serum-replacement (15% KSR, Invitrogen) conditions supplemented with factors, and DMSO (Sigma).
  • Recombinant proteins were obtained commercially (R&D) and stock solutions were prepared according to manufacturer's recommendation.
  • the GSK-3B inhibitor CHIR99021 and the BMP type I receptors inhibitor Dorsomorphin were purchased from Stemgent and prepared according to the manufacturer's recommendations. Fluorescent reporter analysis and image acquisition were done on a Zeiss Axiovert system. FACS analysis and cell sorting
  • Mouse E9.5 embryo PSMs were microdissected and processed as previously described (Krol et al, 2011), and prepared samples were run on Affymetrix GeneChip arrays.
  • iPAMs were sorted by flow-cytometry based on their Msgnl- YFP+ expression, and samples were prepared for microarrays. Experiments were conducted on biological triplicates. Datasets were processed using Affymetrix Microarray Suite (MAS) 5.0.
  • MAS Affymetrix Microarray Suite
  • a gene expression reference was created by using all microarrays of wild-type mouse tissues deposited in GEO corresponding to Affymetrix Mouse Genome 430 2.0 Arrays (GEO platform id GPL1261). Normalization was done by calculating the mean values for each microarray. The median values for the distribution of those mean values across all microarrays were determined. This median is then used as a scaling factor for each value on each microarray. Once all microarrays have been normalized, the median expression value for each probeset is defined as the reference value. Gene signature list specific to one experimental condition was generated by normalizing the corresponding microarray data like the reference dataset. Probeset whose normalized expression value is 10 times higher than the corresponding reference value is considered to be a signature gene of that condition.
  • Total R A was extracted from ES cell cultures using Trizol (Invitrogen) or with the Rnaeasy plus mini-kit (Qiagen). RT-PCR was performed on 5ng total RNA using QuantiFast SYBR Green RT-PCR Kit (Qiagen), appropriate primers and run on a LightCycler 480II (Roche). GAPDH was used as the internal control.
  • MsgnlRepV ES cell cultures were trypsinized after 4 days of differentiation, and iPAMs were sorted based on YFP fluorescence using a FACS Aria, or Mof ow Astrios (BD).
  • iPAMS were permanently labeled by transduction overnight with a CAG-GFP lentivirus (MOI of 20-30).
  • MOI CAG-GFP lentivirus
  • Antibodies used in this study are anti-Dystrophin (Sigma), anti-Laminin (Sigma), Myogenin (Dako), Pax7 (DSHB) and GFP (Abeam).
  • Antibodies for Myosins isoforms have been described in (S.J.Mathew Dev 138, 371 (2011). Tissue sections were incubated overnight with primary antibodies. Secondary antibodies conjugated with AlexaFluor (Molecular probes) were used at 1 :500. Imaging was performed on a Zeiss Axio observer and images processed with Adobe photoshop.
  • Example 1 Use of an activator of the Wnt signaling pathway, - -
  • iPAM induced Paraxial mesoderm progenitor
  • transgenic mouse ES cell line harboring a fluorescent reporter specifically expressed in paraxial mesoderm progenitors.
  • Msgnl a gene specific for the presomitic mesoderm
  • the transgenic MsgnlRepV (Mesogeninl Reporter Venus) mouse ES cell line was subsequently validated using the tetraploid aggregation method to generate embryos entirely derived from the transgenic ES cells.
  • transgenic mouse embryos exhibit fluorescently labeled paraxial mesoderm tissue, thus, validating the tissue specificity of Venus expression in the transgenic ES cell line.
  • the MsgnlRepV reporter cells were plated at a defined density in 24-well plates coated with gelatin (0.1%>). Two basal culture media were selected: a DMEM based medium containing 15% fetal bovine serum (FBS, high serum) and a defined serum- free medium containing 15% KSR (Invitrogen/Gibco). These basal media were supplemented with candidate factors on day 0 of differentiation. Control and experimental conditions were cultured in parallel. Cells were left to differentiate for three to four days with medium changed on day 2 or 3. Cell cultures were analyzed on day 3 and 4 of differentiation visually and by flow cytometry for YFP+ population quantification. - -
  • YFP+ population While at day 0, the YFP+ population is ⁇ 1% of the cells, in R-spondin3 supplemented differentiation medium, YFP+ population can represent more than 50%, up to 70% of the cells at day4 ( Figure 2B).
  • induction of the paraxial mesoderm progenitor markers, Brachyury, PDGFRa, Tbx6 and Msgnl is observed after 3 to 10 days of culture in 15% FBS containing medium when treating huESl with R- spondin3 ( Figure 10). Paraxial mesoderm progenitors characterization.
  • ES cells were cultured in medium containing recombinant R-spondin proteins (R-spondins 1-4) supplemented with 15% FBS and allowed to differentiate for 4 days (Figure 4B).
  • R-spondins 1-4 recombinant R-spondin proteins
  • Figure 4B Two members of the family, R-spondin2 and R-spondin3, exhibit comparable activities and significantly increase the number of YFP+ cells.
  • Luciferase was strongly activated by R-spondin3 treatment suggesting that it activates the canonical Wnt pathway in this context.
  • DMSO Dimethyl sulfoxide
  • EC Embryonic Carcinoma
  • paraxial mesoderm progenitor cells are fated to become skeletal muscles, vertebral tissues (cartilage, bone), dorsal dermis, endothelium, and other tissues such as adipose tissues.
  • ES cells were exposed to optimized differentiation conditions (ie. R-spondin3 lOng/mL, DMSO 0.5%, in 15% FBS basal medium).
  • culture medium was changed and cells were exposed to specific differentiation media until day 18, with medium replacement every 3 days.
  • cell cultures were fixed and analyzed by tissue specific histochemical stainin g o r immunofluorescence ( Figure 8).
  • tissue specific histochemical stainin g o r immunofluorescence Figure 8
  • cell cultures were positive for Cartilage (Alcian blue positive nodules), Muscles (Desmin positive fibers) and Endothelium (CD31/PECAM1).
  • 4 days in differentiation conditions ie.
  • iPAM induced paraxial mesoderm progenitors
  • Induced paraxial mesoderm progenitors (iPAM) cells can be cultured in a hanging drop for 3 days at 800cells/20uL in differentiation medium, composed of DMEM supplemented with 10%> fetal calf serum (FCS), 5% horse serum (Sigma), 0.1 mM 2- mercaptoethanol, 0.1 mM nonessential aminoacids, and 50 ug/ml penicillin/streptomycin. After 3 days, the medium is changed and cell aggregates are transferred on a low attachement plate. At day 6, cells are plated and cultured in differentiation medium on plates coated with Matrigel (BD Bioscience, Bedford, MA, USA). Myogenic differentiation is achieved by withdrawal of FBS from confluent cells and addition of 10 ug/ml insulin, 5 ug/ml transferrin, and 2% horse serum.
  • FCS fetal calf serum
  • horse serum Sigma
  • 0.1 mM 2- mercaptoethanol 0.1 mM nonessential aminoacids
  • Induced paraxial mesoderm progenitors can also be cultured for 3 weeks in Skeletal Muscle Cell Medium (Lonza, Chemicon) complemented with EGF, insulin, Fetuin, dexamethasone, and bFGF (100 ng/mL).
  • Skeletal Muscle Cell Medium Longza, Chemicon
  • EGF EGF
  • insulin insulin
  • Fetuin Fetuin
  • dexamethasone bFGF (100 ng/mL).
  • Induced paraxial mesoderm progenitors iPAM cells are exposed to 200 ng/ml human or mouse recombinant BMP4 or a combination of 1 uM retinoic acid and 10 mM Lithium Chloride.
  • cells are plated on gelatin-coated plates at a density of 1-3 x 10 A 3 per well (24-well plate) and cultured for 28 days in bone differentiation medium (DMEM, 10%FBS, 2 mM 1-Glutamine, 1 * Penicillin/streptomycin (P/S), 0.1 ⁇ dexamethasone, 50 ⁇ ascorbic acid 2 -phosphate, 10 mM ⁇ -glycerophosphate, 10 ng/mL BMP4) in order to observe cells expressing bone specific markers or secreting alcian blue positive extracellular matrix.
  • Differentiated skeletal cell lineages are identified using specific stainings for extracellular matrix components of bone and cartilage including alcian blue or alizarin red, as well as by immunofluorescence using chondrocyte- and/ or osteocyte specific antibodies.
  • Induced paraxial mesoderm progenitors (iPAM) cells can also be differentiated into the bone lineage using the following differentiation medium composed of DMEM, 10% FBS, 2 mM L-Glutamine, l x P/S, 0.1 mM Dexamethasone, 50 mM ascorbic acid 2-phosphate,10 mM beta-glycerophosphate, and 10 ng/mL BMP4, and vitamin D3 for 20 days, with medium changed every 3 days. Bone formation can be confirmed by staining the differentiating culture with Alizarin red, well known in the art that results to stain differentiated bone in red. Extracellular accumulation of calcium can also be visualized by von Kossa staining.
  • differentiating cells can be lysed and assayed for ALP activity using BBTP reagent.
  • differentiating cells can be analyzed for osteoblast lineage markers expression, for example Osterix(Osx) and Cbfal/Runx2, alkaline phosphatase, collagen type I, osteocalcin, and osteopontin.
  • iPAM induced paraxial mesoderm progenitors
  • cartilage differentiation medium MEM, 10%FBS, 2 mM 1-Glutamine, l x P/S, 0.1 ⁇ Dexamethasone, 170 ⁇ ascorbic acid 2-phosphate.
  • cartilage differentiation medium MEM, 10%FBS, 2 mM 1-Glutamine, l x P/S, 0.1 ⁇ Dexamethasone, 170 ⁇ ascorbic acid 2-phosphate.
  • cartilage differentiation medium MEM, 10%FBS, 2 mM 1-Glutamine, l x P/S, 0.1 ⁇ Dexamethasone, 170 ⁇ ascorbic acid 2-phosphate.
  • cartilage differentiation medium 10 ng/mL TGF beta3
  • the medium is replaced with cartilage differentiation medium supplemented with - - lOng/mL Bmp2.
  • cartilaginous nodules secreting extracellular matrix can be observed.
  • Induced paraxial mesoderm progenitors (iPAM) cells can also be differentiated into cartilage cells using a differentiation medium based on alphaMEM, 10%FBS, 2 mM L- Glutamine, l x P/S, 0.1 mM Dexamethasone, and 170 mM ascorbic acid 2-phosphate or DMEM supplemented with 0.1 mM dexamethasone, 0.17 mM ascorbic acid, 1.0 mM sodium pyruvate, 0.35mM L-proline, 1% insulin-transferrin sodium, 1.25 mg/ml bovine serum albumin, 5.33 ug/ml linoleic acid, and 0.01 ug/ml transforming growth factor-beta), as well as TGFbeta3 or BMP2.
  • a differentiation medium based on alphaMEM, 10%FBS, 2 mM L- Glutamine, l x P/S, 0.1 mM Dexa
  • Cells are cultured for several weeks, with medium changed every 3 days. Differentiation can also be performed at high density on 3D scaffold such as Alginate beads in a DMEM based medium containing 10% FBS and antibiotic supplemented with 100 ng/ml recombinant human Bone Morphogenic Protein-2 (BMP-2) and 50 mg ascorbic acid. Cartilage formation can be confirmed by Alcian Blue staining of the differentiating culture, well known in the art that results in the staining of Muco-glycoproteins in blue. Alternatively, a safranin O staining can be performed.
  • Induced paraxial mesoderm progenitors can be differentiated into dermal fibroblasts by culturing them on a scaffold of collagen in medium containing a fibroblast growth factor such as bFGF (basic Fibroblast Growth Factor) or a member of the Wnt family of growth factors.
  • a fibroblast growth factor such as bFGF (basic Fibroblast Growth Factor) or a member of the Wnt family of growth factors.
  • iPAM paraxial mesoderm progenitors
  • Example 2 Use of an activator of the Wnt signaling pathway and an inhibitor of BMP signaling pathway,
  • Msgnl-YFP+ cells express a number of key paraxial mesoderm markers already validated by Q-PCR such as Msgnl and Tbx6 (Figure 3B).
  • Msgnl and Tbx6 the Msgnl -YFP+ population also activates cardiac and angiogenic markers, which are a signature of more ventral/lateral mesoderm derivatives.
  • TGFp/BMP signaling tends to be up-regulated in Msgnl -YFP+.
  • the Msgnl -YFP+ cell population expresses BMP4 at a significant level whereas BMP4 is detected only at very low level in vivo in the PSM. This was problematic because BMP4 has been shown to prevent cells to acquire a paraxial mesoderm fate at the expenses of lateral plate fates (Pourquie O et al, 1996, Tonegawa A et al, 1997).
  • Noggin Noggin
  • iPAM Paraxial Mesoderm progenitor
  • BMP inhibitors including recombinant proteins Noggin (Nog), Chordin (Chd), Chordin-like 1 (Chdll), Follistatin (Fst), Follistatin- like 1 (Fstll), Dan family protein including Cerberus 1 (Cerberus) and Gremlin 1 (Greml); at varying concentration range (10-200ng/mL), and various time-window (day 0-4, day 1-4) and analyzed the impact on BMP4 and the PSM marker Tbx6 expression. Additionally, we tested the chemical compound Dorsomorphin (Compound C), a specific BMP type I receptor inhibitor, at various concentration (0.1-1 ⁇ ) and various time-window (day 0-4, day 1-4).
  • iPAM induced paraxial mesoderm progenitors
  • Noggin Noggin
  • Follistatin Fst
  • Dorsomorphin promoting induced paraxial mesoderm progenitors maturation by counteracting BMP4 and activating Msgnl and Tbx6 expression (data not shown).
  • R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/ ⁇ beta ⁇ -catenin signaling. Proc Natl Acad Sci U S A 108, 11452-7.
  • Tbx6 a mouse T-Box gene implicated in paraxial mesoderm formation at gastrulation. Dev Biol 180, 534-42.
  • Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature.
  • R-spondin2 regulates craniofacial patterning and morphogenesis within the branchial arch through ectodermal-mesenchymal interaction. Dev Biol 352, 1-13.
  • R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling and is required for Xenopus myogenesis. Dev Cell 7, 525-34.
  • Retinoic acid enhances skeletal muscle progenitor formation and bypasses inhibition by bone morphogenetic protein 4 but not dominant negative beta-catenin. BMC Biol 7, 67.
  • McMahon JA Takada S, Zimmerman LB, Fan CM, Harland RM, McMahon AP. Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite. Genes Dev. 1998 May 15; 12(10): 1438-52.
  • Rspo3 binds syndecan 4 and induces Wnt/PCP signaling via clathrin-mediated endocytosis to promote morphogenesis.
  • Muscle cell differentiation of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents.
  • Paraxial mesodermal progenitors derived from mouse embryonic stem cells contribute to muscle regeneration via differentiation into muscle satellite cells. Stem Cells 26, 1865-73.

Abstract

The present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.

Description

METHOD FOR PREPARING INDUCED PARAXIAL MESODERM PROGENITOR
(IPAM) CELLS AND THEIR USE
FIELD OF THE INVENTION:
The present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
BACKGROUND OF THE INVENTION:
Embryonic stem (ES) cell research offers unprecedented potential for understanding fundamental developmental processes, such as lineage differentiation. Embryonic stem cell lines are derived from early embryos and are characterized by their ability to self-renew, that is, to be maintained indefinitely in a proliferative and undifferentiated state in culture. ES cells are also pluripotent, meaning they retain the capacity to differentiate into the three embryonic lineages: ectoderm, mesoderm and endoderm plus all of their derivatives (Chambers I., 2004). The recent development of reprogramming technologies now allows ES- like stem cells to be generated from somatic cells, such as fibroblasts. Introduction into somatic cells of a small set of specific transcription factors - Oct4, Sox2, c-Myc, and Klf4 in the mouse (Takahashi and Yamanaka, 2006) and human (Park et al., 2008b; Takahashi et al., 2007), or Oct4, Sox2, Nanog and Lin28 in human (Yu et al, 2007) - can reprogram various differentiated cell types to an ES-like stem cell state (induced pluripotent stem cells or iPS). This strategy now allows the generation of ES-like cell lines from individual patients and, thus, offers the possibility to create highly relevant in vitro models of human genetic diseases. Such reprogrammed cell lines have already been generated from patients with a variety of diseases, such as Duchenne Muscular Dystrophy or Amyotrophic lateral sclerosis (ALS) and differentiation of the reprogrammed cells into the deficient tissue has been achieved for iPS cells from patients affected with several diseases such as ALS, thus, demonstrating the feasibility of the approach (Dimos et al, 2008; Park et al, 2008a). - -
Whereas some lineages such as cardiac myocytes or neurons are easily generated in vitro from ES cells, differentiating paraxial mesoderm derivatives such as skeletal muscle, dermis, cartilage or bone from ES or iPS cells has proven to be challenging. Given the promises offered by cellular replacement therapy for the cure of some muscular degenerative diseases or for orthopaedic surgery, the development of protocols for production of precursors of muscle and skeletal lineages is of key importance. In the embryo, the muscles, the dorsal dermis and the axial skeleton of the body derive from the paraxial mesoderm and more specifically from multipotent precursors forming the presomitic mesoderm (PSM). These precursors are characterized by expression of the genes Brachyury (T), Tbx6 and Mesogeninl (Msgnl) (Chapman et al, 1996; Yoon and Wold, 2000) and they mostly differentiate into skeletal muscles, dermis, skeletal lineages, as well as in a variety of other derivatives including adipocytes and endothelial cells. In the mouse embryo, Rspo3 (also called Cristinl, Thsd2) is strongly expressed in the PSM and somites, as well as later in condensing mesenchymal cells, (Kazanskaya et al, 2004; Nam et al, 2007). R-spondins (Rspo l to 4 genes) are secreted molecules containing a thrombospondin domain, that can activate canonical Wnt signaling and Beta-Catenin, via the Fzd/LRP/Lgr4/Lgr5 co-receptors complex (Carmon et al, 2011 ; de Lau et al, 2011 ; Kim et al, 2008; Nam et al, 2006), but they were also shown to bind Syndecan4 and induce Wnt/PCP signaling (Ohkawara et al, 2011). Interestingly, biochemical assays show that Rspo2 and 3 are more potent to activate Wnt signaling than Rspol and 4 (Kim et al, 2008). R-spondins have also been shown to be implicated in bone formation and chondro genesis (Hankenson et al, 2010; Jin et al, 2011 ; Ohkawara et al, 2011), myogenesis (Han et al, 2011; Kazanskaya et al, 2004) and angiogenesis (Kazanskaya et al, 2008).
Bone Morpho genetic Proteins (BMPs) are secreted molecules of the TGFbeta superfamily that can dimerize and activate BMP signaling and bind to a receptor complex constituted of BMP receptor type I and type II (BMPR-I and -II). More precisely, BMPR-I can consist of Activin receptor-like kinase (ALK)-2/3 and 6 (also known as ActR-IA , BMPR-IA and BMPR-IB respectively). Similarly, BMPR-II can consist of BMPR-II, ActR- IIA and ActR-IIB. The BMP receptor complex is formed by an heterotetrameric complex of two BMPR-I and two BMPR-II. The BMP receptor contains an intracytoplasmic serine/threonine kinase domain which allows the phosphorylation of Smad 1/5/8 upon binding of the BMP dimer. Phosphorylated Smadl/5/8 then associate to Smad4 and shuttle to the nucleus to activate target genes, which include the inhibitor of DNA binding (Id) 1/2/3 genes - -
[Hollnagel A et al, 1999]. Importantly, numerous BMP/TGFP secreted agonists and antagonists have been described to regulate and fine-tune BMP signaling during development. Most notably noggin, chordin, follistatin and gremlin block BMP signaling by sequestrating secreted BMP, preventing its binding to the receptor. BMP ligands (prominently BMP2, 4 and 7), BMP receptors, Smads, Co-Smads and BMP agonists/antagonists have been implicated in mesoderm specification and organogenesis during development [Derynck Rik, 2008; Reshef R. et al, Gen Dev 1998; Wijgerde M. et al, 2005; McMahon JA et al, 1998; Stafford DA et al, 2011; Pourquie O. et al, 1996 and Tonegawa A. et al, 1997].
Differentiation of ES cells into paraxial mesoderm and its derivatives is highly inefficient in vitro. Limited spontaneous skeletal muscle differentiation has been described following culture of mouse embryoid bodies and DMSO treatment (Dinsmore et al, 1996; Rohwedel et al, 1994), or Retinoic acid treatment (Kennedy et al, 2009). Two distinct strategies to differentiate mouse and human ES cells in vitro to the muscle lineage have been reported. The first one involves the sorting of precursors using surface markers. For instance, Studer's group reported the isolation of human ES cells-derived CD73+ mesenchymal precursors and their subsequent differentiation into skeletal muscle following a culture period in serum containing medium (Barberi et al, 2007). The antibody against satellite cells SM/C- 2.6 was also used to isolate myogenic cells differentiated from mouse ES and iPS cells (Fukada et al, 2004; Mizuno et al, 2010). Finally, mesoderm precursors differentiated from mouse ES cells were also isolated based on their expression of other surface markers such as the Platelet derived growth factor receptor alpha (PDGFRa) or Vascular endothelial growth factor receptor 2 (VEGFR2) ((Sakurai et al, 2009; Sakurai et al, 2008) Sakurai H. et al, 2006; Takebe A. et al, 2006). Whether this combination of markers is strictly specific for paraxial mesoderm precursors has however not been demonstrated. The second strategy is based on forced expression of the transcription factors Pax3 or MyoD, or of the secreted factor Insulin Growth Factor 2 (IGF-2) in mouse ES cells (Darabi et al, 2008; Darabi et al, 2011; Dekel et al, 1992; Prelle et al, 2000; Shani et al, 1992). However, these strategies show either limited efficiency or require introduction of exogenous DNA in the ES cells which is a major hurdle for the development of safe cell therapies and the differentiated cells often show limited proliferation and engraftment potential.
Therefore, there is a need to develop better ES and iPS cell differentiation strategies to produce muscle cells and paraxial mesoderm derived lineages for the development of applications in regenerative medicine. - -
The present invention fulfils this need by providing a method for preparing multipotent progenitor cell lines expressing markers of the paraxial mesoderm progenitors and referred to as induced Paraxial Mesoderm progenitor cells or iPAM to distinguish them from the natural embryo Paraxial Mesoderm progenitor cells. Like their in vivo counterpart, the iPAM cells are capable of giving rise to cell lineages of the muscular, skeletal (bone and cartilage), dermal tissue, and derivatives such as adipocytes and endothelium. The inventors have shown that embryonic stem cells or pluripotent reprogrammed cells (iPS) can be differentiated into induced Paraxial Mesoderm progenitor (iPAM) cells using a limited number of factors. In particular, the inventors have made the surprising finding that it is possible to efficiently obtain induced Paraxial Mesoderm progenitor (iPAM) cells by treatment with specific factors, without any genetic modification of the target cells. They have shown that the obtained induced Paraxial Mesoderm progenitor (iPAM) cells exhibit characteristics of endogenous Paraxial mesoderm progenitor cells. To the applicant's knowledge, the invention is the first description of a method for obtaining unlimited amounts of cells suitable for use as progenitor cells for regenerating either muscle, skeletal, adipose or dermal tissues and paraxial mesoderm derived endothelium. Therefore the invention is highly useful in particular in regenerative medicine.
SUMMARY OF THE INVENTION:
Thus, the present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signalling pathway.
Particularly, the present invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
More particularly, the invention relates to an ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of a - - member of the R-spondin family and an effective amount of an inhibitor of the Bone Morpho genetic Protein (BMP) signaling pathway.
The invention also relates to an alternate method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of a member of an inhibitor of the GSK-3B and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
DETAILED DESCRIPTION OF THE INVENTION: Method for preparing induced paraxial mesoderm progenitor (iPAM) cells
A first aspect of the invention relates to an ex vivo method for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway.
In another particular aspect, the invention also relates to an ex vivo method for preparing a population of induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway.
In a particular aspect, the invention also relates to an ex vivo method for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
In a particular aspect, the invention relates to an ex vivo method for preparing a population of induced Paraxial Mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway. - -
As used herein, the term "Wnt signaling pathway" denotes a signaling pathway which may be divided in two pathways: the "canonical Wnt/beta catenin signaling pathway" and the "Wnt/PCP signaling pathway". As used herein, the term "canonical Wnt/beta catenin signaling pathway" or "Wnt/PCP signaling pathway" in its general meaning denotes a network of proteins and other bioactive molecules (lipids, ions, sugars...) best known for their roles in embryogenesis and cancer, but also involved in normal physiological processes in adult animals. The "canonical Wnt/beta catenin signaling pathway" is characterized by a Wnt dependant inhibition of glycogen synthase kinase 3B (GSK-3B), leading to a subsequent stabilization of B-catenin, which then translocates to the nucleus to act as a transcription factor. The "Wnt/PCP signaling pathway" does not involve GSK-3 B or B-catenin, and comprises several signaling branches including Calcium dependant signaling, Planar Cell Polarity (PCP) molecules, small GTPases and C-Jun N-terminal kinases (JNK) signaling. These pathways are well described in numerous reviews such as (Clevers, 2006; Montcouquiol et al, 2006; Schlessinger et al, 2009).
In one embodiment, the Wnt signaling pathway is the canonical Wnt/B-catenin signaling pathway.
In another preferred embodiment, the Wnt signaling pathway is the Wnt/PCP signaling pathway.
In another preferred embodiment, the Wnt signaling pathway is the canonical Wnt/ B- catenin signaling pathway and Wnt/PCP signaling pathway.
As used herein the term "activator" denotes a substance that enhances Wnt signaling activity. For example, for the canonical Wnt/ B-catenin signaling pathway, this activity can be measured by Wnt reporter activity using established multimers of LEF/TCF binding sites reporters, and/or inhibition of GSK-3B, and/or activation of canonical Wnt target genes such as T, Tbx6, Msgnl, or Axin2.
As used herein the term "induced Paraxial Mesoderm progenitor cells" or "iPAM" refers to cells derived from any cell type but exhibiting characteristics of progenitor cells of the Paraxial Mesoderm. In one embodiment, the iPAM cells are characterized by the following properties:
a) they express biomarkers characteristic of Paraxial mesoderm progenitor cells such as Tbx6, EphrinAl, EphrinB2, EPHA4, PDGFRalpha, Salll, Sall4, Dill, D113, Papc - -
(Pcdh8), Lfng, Hes7, Ripply 1, Ripply2, Brachyury (T), Cdx2, Cdx4, Evxl, Cxcr4, I117rd, Fgf , Fgfl7, Gbx2, Wnt3a, Wnt5b, Rspo3, SP5, SP8, Has2, Dkkl, Dactl, Pax3, Pax7, Mespl, Mesp2 or Msgnl genes. Preferentially Msgnl gene as measured for example with a gene reporter assay comprising the Msgnl promoter, and;
b) they are multipotent cells, capable of differentiating into at least skeletal, dermis or muscle cell lineages;
c) optionally, they may have long term self-renewal properties, e.g., they can be maintained in culture more than 6 months. The multipotency of said induced Paraxial Mesoderm progenitor (iPAM) cells can be tested in vitro, e.g., by in vitro differentiation into skeletal, dermal or muscle cell lineages using the protocols described below, and in particular in the Examples.
As used herein, the term "multipotent" refers to cells that can differentiate in more than one cell lineage depending on the environmental and culture conditions. Contrary to embryonic stem cells which are pluripotent and can differentiate into all types of somatic cell lineages, the induced paraxial mesoderm progenitor cells of the present invention have limited differentiation capacity.
The term "pluripotent cells" as used herein refers to mammalian undifferentiated cells which can give rise to a variety of different cell lineages. Typically, pluripotent cells may express the following markers Oct4, SOX2, Nanog, SSEA 3 and 4, TRA 1/81, see International Stem Cell Initiative recommendations, 2007.
In one embodiment, the pluripotent cells are human pluripotent cells.
In another embodiment, the pluripotent cells are non-human mammalian pluripotent cells.
In one embodiment, the pluripotent cells are stem cells.
Typically, said stem cells are embryonic stem cells.
In another embodiment, the pluripotent cells are human embryonic stem cells (hES cells). Typically, hES cell lines (Loser et al, 2010) such as the one described in the following table may be employed for the method of the invention: - - passage country of
line karyotype available origin origin
SA01 46XY 25 Sweden Cellartis AB
VUB01 46XY 73 Belgium AZ-VUB Bruxel
HUES 24, 46XY 26 USA Harvard
HI 46XY, Wicell research
20ql l .21 26 USA Institute
H9 Wicell research
46XX 27 USA Institute
WT3 46XY 35 UK UKSCB
HUES1
46XX 33 USA Harvard
In one embodiment, the pluripotent cells are non-human embryonic stem cells, such as mouse stem cells, rodent stem cells or primate stem cells.
In one embodiment, the pluripotent cells are induced pluripotent stem cells (iPS).
Induced pluripotent stem cells (iPS cells) are a type of pluripotent stem cells artificially derived from a non-pluripotent, typically an adult somatic cell, by inducing a "forced" expression of certain genes. iPS cells were first produced in 2006 from mouse cells (Takahashi and Yamanaka, 2006) and in 2007 from human cells (Takahashi et al., 2007; Yu et al., 2007).
In another embodiment, the activator of the canonical Wnt/B-catenin signaling pathway or the Wnt/PCP signaling pathway according to the invention is a member of the R- spondin family, originating from a vertebrate species or modified.
In another embodiment, the member of the R-spondin family is a member of the mammalian R-spondin family. - -
In a particular embodiment, the member of the R-spondin family according to the invention is selected in the group consisting of R-spondin 1, R-spondin 2, R-spondin 3 and R- spondin 4.
In a particular embodiment, the member of the R-spondin family according to the invention is R-spondin 3.
In a particular embodiment, the member of the R-spondin family according to the invention is R-spondin 2.
As used herein, the term "R-spondin3" or "R-spondin2" refers to members of the family of secreted proteins in vertebrates that activate the Wnt signaling pathway.
An exemplary sequence for human R-spondin3 protein is deposited in the database under accession number NP l 16173.2 (SEQ ID NO: l). An exemplary sequence for mouse R- spondin3 protein is deposited in the database under accession number NP 082627.3 (SEQ ID NO:2). An exemplary sequence for human R-spondin2 protein is deposited in the database under accession number NP 848660.3 (SEQ ID NO:3). An exemplary sequence for mouse R- spondin2 protein is deposited in the database under accession number NP 766403.1 (SEQ ID NO:4).
As used herein, the term "R-spondin3" also encompasses any functional variants of R- spondin3 wild type (naturally occurring) protein, provided that such functional variants retain the advantageous properties of differentiating factor for the purpose of the present invention. In one embodiment, said functional variants are functional homologues of R-spondin3 having at least 60%, 80%, 90% or at least 95% identity to the most closely related known natural R- spondin3 polypeptide sequence, for example, to human or mouse polypeptide R-spondin3 of SEQ ID NO: l or SEQ ID NO:2 respectively, and retaining substantially the same Wnt activation activity as the related wild type protein. In another embodiment, said functional variants are fragments of R-spondin3, for example, comprising at least 50, 100, or 200 consecutive amino acids of a wild type R-spondin3 protein, and retaining substantially the same Wnt activation activity. In another embodiment, such functional variant can consist in R-spondin3 gene product isoforms such as the isoform 2 of the human R-spondin3 as described under the ref. Q9BXY4-2 and CAI20142.1 (SEQ ID NO:5).
As used herein, the term "R-spondin2" also encompasses any functional variants of R- spondin2 wild type (naturally occurring) protein, provided that such functional variants retain the advantageous properties of differentiating factor for the purpose of the present invention. - -
In one embodiment, said functional variants are functional homologues of R-spondin2 having at least 60%, 80%, 90% or at least 95% identity to the most closely related known natural R- spondin2 polypeptide sequence, for example, to human or mouse polypeptide R-spondin2 of SEQ ID NO:3 or SEQ ID NO:4 respectively, and retaining substantially the same Wnt activation activity as the related wild type protein. In another embodiment, said functional variants are fragments of R-spondin2, for example, comprising at least 50, 100, or 200 consecutive amino acids of a wild type R-spondin2 protein, and retaining substantially the same Wnt activation activity. In another embodiment, said functional variants can consist in R-spondin2 gene product isoforms such as the isoform 2 or the isoform 3 of the human R- spondin2 such as described respectively under the ref. Q6UXX9-2 (SEQ ID NO: 6) or under the ref. Q6UXX9-3 (SEQ ID NO:7).
As used herein, the percent identity between the two amino-acid sequences is a function of the number of identical positions shared by the sequences (i. e., % identity = # of identical positions/total # of positions x 100), taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm, as described below.
The percent identity between two amino-acid sequences can be determined using the algorithm of E. Meyers and W. Miller (Comput. Appl. Biosci., 4: 11-17, 1988) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4.
In another embodiment, the activator according to the invention is a combination of the R-spondin 3 and R-spondin 2.
In another embodiment, the activator according to the invention may be the human R- spondin-3 isoform 2 of sequence SEQ ID NO:5.
In another embodiment, the activator according to the invention may be the human R- spondin-2 isoform 2 of sequence SEQ ID NO:6, or the human R-spondin-2 isoform 3 of sequence SEQ ID NO:7.
In a particular embodiment, the concentration of R-spondin3 used for culture of pluripotent cells is between 0.1 ng/ml and 500 ng/ml, preferably between 1 ng/ml and 500 ng/ml and more preferably between 5 ng/ml and 30 ng/ml. - -
In a particular embodiment, the concentration of R-spondin2 used for culture of pluripotent cells is between 1 ng/ml and 500 ng/ml, preferably between 5 ng/ml and 30 ng/ml.
In a particular embodiment, the concentration of R-spondin3 or R-spondin2 is 10 ng/ml. With a concentration of 10 ng/ml, more than 50% up to 70% of pluripotent cells are differentiated in induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, pluripotent cells are cultured with R-spondin3 or R-spondin2 during 1 to 15 days, or for a shorter time period. In a particular embodiment, pluripotent cells are cultured with R-spondin3 or/and R-spondin2 during at least 10 days at a concentration of 10 ng/ml.
As used herein, the term '"inhibitor of the BMP signaling pathway" denotes any compound, natural or synthetic, which results in a decreased activation of the BMP (bone morphogenetic protein) signaling pathway, which is characterized by the binding of a dimer BMP proteins to an heterocomplex constituted of BMP type I and type II receptors, which results in a phosphorylation cascade leading to the phosphorylation of Smadl/5/8, and resulting in target genes activation, such as Id genes. Typically, an inhibitor of the BMP signaling pathway provokes a decrease in the levels of phosphorylation of the proteins Smad 1, 5 and 8 (Gazzero and Minetti, 2007).
The skilled person in the art knows how to assess whether a given compound is an inhibitor of the BMP signaling pathway. Typically, a compound is deemed to be an inhibitor of the BMP signaling pathway if, after culturing cells in the presence of said compound, the level of phosphorylated Smad 1, 5 or 8 is decreased compared to cells cultured in the absence of said compound. Levels of phosphorylated Smad proteins can be measured by Western blot using antibodies specific for the phosphorylated form of said Smad proteins.
Target genes activation, such as Id genes, can typically be measured by direct Idl/2/3 transcripts (mRNA) production, via quantitative real-time PCR (qRT-PCR) and expression levels can be compared to control situation, in the absence of said compound. The inhibitor of the BMP signaling pathway may be a BMP antagonist, a chemical compound that blocks BMP type I and/or type II receptors activity (BMP type I/II receptor inhibitor), an inhibitor of BMP type I and/or type II gene expression, or a molecule which inhibits any downstream step of the BMP signaling pathway. The inhibitor of BMP signaling - - may be a natural or a synthetic compound. When the inhibitor of the BMP signaling pathway is a protein, it may be a purified protein or a recombinant protein or a synthetic protein.
In one embodiment, the inhibitor of the BMP signaling pathway is a BMP type I receptors inhibitor.
Many methods for producing recombinant proteins are known in the art. The skilled person can readily, from the knowledge of a given protein's sequence or of the nucleotide sequence encoding said protein, produce said protein using standard molecular biology and biochemistry techniques. In one embodiment of the invention, the inhibitor of the BMP signaling pathway is selected from the group consisting of Noggin, Chordin and related proteins (Chordin-like 1/2/3), Follistatin and related proteins (Follistatin-like 1/2/3/4/5), proteins of the Dan family (including Cerberus 1, Gremlin 1 and 2, Cerl-2 (Coco), SOST (Sclerostin), SOSTDC1 (Wise)) and variants and fragments thereof which inhibit the BMP signaling pathway.
In another embodiment of the invention, the inhibitor of the BMP signaling pathway is selected from the group consisting of BMP- 1/Tolloid- like proteins, TWSGl (twisted gastrulation), TMEFFs (Tomoregulins), Biglycan, TSK (Tsukushi), BMPER (Crossveinless 2), Ogon (Sizzled), AMN (Amnionless), CTGF (Connective Tissue Growth Factor), and HSPGs (including Glypican3 and Syndecan4).
In another embodiment, the inhibitor of the BMP signaling pathway is noggin. Noggin can be murine (mouse noggin exemplified by GenPept accession number NP 032737, SEQ ID NO: 10) or human noggin (human noggin exemplified by GenPept accession number EAW94528, SEQ ID NO: l 1). It may be purified or recombinant. It may be in monomeric or dimeric form.
In one embodiment, the inhibitor of the BMP signaling pathway is a compound that inhibits BMP signaling transduction cascade. In a particular embodiment, the compound that inhibits BMP signaling transduction cascade is a synthetic or a chemical compound. In another embodiment, the inhibitor of the BMP signaling pathway is an inhibitor of
BMP type I receptors.
As used herein, the term "BMP type I receptors" for "Bone Morphogenetic Protein" denotes transmembrane proteins with serine/threonine protein kinase activity that mediates the addition of phosphate molecules on certain serine and threonine amino acids on particular - - cellular substrates. It is well known in the art that an inhibitor of BMP type I receptors may block the BMP signaling pathway, see for example Yu et al, Nat Chem Bio 1.2008.
In a preferred embodiment, the inhibitor of BMP type I receptors is Dorsomorphin, a chemical compound or any derivatives generated by structure-activity studies [Cuny GD et al, 2008]. Dorsomorphin (6-[4-(2-Piperidin-l-yl-ethoxy)phenyl]-3-pyridin-4-yl-pyrazolo[l,5- ajpyrimidine , also known as Compound C) is inhibiting specifically BMP type I receptors (ALK2, 3, and 6) [Yu PB et al, 2008].
Recombinant Noggin can be purchased from R&D Systems or Peprotech or can be produced using standard techniques as described above.
Typically, the inhibitor of the BMP signaling pathway is added to the culture medium of the invention in a concentration ranging from 1 to 10000 ng/ml, preferably from 5 to 1000 ng/ml, preferably from 5 to 500 ng/ml, preferably from 10 to 200 ng/ml, even more preferably at about 200 ng/ml.
Typically, noggin is added to the culture medium of the invention at a concentration ranging from 1 to 1000 ng/ml, preferably from 10 to 200 ng/ml, even more preferably at about 200 ng/ml.
Typically, Dorsomorphin is added to the culture medium of the invention in a concentration ranging from 0.1 to 2 μΜ, preferably at 1 μΜ.
In one embodiment, pluripotent cells are cultured with the inhibitor of the BMP signaling pathway during 1 to 4 days.
In one embodiment, the culture medium according to the invention comprises a Wnt activator and an inhibitor of BMP signalling pathway according to the invention to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In one embodiment, the Wnt activator is R-spondin3 and the inhibitor of BMP signalling pathway is Noggin.
In another embodiment, the culture medium according to the invention may further comprise DMSO (Dimethyl sulfoxide) or an equivalent of the DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
As used herein, the term "equivalent" means a substance exhibiting the same properties as DMSO which is a solvent that dissolves both polar and nonpolar compounds. - -
In another embodiment, the culture medium according to the invention comprises R- spondin 3, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, the culture medium according to the invention comprises R- spondin 3 and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, the culture medium according to the invention comprises R- spondin 2, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, the culture medium according to the invention comprises R- spondin 2 and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, the culture medium according to the invention comprises R- spondin 3, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, the culture medium according to the invention comprises R- spondin 2, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In still another embodiment, the culture medium according to the invention comprises R-spondin 3, R- spondin 2, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In still another embodiment, the culture medium according to the invention comprises R-spondin 3, R-spondin 2 and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In still another embodiment, the culture medium according to the invention comprises R-spondin 3, R-spondin 2, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
Vertebrate recombinant R-spondins can be purchased commercially, or produced as conditioned culture medium. This involves expressing a construct containing the coding sequence of an R-spondin protein into competent cells, such as COS cells. R-spondin protein is secreted in the culture medium. Conditioned medium can be applied directly to pluripotent cells or prediluted in basal medium. - -
In another embodiment, the activator of the Wnt signaling pathway is an inhibitor of GSK-3 β.
As used herein, the term "GSK-3 β" for "Glycogen synthase kinase 3 beta" denotes a serine/threonine protein kinase that mediates the addition of phosphate molecules on certain serine and threonine amino acids on particular cellular substrates. It is well known in the art that an inhibitor of GSK-3 β may activate the Wnt signaling pathway, see for example (Cohen and Goedert, 2004; Sato et al, 2004; Taelman et al, 2010; Wu and Pan, 2010).
In a preferred embodiment, the inhibitor of GSK-3 β is CHIR99021.
In another preferred embodiment, the following alternatives may be used for increasing the activity of R-spondin factor in the system:
1. enhancing endogenous expression of the gene encoding said R-spondin factor or a modified form of R-spondin,
2. allowing ectopic expression of said R-spondin factor by introducing an expression vector comprising a coding sequence of R-spondin factor operably linked to control sequences into the pluripotent cells to be differentiated, or by introducing in the cells coding RNA for R-spondin factor
3. introducing directly into the cells environment an appropriate amount of R-spondin factor, for example as recombinant R-spondin factor (family of R-spondinl, 2 ,3 and 4) in the culture medium, or conditioned medium, or as substrate coating.
4. activating or inhibiting endogeneous expression of a gene involved in R-spondin factor signalling in said target cells; or,
5. overexpressing proteins involved in controlling R-spondin factor expression level, maturation and overall regulation in said target cells.
In one embodiment, the culture medium according to the invention comprises CHIR99021 and an inhibitor of BMP signaling pathway according to the invention which is Dorsomorphin to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In one embodiment, the culture medium according to the invention comprises CHIR99021, Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In one embodiment, the culture medium according to the invention comprises a Wnt activator which is a combination of R-spondin2, R-spondin3 and CHIR99021 ; and an - - inhibitor of BMP signaling according to the invention which is a combination of Noggin and Dorsomorphin to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In still another embodiment, the culture medium according to the invention comprises R-spondin 3, R-spondin 2, CHIR99021 , Dorsomorphin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In still another embodiment, the culture medium according to the invention comprises R-spondin 3, R-spondin 2, CHIR99021, Noggin and DMSO to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
In another embodiment, introducing directly into the cells environment an appropriate amount of pharmacological GSK-3P inhibitor, for example the chemical compound CHIR99021 is used as an alternative for increasing the activity of Wnt signaling pathway in the system, alone or in combination with R-spondin.
The invention relates to a composition for preparing induced Paraxial Mesoderm progenitor (iPAM) cells from pluripotent cells wherein said composition comprises an effective amount of an activator of the Wnt signaling pathway according to the invention and an effective amount of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway.
The invention also relates to a composition for preparing induced Paraxial Mesoderm progenitor (iPAM) cells from pluripotent cells wherein said composition comprises an effective amount of an activator of the Wnt signaling pathway according to the invention.
The invention also relates to a kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said kit comprising:
a) an activator of the Wnt signaling pathway
b) an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway. c) optionally, instructions for preparing induced Paraxial Mesoderm progenitor (iPAM) cells. - -
The invention also relates to a kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells, said kit comprising:
a) an activator of the Wnt signaling pathway
b) optionally, instructions for preparing induced Paraxial Mesoderm progenitor (iPAM) cells.
In a preferred embodiment, the activator is a member of the R-spondin family.
In another embodiment, the activator is selected from the group consisting of R- spondin 1, R-spondin 2, R-spondin 3 and R-spondin 4.
In another preferred embodiment, the activator is the R-spondin 2 or the R-spondin 3.
In another preferred embodiment, the activator is an inhibitor of GSK-3P such as CHIR99021.
In another embodiment, the inhibitor according to the invention is a secreted antagonist of the BMP/TGFbeta family.
In another embodiment, the inhibitor of BMP signaling pathway is selected from the group consisting of Noggin, Chordin, Chordin-like 1/2/3, Follistatin, Follistatin-like 1/2/3/4/5, a member of the Dan family, including Cerberus 1, Gremlin 1/2.
In another preferred embodiment, the inhibitor is Noggin or Follistatin.
In another preferred embodiment, the inhibitor is a chemical inhibitor of BMP signaling such as Dorsomorphin.
In a specific embodiment, said kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells comprises,
a) a composition comprising members of the R-spondin family;
b) a composition comprising an inhibitor of the BMP signaling pathway and c) DMSO or an equivalent.
In another specific embodiment, said kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells comprises,
a) a composition comprising members of the R-spondin family;
b) DMSO or an equivalent.
In a specific embodiment, said kit for preparing induced Paraxial Mesoderm progenitor (iPAM) cells comprises, - - a) a composition comprising a chemical compound inhibitor of GSK-3P; b) a composition comprising a chemical compound inhibitor of BMP type I receptors and
c) DMSO or an equivalent.
Populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells obtainable from the methods of the invention The invention further relates to populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells obtainable from the method as described above.
These populations typically may comprise other cell types in addition to induced Paraxial Mesoderm progenitor (iPAM) cells. In one embodiment, the populations of the invention are characterized in that they comprise at least 10%, 20%>, 30%>, 40%>, 50%>, 60%>, 70%, 80% and preferably at least 90% of cells that exhibit high expression of at least one biomarker characteristic of paraxial mesoderm progenitor cells, for example Msgnl gene product. Other biomarkers characteristic of paraxial mesoderm progenitor cells include, without limitation, one or more of the following proteins: Tbx6, EphrinAl, EphrinB2, EPHA4, PDGFRalpha, Salll, Sall4, Dill, D113, Papc (Pcdh8), Lfng, Hes7, Ripplyl, Ripply2, Brachyury (T), Cdx2, Cdx4, Evxl, Cxcr4, I117rd, Fgf8, Fgfl7, Gbx2, Wnt3a, Wnt5b, Rspo3, SP5, SP8, Has2, Dkkl, Dactl, Pax3, Pax7, Mespl, Mesp2.
Any methods known in the art for measuring gene expression may be used, in particular, quantitative methods such as, real time quantitative PCR or microarrays, or methods using gene reporter expression, said gene reporter comprising Msgnl promoter as described in the Examples, or qualitative methods such as immuno staining or cell sorting methods identifying cells exhibiting specific biomarkers, including cell surface markers.
As used herein, the Msgnl gene refers to the gene encoding Mesogeninl . Examples of a nucleotide sequence of a gene encoding Mesogeninl in mouse and human are given in SEQ ID NO:8 (NM 019544.1) and SEQ ID NO:9 (NM 001105569.1) respectively. - -
In one embodiment, expression of Msgnl is considered high if expression is detectable in a quantitative assay for gene expression. In another embodiment, it is high if the expression level is significantly higher than the expression level observed in the original pluripotent cells, or in cells differentiating under non specific conditions such as Basal culture medium without LIF (Leukemia Inhibitory Factor) for mouse pluripotent cells or without FGF (Fibroblast Growth Factor) for human pluripotent cells. Expression levels between the control and the test cells may be normalized using constitutively expressed genes such as GAPDH or Beta Actin. Populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells may be cultured indefinitely under appropriate growth conditions. Appropriate growth conditions may be established by the skilled person in the art based on established growth conditions for embryonic stem cells or induced pluripotent stem cells (iPS cells) for example or as described in the Examples below. Growth conditions may advantageously comprise for example the use of serum replacement medium, KSR (Gibco), ESGRO (Chemicon/Millipore) supplemented with growth factors like FGFs, WNTs, or chemical compounds modulating the respective signaling pathways.
The induced Paraxial Mesoderm progenitor (iPAM) cells may be purified or the populations may be enriched in induced Paraxial Mesoderm progenitor (iPAM) cells by selecting cells expressing markers specific of induced Paraxial Mesoderm progenitor (iPAM) cells. In one embodiment, markers specific of induced Paraxial Mesoderm progenitor (iPAM) cells for purification or enrichment of a population of induced Paraxial Mesoderm progenitor (iPAM) cells may be selected among one or more of the following markers: Msgnl, Tbx6, EphrinAl , EphrinB2, EPHA4, PDGFRalpha, Salll , Sall4, Dill , D113, Papc (Pcdh8), Lfng, Hes7, Ripply 1, Ripply2, Brachyury (T), Cdx2, Cdx4, Evxl, Cxcr4, I117rd, Fgf8, Fgfl7, Gbx2, Wnt3a, Wnt5b, Rspo3, SP5, SP8, Has2, Dkkl, Dactl, Pax3, Pax7, Mespl, Mesp2, or selected negatively with markers of other lineages/cell type such as neural fate. Purification or induced Paraxial Mesoderm progenitor (iPAM) cells enrichment may be achieved using cell sorting technologies, such as fluorescence activated cell sorting (FACS) or magnetic beads comprising specific binders of said cell surface markers of induced Paraxial Mesoderm progenitor (iPAM) cells, or fluorescent reporters for paraxial mesoderm progenitor markers. Another method consists in taking advantage of the differential adhesion - - properties of induced Paraxial Mesoderm progenitor (iPAM) cells, by selective attachment on defined substrates.
After purification or enrichment, the population may thus comprise more than 10%, 20%, 30%, 40%, 50%, 60%; 70%, 80%, 90% or more than 95% of cells having a high expression of a biomarker characteristic of induced Paraxial Mesoderm progenitor (iPAM) cells, for example, Msgnl gene product.
In another preferred embodiment, the invention relates to a composition comprising a population of induced Paraxial Mesoderm progenitor (iPAM) cells obtainable from the method as described above.
Methods for preparing cell lineages by differentiation of induced Paraxial
Mesoderm progenitor (iPAM) cells
The induced Paraxial Mesoderm progenitor (iPAM) cells may advantageously be cultured in vitro under differentiation conditions to generate skeletal muscle, bone, cartilage, dermal cells, as well as other derivatives of the paraxial mesoderm including but not restricted to adipocytes or endothelial cells.
Thus, the invention relates to a method for preparing populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages said method comprising the steps of
(a) providing a population comprising induced Paraxial Mesoderm progenitor
(iPAM) cells; and,
(b) culturing said population comprising induced Paraxial Mesoderm progenitor (iPAM) cells, under appropriate conditions for their differentiation into the desired cell lineages selected among the paraxial mesoderm derivatives which include skeletal muscle, bone, cartilage, dermal cell, adipocyte or endothelial cell lineages.
The invention further relates to a composition for preparing populations of cell lineages comprising induced Paraxial Mesoderm progenitor (iPAM) cells according to the invention and appropriate conditions for their differentiation into the desired cell lineages. - -
In one specific embodiment, the present invention provides a method for preparing a population comprising skeletal muscle cell lineages, said method comprising the steps of
(a) providing a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells;
(b) culturing said population comprising induced Paraxial Mesoderm progenitor (iPAM) cells cells in the presence of a differentiation medium comprising at least the following components:
(i) an extracellular matrix material; and,
(ii) compounds activating or inhibiting the signaling pathways known to control of the differentiation of said lineages which include but are not restricted to retinoic acid, BMP, TGFB (Transforming Growth FactorB), Hedgehog, Notch, FGF, Wnt, myostatin, insulin, PDGF, VEGF, MAPK, PI3K; and,
(c) optionally, culturing said population obtained from step (b) in a second differentiation medium comprising at least one or more compounds activating or inhibiting the Wnt, FGF, HGF (Hepatocyte growth factor), Activin, EGF (Epidermal growth factor), insulin, and IGF signaling pathways or compounds known to promote myogenic differentiation such as horse serum or transferrin,
thereby obtaining a population comprising skeletal muscle cell lineages, that can be identified by markers such as Desmin, or Myosin Heavy Chain.
The use of engineered extracellular matrices or three dimensional scaffolds has been widely described in the Art (Metallo et al, 2007). In specific embodiments, the extracellular matrix material is selected from the group consisting of Collagen I, Collagen IV, Fibronectin, Laminin, gelatin, poly-lysine, PDMS and Matrigel.
The invention further relates to a composition for preparing skeletal muscle cell lineages from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises:
i. an extracellular matrix material,
ii. at least one or more compounds activating or inhibiting the retinoic acid, BMP (Bone morphogenetic protein), TGFB, Hedgehog, Notch, FGF, Wnt, myostatin, insulin, PDGF (Platelet derived growth factor), VEGF (Vascular endothelial growth factor), MAPK, PI3K pathways. The composition further comprises at - - least another compound activating or inhibiting the Wnt, FGF, HGF, Activin, EGF, insulin, and IGF signaling pathways or compounds known to promote myogenic differentiation such as horse serum or transferrin. In another embodiment, the present invention provides a method for preparing a population comprising dermal cell lineages, said method comprising the steps of culturing a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of an efficient amount of at least one or more compounds activating or inhibiting BMP, TGFB, Wnt, FGF, EGF, retinoic acid, Notch and Hedgehog pathways. Dermal cells can be identified using markers such as Dermo- 1.
The invention further relates to a composition for preparing dermal cell lineages from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one or more compounds activating or inhibiting BMP, TGFB, Wnt, FGF, EGF, retinoic acid, Notch and Hedgehog pathways.
In another specific embodiment, the present invention provides a method for preparing a population comprising bone or cartilage cell lineages, comprising the step of culturing a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of an efficient amount of at least one or more compounds activating or inhibiting the retinoic acid, Wnt, Hedgehog, PTHrP, TGFB, BMP pathways, or compounds known to promote bone or cartilage differentiation such as dexamethasone, ascorbic acid, vitamin D3, and beta- glycerophosphate. Cartilage cells can be identified by classical staining such as Alcian Blue and bone cells with alizarin red or Von Kossa stain.
The invention further relates to a composition for preparing bone or cartilage cell lineages from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one or more compounds activating or inhibiting retinoic acid, Wnt, Hedgehog, PTHrP, TGFB, BMP pathways, or compounds known to promote bone or cartilage differentiation such as dexamethasone, ascorbic acid, vitamin D3 and beta-glycerophosphate.
In yet another embodiment, the present invention provides a method for preparing a population comprising adipocytes, said method comprising the steps of culturing the population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of - - an efficient amount of at least one or more compounds known to promote adipocyte differentiation including dexamethasone, isobutylxanthine and insulin. Adipocytes can be detected by OilRedO staining.
The invention further relates to a composition for preparing adipocytes from induced Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one compound known to promote adipocyte differentiation including dexamethasone, isobutylxanthine and insulin.
In yet another embodiment, the present invention provides a method for preparing a population comprising endothelial cells, said method comprising the steps of culturing the population comprising induced Paraxial Mesoderm progenitor (iPAM) cells in the presence of an efficient amount of at least one or more compounds activating or inhibiting the VEGF or FGF pathways. Endothelium can be detected by PECAM-1 (CD31) immuno staining. The invention further relates to a composition for preparing endothelial cells from induced
Paraxial Mesoderm progenitor (iPAM) cells, characterized in that it further comprises at least one or more compounds activating or inhibiting the VEGF or FGF pathways.
Several examples of suitable conditions for differentiating induced Paraxial Mesoderm progenitor (iPAM) cells into cartilage, muscles or endothelial cells are described in Examples below.
In another embodiment, the present invention provides populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages as well as other derivatives derived from induced Paraxial Mesoderm progenitor (iPAM) cells.
In a preferred embodiment, the present invention provides populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages as well as other derivatives derived from induced Paraxial Mesoderm progenitor (iPAM) cells obtained with a composition according to the invention.
In another embodiment, the invention relates to a composition comprising skeletal muscle, bone, cartilage, dermal cell, adipocyte or endothelial cell lineages obtainable by a method according to the invention. - - iPAM cells, population of cells derived from iPAM cells and uses thereof
Another aspect of the invention relates to the use of said populations comprising induced Paraxial Mesoderm progenitor (iPAM) cells, or said populations comprising skeletal muscle, bone, cartilage or dermal cell lineages derived from differentiation of induced Paraxial Mesoderm progenitor (iPAM) cells, but also adipose tissue and endothelial paraxial mesoderm derivatives, hereafter referred as the Populations of the Invention.
The Populations of the Invention may be used in a variety of applications, in particular, in research or therapeutic field.
One major field of application is cell therapy or regenerative medicine. For example, cells obtained from a patient suffering from a genetic defect may be cultured and genetically corrected according to methods known in the art, and subsequently reprogrammed into iPS cells and differentiated into induced Paraxial Mesoderm progenitor (iPAM) cells or its derivatives for re-administration into the patient.
Similarly, regenerative medicine can be used to potentially cure any disease that results from malfunctioning, damaged or failing tissue by either regenerating the damaged tissues in vivo by direct in vivo implantation of a population comprising induced Paraxial Mesoderm progenitor (iPAM) cells or their derivatives comprising appropriate progenitors or cell lineages.
Therefore, in one aspect, the invention relates to the induced Paraxial Mesoderm progenitor (iPAM) cells or their derivatives or the Populations of the Invention for use as a cell therapy product for implanting into a mammal, for example human patient.
In one specific embodiment, the invention relates to a pharmaceutical composition comprising a population of induced Paraxial Mesoderm progenitor (iPAM) cells obtained according to the invention. In another preferred embodiment, the invention relates to a pharmaceutical composition comprising a population of induced Paraxial Mesoderm progenitor (iPAM) cells including for example at least 102, 103, 104, 105, 106, 107, 108, or at least 109 Msgnl expressing cells. In another embodiment, this composition comprises a pharmaceutically acceptable vehicle. - -
In one specific embodiment, the Populations of the Invention are used for the treatment of a muscle genetic disorder, for example Duchenne muscular dystrophy, or any other genetic muscular dystrophy. In an embodiment, induced Paraxial Mesoderm progenitor (iPAM) cells are co- cultured with various cell types to induce their differentiation toward the desired lineage. In another embodiment, induced Paraxial Mesoderm progenitor (iPAM) cells are directly grafted into a recipient host. For regenerative medicine purposes, induced Paraxial Mesoderm progenitor (iPAM) cells can be grafted after genetic correction by methods known in the art.
In another specific embodiment, the Populations of the Invention are used in orthopaedic surgery for the treatment of joint or cartilage or bone damages caused by aging, disease, or by physical stress such as occurs through injury or repetitive strain.
In another specific embodiment, the Populations of the Invention may also be used advantageously for the production of dermal tissues, for example, skin tissues, for use in regenerative medicine or in research, in particular in the cosmetic industry or for treatment of burns and plastic surgery.
In another preferred embodiment, the invention relates to a composition comprising the Populations of the Invention. The composition comprising the Population of the invention may be used in cell therapy or regenerative medicine.
The invention will be further illustrated by the following figures and examples. However, these examples and figures should not be interpreted in any way as limiting the scope of the present invention.
FIGURES and TABLES:
Table 1: Sequences of the invention
Proteins Bank SEQ ID Sequences
Reference - - number
hRspondin NP-1 16173.2 SEQ ID MHLRLISWLF IILNFMEYIG SQNASRGRRQ
RRMHPNVSQG CQGGCATCSD YNGCLSCKPR
(CAI20141 .1 ) NO:1 LFFALERIGM KQIGVCLSSC PSGYYGTRYP
DINKCTKCKA DCDTCFNKNF CTKCKSGFYL
or HLGKCLDNCP EGLEANNHTM ECVSIVHCEV
SEWNPWSPCT KKGKTCGFKR GTETRVREII
Q9BXY4-1 QHPSAKGNLC PPTNETRKCT VQRKKCQKGE
RGKKGRERKR KKPNKGESKE AIPDSKSLES SKEIPEQREN KQQQKKRKVQ DKQKSVSVST VH mRspondin NP-082627.3 SEQ ID MHLRLISCFF IILNFMEYIG SQNASRGRRQ
RRMHPNVSQG CQGGCATCSD YNGCLSCKPR NO:2 LFFVLERIGM KQIGVCLSSC PSGYYGTRYP
DINKCTKCKV DCDTCFNKNF CTKCKSGFYL HLGKCLDSCP EGLEANNHTM ECVSIVHCEA SEWSPWSPCM KKGKTCGFKR GTETRVRDIL QHPSAKGNLC PPTSETRTCI VQRKKCSKGE RGKKGRERKR KKLNKEERKE TSSSSDSKGL ESSIETPDQQ ENKERQQQQK RRARDKQQKS VSVSTVH hRspondin NP-848660.3 SEQ ID MQFRLFSFAL IILNCMDYSH CQGNRWRRSK
RASYVSNPIC KGCLSCSKDN GCSRCQQKLF
or NO:3 FFLRREGMRQ YGECLHSCPS GYYGHRAPDM
NRCARCRIEN CDSCFSKDFC TKCKVGFYLH
Q6UXX9-1 RGRCFDECPD GFAPLEETME CVEGCEVGHW
SEWGTCSRNN RTCGFKWGLE TRTRQIVKKP VKDTILCPTI AESRRCKMTM RHCPGGKRTP KAKEKRNKKK KRKLIERAQE QHSVFLATDR ANQ mRspondin NP-766403.1 SEQ ID MRFCLFSFAL IILNCMDYSQ CQGNRWRRNK
RASYVSNPIC KGCLSCSKDN GCSRCQQKLF NO:4 FFLRREGMRQ YGECLHSCPS GYYGHRAPDM
NRCARCRIEN CDSCFSKDFC TKCKVGFYLH RGRCFDECPD GFAPLDETME CVEGCEVGHW SEWGTCSRNN RTCGFKWGLE TRTRQIVKKP AKDTIPCPTI AESRRCKMAM RHCPGGKRTP KAKEKRNKKK RRKLIERAQE QHSVFLATDR VNQ
hRspondin CAI20142.1 SEQ ID MHLRLISWLF IILNFMEYIG SQNASRGRRQ
RRMHPNVSQG CQGGCATCSD YNGCLSCKPR
isoform2 or NO:5 LFFALERIGM KQIGVCLSSC PSGYYGTRYP
DINKCTKCKA DCDTCFNKNF CTKCKSGFYL
Q9BXY4-2 HLGKCLDNCP EGLEANNHTM ECVSIVHCEV
SEWNPWSPCT KKGKTCGFKR GTETRVREII QHPSAKGNLC PPTNETRKCT VQRKKCQKGE RGKKGRERKR KKPNKGESKE AIPDSKSLES SKEIPEQREN KQQQKKRKVQ DKQKSGIEVT LAEGLTSVSQ RTQPTPCRRR YL hRspondin Q6UXX9.2 SEQ ID MRQYGECLHS CPSGYYGHRA PDMNRCARCR
IENCDSCFSK DFCTKCKVGF YLHRGRCFDE
isoform2 NO:6 CPDGFAPLEE TMECVEGCEV GHWSEWGTCS
RNNRTCGFKW GLETRTRQIV KKPVKDTILC PTIAESRRCK MTMRHCPGGK RTPKAKEKRN - -
KKKKRKLIER AQEQHSVFLA TDRANQ
hRspondin Q6UXX9-3 SEQ ID FRLFSFAL IILNCMDYSH CQGNRWRRSK
RGCRIENCDS CFSKDFCTKC KVGFYLHRGR
isoform3 NO:7 CFDECPDGFA PLEETMECVG CEVGHWSEWG
TCSRNNRTCG FKWGLETRTR QIVKKPVKDT ILCPTIAESR RCKMTMRHCP GGKRTPKAKE KRNKKKKRKL IERAQEQHSV FLATDRANQ
mMsgnl NM.019544.1 SEQ ID ATGGACAACC TGGGTGAGAC CTTCCTCAGC
CTGGAGGATG GCCTGGACTC TTCTGACACC NO:8 GCTGGTCTGC TGGCCTCCTG GGACTGGAAA
AGCAGAGCCA GGCCCTTGGA GCTGGTCCAG GAGTCCCCCA CTCAAAGCCT CTCCCCAGCT CCTTCTCTGG AGTCCTACTC TGAGGTCGCA CTGCCCTGCG GGCACAGTGG GGCCAGCACA GGAGGCAGCG ATGGCTACGG CAGTCACGAG GCTGCCGGCT TAGTCGAGCT GGATTACAGC ATGTTGGCTT TTCAACCTCC CTATCTACAC ACTGCTGGTG GCCTCAAAGG CCAGAAAGGC AGCAAAGTCA AGATGTCTGT CCAGCGGAGA CGGAAGGCCA GCGAGAGAGA GAAACTCAGG ATGCGGACCT TAGCCGATGC CCTCCACACG CTCCGGAATT ACCTGCCGCC TGTCTACAGC CAGAGAGGCC AACCGCTCAC CAAGATCCAG ACACTCAAGT ACACCATCAA GTACATCGGG GAACTCACAG ACCTCCTCAA CAGCAGCGGG AGAGAGCCCA GGCCACAGAG TGTGTGA hMsgnl NM001 10556 SEQ ID ATGGACAACC TGCGCGAGAC TTTCCTCAGC
CTCGAGGATG GCTTGGGCTC CTCTGACAGC
9.1 NO:9 CCTGGCCTGC TGTCTTCCTG GGACTGGAAG
GACAGGGCAG GGCCC I I I GA GCTGAATCAG GCCTCCCCCT CTCAGAGCCT TTCCCCGGCT CCATCGCTGG AATCCTATTC TTCTTCTCCC TGTCCAGCTG TGGCTGGGCT GCCCTGTGAG CACGGCGGGG CCAGCAGTGG GGGCAGCGAA GGCTGCAGTG TCGGTGGGGC CAGTGGCCTG GTAGAGGTGG ACTACAATAT GTTAGCTTTC CAGCCCACCC ACCTTCAGGG CGGTGGTGGC CCCAAGGCCC AGAAGGGCAC CAAAGTCAGG ATGTCTGTCC AGCGGAGGCG GAAAGCCAGC GAGAGGGAGA AGCTCAGGAT GAGGACCTTG GCAGATGCCC TGCACACCCT CCGGAATTAC CTGCCACCTG TCTACAGCCA GAGAGGCCAG CCTCTCACCA AGATCCAGAC ACTCAAGTAC ACCATCAAGT ACATCGGGGA ACTCACAGAC CTCCTTAACC GCGGCAGAGA GCCCAGAGCC CAGAGCGCGT GA mNoggin NP_032737 SEQ ID MERCPSLGVT LYALVVVLGL RAAPAGGQHY
LHIRPAPSDN LPLVDLIEHP DPIFDPKEKD NO:10 LNETLLRSLL GGHYDPGFMA TSPPEDRPGG
GGGPAGGAED LAELDQLLRQ RPSGAMPSEI KGLEFSEGLA QGKKQRLSKK LRRKLQMWLW SQTFCPVLYA WNDLGSRFWP RYVKVGSCFS KRSCSVPEGM VCKPSKSVHL TVLRWRCQRR GGQRCGWIPI QYPIISECKC SC - -
Figure imgf000029_0001
Figure 1: R-spondin induces induced Paraxial Mesoderm progenitor (iPAM) cells fate.
(A) Comparison of fluorescent Msgnl Reporter activation (YFP positive cells (YFP+ cells)) after 4 days of differentiation of mES cells (Msgnl Rep V), under default culture conditions in 15% FBS or 15% KSR medium, with or without recombinant mouse Rspo3 (lOng/mL). YFP channel, 50X. (B) Robustness of iPAM cells induction in response to mouse Rspo3 in 15% FBS medium. Triplicate wells measurements by flow cytometry. Error bar is s.e.m.
Figure 2: Flow-cytometry analysis of the induction of the Msgnl-YFP+ (induced Paraxial Mesoderm progenitor (iPAM) cells) population upon treatment with Rspo3.
(A) Flow-cytometry analysis on the MsgnlRepV mES cells at dayO of differentiation in 15% FBS medium. YFP+ population represents less than 1%. (B) Flow-cytometry analysis at day 4 of differentiation in 15% FBS medium supplemented with R-spondin3 lOng/mL. YFP+ population represents more than 70% of the total population.
Figure 3: Paraxial mesoderm progenitors (induced Paraxial Mesoderm progenitor (iPAM) cells) characterization.
(A) Differentiation of mouse MsgnlRepV reporter mES cells into iPAM cells after 4 days in culture labeled with an anti-YFP antibody and co-stained with Hoechst, xlO. (B) qRT - PCR analysis of FACS sorted iPAM YFP positive population for the paraxial mesoderm progenitors specific genes Msgnl and Tbx6, relative expression normalized to non iPAM YFP negative population expression level (fold enrichment).
Figure 4: R-spondin activity in induced Paraxial Mesoderm progenitor (iPAM) cells mediated by canonical Wnt signaling. - -
(A) Comparison of the effect of different doses of Rspo3 on iPAM induction (%YFP positive cells) after 4 days of differentiation in 15% FBS medium in the presence or absence of the canonical Wnt inhibitor Dkkl . Concentrations in ng/mL. (B) Comparison of the efficiency of the four recombinant Rspo family members on iPAM induction (%YFP positive cells) after 4 days in differentiation in 15% FBS medium. Concentrations in ng/mL. (C) Luciferase detection in MsgnlRepV reporter mES cells transfected with a Batluc reporter construct for canonical Wnt signaling activation and cultured in the presence of Rspo 3 (lOng/mL), Dkkl (50ng/mL) and LiCl (5mM) in low serum (1% FBS) containing medium. Treatment with Rspo3 strongly activates the canonical Wnt response in differentiating ES cells.
Figure 5: R-spondin activity can be mimicked by the GSK3beta inhibitor CHIR99021.
Comparison of the efficiency of Rspo3 and CHIR99021 on iPAM induction (%YFP positive cells) after 3 and 4 days in differentiation 15% FBS medium. Concentrations are in ng /mL for Rspo3 or in μΜ for CHIR99021.
Figure 6: DMSO has a positive effect on induced Paraxial Mesoderm progenitor (iPAM) cells induction.
iPAM induction after 4 days of differentiation in 15% FBS medium containing 0.5% DMSO. Optimal iPAM induction is obtained by combining R-spondins and DMSO. Concentrations in ng/mL.
Figure 7: Rspo2 and 3 activity in defined medium.
Analysis of the effect of recombinant mouse and human R-spondin 2 and 3 effect on iPAM induction (% of YFP positive cells) after 4 days of differentiation in 1% FBS (A) or 15% KSR (B) media. Concentrations in ng/mL.
Figure 8: Characterization of Populations of the Invention at day 18 of differentiation.
From day 0 to day 4, mouse ES cells were differentiated in presence of Rspo 3, followed by 15% FBS medium until day 18. Cell types were identified by tissue- specific antibody staining, namely Muscle (Desmin, green), Endothelium (PECAM1/CD31, green) and Cartilage (Alcian Blue). - -
Figure 9: Dermal and myogenic differentiation of the iPAM cells after 5 days of culture.
ES cells were cultured for 4 days in FBS 15%, DMSO 0.5% and 10 ng/ml Rspo3 and then switched to FBS 15% or FBS 1% or FBS1% plus Sonic Hedgehog (Shh) and Retinoic acid (FlShhRA) or plus Shh, Noggin and LiCl (FISNLi). Cells were harvested the next day and analyzed by qRT-PCR for the dermal marker Dermo 1 (A) and the muscle marker Myf5 (B). Graphs show fold enrichment. Figure 10: R-spondin induces iPAM fate in human ES cells.
Comparison of the expression of paraxial mesoderm progenitor markers Brachyury (A), PDGFRa (B), Tbx6 (C), Msgnl (D) measured by Q RT-PCR in HUES1 undifferentiated or cultured in 15% FBS containing medium with or without Rspo3 for up to 10 days. Relative expression to undifferentiated HUES1 cells is shown (fold induction).
Figure 11: Noggin promotes iPAM fate by counteracting BMP4 activity.
BMP4 expression at day3 (d3) and day4 (d4) of differentiation in the presence of 10 ng/ml Rspo3 and DMSO 0.5%, with (RDN) or without (RD) addition of Noggin (200ng/ml) Data are shown as normalized expression value. Grey colored data point is considered non significant. Data points are means of biological triplicate samples.
Figure 12: Molecular characterization of early stages of paraxial mesoderm differentiation.
(A) . Venn diagram comparing gene signature lists (GSL method) of posterior and anterior PSM domains and of the in vitro differentiated MsgnlRepV Venus positive ES cells harvested at day3 and 4, respectively, in presence of of 10 ng/ml Rspo3, DMSO 0.5%> and 200ng/ml Noggin. Key signature genes shared between the PSM in vivo and in vitro the Venus positive ES cells (induced Paraxial Mesoderm progenitor (iPAM) cells) are highlighted (black boxes). Red arrow shows the gene signature shift between day3 and day4 of ES cells differentiation.
(B) Representative genes from the Signature Gene lists common to the PSM and to the MsgnlRepV cells differentiated in vitro for 3 and 4 days (induced Paraxial Mesoderm progenitor (iPAM) cells). The genes shown were validated as strongly expressed and paraxial mesoderm specific by in-situ hybridization (data not shown). Whereas genes activated at day - -
3 were mostly specific to the posterior PSM, genes activated at day 4 clearly showed acquisition of anterior PSM identity.
Figure 13: Pax3-positive PSM progenitors differentiated from ES cells in vitro can generate muscle fibers in vivo.
(A) Tibialis anterior muscles were collected after 1 month post-transplantation with in vitro differentiated Pax3-positive cells (induced Paraxial Mesoderm progenitor (iPAM) cells) labeled with a CAG-GFP lentivirus (Transverse section). Control (Crtl, non-grafted area) and engrafted area were stained (red) with antibodies against Dystrophin (DYS) and Laminin (LAMA1). Engrafted progenitors produce large areas of muscle fibers expressing dystrophin and Laminin. Nuclei are counterstained with Hoechst. Scale bar, ΙΟΟμιη.
(B) Tibialis anterior muscle collected after 1 month post-transplantation (Transverse section). Control (Crtl) and Engrafted area were stained with antibodies against Myogenin (MYOG) and PAX7 (red). Nuclei are counterstained with Hoechst. For PAX7 panels, insert panels show GFP distribution. Scale bar, ΙΟΟμιη.
(C) Engrafted cells express the embryonic myosin MyHCemb, MyHCI (slow) and MyHCperi /MyHC II (fast) (Left panel), and overlay with corresponding GFP/Hoechst conterstaining is shown in Right panel. For each antibody, grafted tissue is shown on top and control tissue is shown below. Scale bar, 200μιη.
EXAMPLES: Material & Methods Cell culture
Undifferentiated mouse ES cells MsgnlRepV (El 4 derived) were maintained on gelatin-coated dishes in DMEM supplemented with 15% fetal bovine serum (FBS; from PAA), penicillin, streptomycin, 2mM L-Glutamine, O. lmM non essential amino acids, ImM sodium pyruvate, 0.2%> β-mercaptoethanol and 1 ,500 U/mL LIF. ES cells were co-cultured with mytomicin- inactivated MEFs (feeders). Undifferentiated human ES cells were cultured on plates coated with matrigel (BD Biosciences) in mTeSR medium (STEMCELL Technologies). Cultures were maintained in an incubator in 5% C02 at 37°C.
Differentiation of ES cells - -
ES cells were trypsinized and plated at various densities in gelatin coated, feeder-free, 24 well plates directly in serum-based (15% FBS) or serum-replacement (15% KSR, Invitrogen) conditions supplemented with factors, and DMSO (Sigma). Recombinant proteins were obtained commercially (R&D) and stock solutions were prepared according to manufacturer's recommendation. The GSK-3B inhibitor CHIR99021 and the BMP type I receptors inhibitor Dorsomorphin were purchased from Stemgent and prepared according to the manufacturer's recommendations. Fluorescent reporter analysis and image acquisition were done on a Zeiss Axiovert system. FACS analysis and cell sorting
Cell cultures were dissociated by trypsinization, analyzed by flow cytometry on a FACScalibur (BD Biosciences) according to YFP expression. Data were further analyzed with MoFlo software (Beckman Coulter) and FlowJo software. DNA microarrays
Mouse E9.5 embryo PSMs were microdissected and processed as previously described (Krol et al, 2011), and prepared samples were run on Affymetrix GeneChip arrays. For differentiated cell cultures, iPAMs were sorted by flow-cytometry based on their Msgnl- YFP+ expression, and samples were prepared for microarrays. Experiments were conducted on biological triplicates. Datasets were processed using Affymetrix Microarray Suite (MAS) 5.0.
Gene signature lists method
A gene expression reference was created by using all microarrays of wild-type mouse tissues deposited in GEO corresponding to Affymetrix Mouse Genome 430 2.0 Arrays (GEO platform id GPL1261). Normalization was done by calculating the mean values for each microarray. The median values for the distribution of those mean values across all microarrays were determined. This median is then used as a scaling factor for each value on each microarray. Once all microarrays have been normalized, the median expression value for each probeset is defined as the reference value. Gene signature list specific to one experimental condition was generated by normalizing the corresponding microarray data like the reference dataset. Probeset whose normalized expression value is 10 times higher than the corresponding reference value is considered to be a signature gene of that condition.
Quantitative RT-PCR - -
Total R A was extracted from ES cell cultures using Trizol (Invitrogen) or with the Rnaeasy plus mini-kit (Qiagen). RT-PCR was performed on 5ng total RNA using QuantiFast SYBR Green RT-PCR Kit (Qiagen), appropriate primers and run on a LightCycler 480II (Roche). GAPDH was used as the internal control.
Differentiated Culture Phenotyping
Cell cultures were fixed with PFA 4% overnight at 4°C. Cells were incubated 20 minutes with a blocking solution composed of 1% fetal bovine serum and 0.1%Triton in Tris Buffered Saline (TBS). Primary antibodies incubation was performed overnight at 4°C and antibodies working dilutions were as follow: anti-GFP (Abeam) was 1 : 1 ,000 , anti- Desmin (DSHB) was 1 : 100, anti-CD31 (BD Pharmingen) was 1 : 100. After TBS washes, cells were incubated with AlexaFluor488-conjugated secondary antibodies (Molecular probes) at 1 :500 for 30 minutes, and counterstained with Hoechst. Alcian Blue staining was done according to standard protocol.
Cells preparation and transplantations in injured Tibialis Anterior muscles
MsgnlRepV ES cell cultures were trypsinized after 4 days of differentiation, and iPAMs were sorted based on YFP fluorescence using a FACS Aria, or Mof ow Astrios (BD). iPAMS were permanently labeled by transduction overnight with a CAG-GFP lentivirus (MOI of 20-30). To remove non-integrated viral particules, cells were washed several time and reincubated, 1 hour in medium before preparation for transplantation. Grafted muscles were collected after 1 month and processed for immunohistochemistry. Dissected Tibialis Anterior muscles were prepared for cryosections (12μιη) as described previously [B. Gayraud-Morel B. et al, 2012]. Antibodies used in this study are anti-Dystrophin (Sigma), anti-Laminin (Sigma), Myogenin (Dako), Pax7 (DSHB) and GFP (Abeam). Antibodies for Myosins isoforms have been described in (S.J.Mathew Dev 138, 371 (2011). Tissue sections were incubated overnight with primary antibodies. Secondary antibodies conjugated with AlexaFluor (Molecular probes) were used at 1 :500. Imaging was performed on a Zeiss Axio observer and images processed with Adobe photoshop.
Results
Example 1: Use of an activator of the Wnt signaling pathway, - -
Production of induced Paraxial mesoderm progenitor (iPAM) cells in-vitro.
We first aimed at identifying key molecular players promoting differentiation of the paraxial mesoderm lineage from ES cells. First, we investigated the time-course of paraxial mesoderm induction during mouse ES cell differentiation after formation of embryoid bodies, in DMEM based medium supplemented with 15% Fetal Bovine serum (FBS 15%). Differentiation in paraxial mesoderm progenitor cells was characterized by activation of the Brachyury/T, Tbx6, and Msgnl markers detected by PCR. Our data suggest that between day 1 and 4 of culture, some differentiated cells are in a presomitic mesoderm -like stage. The MsgnlRepV reporter ES line characterization.
In order to follow the differentiation of ES cells toward the first stage of paraxial mesoderm differentiation (ie presomitic fate), which represents the first step of skeletal muscle differentiation after acquisition of a mesodermal identity, we generated a transgenic mouse ES cell line harboring a fluorescent reporter specifically expressed in paraxial mesoderm progenitors. We used the promoter from the mouse Msgnl, a gene specific for the presomitic mesoderm, to drive the expression of Venus (a modified YFP). The transgenic MsgnlRepV (Mesogeninl Reporter Venus) mouse ES cell line was subsequently validated using the tetraploid aggregation method to generate embryos entirely derived from the transgenic ES cells. As expected, transgenic mouse embryos exhibit fluorescently labeled paraxial mesoderm tissue, thus, validating the tissue specificity of Venus expression in the transgenic ES cell line.
R-spondins identification.
In order to optimize the differentiation conditions for paraxial mesoderm progenitors, we developed a manual screening assay, testing candidate growth factors and drugs interfering with various signaling pathways on ES cells. The MsgnlRepV reporter cells were plated at a defined density in 24-well plates coated with gelatin (0.1%>). Two basal culture media were selected: a DMEM based medium containing 15% fetal bovine serum (FBS, high serum) and a defined serum- free medium containing 15% KSR (Invitrogen/Gibco). These basal media were supplemented with candidate factors on day 0 of differentiation. Control and experimental conditions were cultured in parallel. Cells were left to differentiate for three to four days with medium changed on day 2 or 3. Cell cultures were analyzed on day 3 and 4 of differentiation visually and by flow cytometry for YFP+ population quantification. - -
After 4 days of differentiation, control differentiation in 15% FBS results in a low and variable induction of YFP+ cells (typically 1 to 15% of the culture), and differentiation in defined medium 15% KSR (Invitrogen) results in an even lower induction (typically 1%). Among the set of candidates tested, we identified the secreted R-spondin3 protein as being able to increase dramatically the induction of YFP+ cells. In our assay, R-spondin3 at lOng/mL is sufficient to increase significantly the induction of YFP+ cells both in FBS based medium and KSR based medium, up to 70% (Figure 1, 2 and 7). The R-spondin3 response saturated between 30 to lOOng/mL. While at day 0, the YFP+ population is <1% of the cells, in R-spondin3 supplemented differentiation medium, YFP+ population can represent more than 50%, up to 70% of the cells at day4 (Figure 2B). In human ES cells, induction of the paraxial mesoderm progenitor markers, Brachyury, PDGFRa, Tbx6 and Msgnl is observed after 3 to 10 days of culture in 15% FBS containing medium when treating huESl with R- spondin3 (Figure 10). Paraxial mesoderm progenitors characterization.
To confirm induction of a paraxial mesoderm progenitor cell fate upon differentiation of ES cells in vitro, we sorted the YFP+ cell population after four days of differentiation in presence of R-spondin3 (Figure 3 A) and analyzed the YFP+ versus YFP- cells by qRT-PCR for the key paraxial mesoderm markers Msgnl and Tbx6 (Figure 3B). We confirmed that the YFP+ population strongly expresses the Msgnl endogenous gene, as well as Tbx6, demonstrating that we are able to generate paraxial mesoderm progenitors (iPAM) in vitro.
R-spondin family and Wnt signaling.
We next asked whether other members of the R-spondin family can induce paraxial mesoderm progenitor iPAM cells (Msgnl -YFP+ paraxial mesoderm progenitors). ES cells were cultured in medium containing recombinant R-spondin proteins (R-spondins 1-4) supplemented with 15% FBS and allowed to differentiate for 4 days (Figure 4B). Two members of the family, R-spondin2 and R-spondin3, exhibit comparable activities and significantly increase the number of YFP+ cells. The activity of R-spondin family proteins has been associated with canonical Wnt/Beta catenin signaling (Kim et al, 2008; Nam et al., 2006) and more recently with Wnt/PCP signaling (Ohkawara et al, 2011). We analyzed the effect of the inhibition of canonical Wnt signaling on R-spondin dependent differentiation using the secreted Dkkl inhibitor, (Figure 4A). Supplementation of the medium with the extracellular Wnt antagonist Dkkl results in a sharp decrease of YFP+ induction. Moreover, - - adding Dkkl to FBS-containing medium blocks the effect of R-spondin3, suggesting that R- spondin3 effect is mediated by the Wnt canonical pathway. We also analyzed the expression of luciferase from a plasmid driven by a promoter responding to canonical Wnt signaling (BAT-luc) transfected in ES cells treated or not with R-spondin3, and with Dkkl or with the compound LiCl which can activate the Wnt pathway (Figure 4C). Luciferase was strongly activated by R-spondin3 treatment suggesting that it activates the canonical Wnt pathway in this context.
To further test whether R-spondin3 effect is mediated by the Wnt canonical pathway, we tested the effect of CHIR99021, a well described GSK-3B inhibitor (Ring et al., 2003). Figure 5 shows that after 4 days, CHIR99021 is as efficient as Rspo-3 in inducing YFP+ cells, suggesting that R-spondin3 effect is mediated by activation of the canonical Wnt pathway.
Dimethyl sulfoxide (DMSO) has been shown to promote the differentiation of several cell types, notably mesoderm from the P19 Embryonic Carcinoma (EC) cell line (McBurney et al, 1982; Skerjanc, 1999). The exact mechanism of action of DMSO in cell culture is not known, and it has been hypothesized that DMSO modifies the plasma membrane properties, making the cells more responsive to extracellular signals present in the differentiation medium. Addition of 0.5% of DMSO to FBS-containing medium, results in an increase of YFP+ cells after 4 days in culture (Figure 5, 6 and 7), although this increase is modest compared to the increase due to the addition of R-spondin2 or R-spondin3, or both. Interestingly, the addition of R-spondins and DMSO synergizes to enhance paraxial mesoderm progenitors differentiation (Figure 5, 6 and7). Optimal conditions for paraxial mesoderm differentiation were observed when both DMSO and R-spondin 2 and/or 3 were combined (Figure 5, 6 and 7). Importantly, this effect is also seen in a serum-free, defined KSR based medium (Figure 7B).
Paraxial mesoderm progenitors differentiation potential
We next explored the differentiation potential of the iPAM cell population. In vivo, paraxial mesoderm progenitor cells are fated to become skeletal muscles, vertebral tissues (cartilage, bone), dorsal dermis, endothelium, and other tissues such as adipose tissues.
Thus, we performed sequential differentiation protocols, aiming at first generating iPAM cells, and then differentiating them further in 15% FBS medium or by applying various described differentiation protocols (see below), in particular « Myogenic » and «Chondrogenic » media. - -
For example, between dayO to day4, ES cells were exposed to optimized differentiation conditions (ie. R-spondin3 lOng/mL, DMSO 0.5%, in 15% FBS basal medium). On day 4, culture medium was changed and cells were exposed to specific differentiation media until day 18, with medium replacement every 3 days. At day 18, cell cultures were fixed and analyzed by tissue specific histochemical stainin g o r immunofluorescence (Figure 8). Under optimized differentiation conditions, cell cultures were positive for Cartilage (Alcian blue positive nodules), Muscles (Desmin positive fibers) and Endothelium (CD31/PECAM1). Alternatively, after 4 days in differentiation conditions (ie. Rspo3 lOng/mL, DMSO 0.5%, in FBS 15% basal medium), cells were switched to FBS 15% or FBS 1 % or FBS 1 % plus Sonic Hedgehog (Shh) and Retinoic acid (F 1 ShhRA) or plus Shh, Noggin and LiCl (FISNLi). Cells were harvested the next day and analyzed by qRT- PCR for the dermal marker Dermol and the muscle marker Myf5 (Figure 9). Significant activation of these markers was observed indicating differentiation of the iPAM cells toward the dermal and muscle lineages respectively.
Myogenic protocol:
Alternatively, induced paraxial mesoderm progenitors (iPAM) cells can be differentiated in two-dimensional culture into muscle cells using SF03 medium complemented with BMP4, ActivinA and IGF-1 for 3 days, followed by 3 days of SF03 medium complemented with LiCl and Shh.
Induced paraxial mesoderm progenitors (iPAM) cells can be cultured in a hanging drop for 3 days at 800cells/20uL in differentiation medium, composed of DMEM supplemented with 10%> fetal calf serum (FCS), 5% horse serum (Sigma), 0.1 mM 2- mercaptoethanol, 0.1 mM nonessential aminoacids, and 50 ug/ml penicillin/streptomycin. After 3 days, the medium is changed and cell aggregates are transferred on a low attachement plate. At day 6, cells are plated and cultured in differentiation medium on plates coated with Matrigel (BD Bioscience, Bedford, MA, USA). Myogenic differentiation is achieved by withdrawal of FBS from confluent cells and addition of 10 ug/ml insulin, 5 ug/ml transferrin, and 2% horse serum.
Induced paraxial mesoderm progenitors (iPAM) cells can also be cultured for 3 weeks in Skeletal Muscle Cell Medium (Lonza, Chemicon) complemented with EGF, insulin, Fetuin, dexamethasone, and bFGF (100 ng/mL). - -
Osteogenic protocol:
For skeletal lineages, Induced paraxial mesoderm progenitors (iPAM) cells are exposed to 200 ng/ml human or mouse recombinant BMP4 or a combination of 1 uM retinoic acid and 10 mM Lithium Chloride. Alternatively, cells are plated on gelatin-coated plates at a density of 1-3 x 10A3 per well (24-well plate) and cultured for 28 days in bone differentiation medium (DMEM, 10%FBS, 2 mM 1-Glutamine, 1 * Penicillin/streptomycin (P/S), 0.1 μΜ dexamethasone, 50 μΜ ascorbic acid 2 -phosphate, 10 mM β-glycerophosphate, 10 ng/mL BMP4) in order to observe cells expressing bone specific markers or secreting alcian blue positive extracellular matrix. Differentiated skeletal cell lineages are identified using specific stainings for extracellular matrix components of bone and cartilage including alcian blue or alizarin red, as well as by immunofluorescence using chondrocyte- and/ or osteocyte specific antibodies.
Induced paraxial mesoderm progenitors (iPAM) cells can also be differentiated into the bone lineage using the following differentiation medium composed of DMEM, 10% FBS, 2 mM L-Glutamine, l x P/S, 0.1 mM Dexamethasone, 50 mM ascorbic acid 2-phosphate,10 mM beta-glycerophosphate, and 10 ng/mL BMP4, and vitamin D3 for 20 days, with medium changed every 3 days. Bone formation can be confirmed by staining the differentiating culture with Alizarin red, well known in the art that results to stain differentiated bone in red. Extracellular accumulation of calcium can also be visualized by von Kossa staining. Alternatively, differentiating cells can be lysed and assayed for ALP activity using BBTP reagent. Alternatively, differentiating cells can be analyzed for osteoblast lineage markers expression, for example Osterix(Osx) and Cbfal/Runx2, alkaline phosphatase, collagen type I, osteocalcin, and osteopontin.
Chondrogenic protocol:
For chondrogenic cell differentiation, induced paraxial mesoderm progenitors (iPAM) cells are plated at a density of 8 x 10A4 per well (24-well plate) and cultured for 30 minutes in a 37C incubator in cartilage differentiation medium ( MEM, 10%FBS, 2 mM 1-Glutamine, l x P/S, 0.1 μΜ Dexamethasone, 170 μΜ ascorbic acid 2-phosphate). Next, an equal amount of cartilage cell differentiation medium with 10 ng/mL TGF beta3 is added to the well. After one week, the medium is replaced with cartilage differentiation medium supplemented with - - lOng/mL Bmp2. After 21 days cartilaginous nodules secreting extracellular matrix can be observed. Induced paraxial mesoderm progenitors (iPAM) cells can also be differentiated into cartilage cells using a differentiation medium based on alphaMEM, 10%FBS, 2 mM L- Glutamine, l x P/S, 0.1 mM Dexamethasone, and 170 mM ascorbic acid 2-phosphate or DMEM supplemented with 0.1 mM dexamethasone, 0.17 mM ascorbic acid, 1.0 mM sodium pyruvate, 0.35mM L-proline, 1% insulin-transferrin sodium, 1.25 mg/ml bovine serum albumin, 5.33 ug/ml linoleic acid, and 0.01 ug/ml transforming growth factor-beta), as well as TGFbeta3 or BMP2. Cells are cultured for several weeks, with medium changed every 3 days. Differentiation can also be performed at high density on 3D scaffold such as Alginate beads in a DMEM based medium containing 10% FBS and antibiotic supplemented with 100 ng/ml recombinant human Bone Morphogenic Protein-2 (BMP-2) and 50 mg ascorbic acid. Cartilage formation can be confirmed by Alcian Blue staining of the differentiating culture, well known in the art that results in the staining of Muco-glycoproteins in blue. Alternatively, a safranin O staining can be performed.
Dermal fibroblast protocol:
Induced paraxial mesoderm progenitors (iPAM) cells can be differentiated into dermal fibroblasts by culturing them on a scaffold of collagen in medium containing a fibroblast growth factor such as bFGF (basic Fibroblast Growth Factor) or a member of the Wnt family of growth factors.
Next, to confirm that R-spondin also induces paraxial mesoderm progenitors (iPAM) cells from human ES cells differentiation, HUES1 cells were plated as single cells and differentiated in 15% FBS containing medium with or without Rspo3. qRT-PCR time course analysis for paraxial mesoderm progenitor markers expression was performed (Figure 10). Strong activation of Msgnl and Tbx6 during hES cells differentiation in presence of R- spondin3, demonstrate that iPAM cells can be differentiated from hES cells.
Example 2: Use of an activator of the Wnt signaling pathway and an inhibitor of BMP signaling pathway,
Characterization of the Msgnl-YFP+ cell population.
Using R-spondin proteins and DMSO, we are able to produce in a single step 70%> of Msgnl -YFP+ cells (Figure. 2B). To directly compare the transcriptome of Msgnl -YFP+ to - - their in-vivo counterpart (ie. presomitic mesoderm, PSM), we used microarrays to generate a global transcriptome profiling of consecutive mouse fragments representing progressively more mature stages of differentiation (data not shown). The PSM fragments spanned from the tail bud level to the somitic region where the myogenic program is first activated. Based on this microarray series, we were able to define sets of signature genes defining the progressive maturation stage of cells from the tail bud (epiblast) to the presomitic mesoderm and somitic stages. In parallel, we differentiated ES cells in presence of R-spondin3 and DMSO (RD), sorted the Msgnl-YFP+ cell population and generated microarrays of this population at day3 and day4 of differentiation respectively. Global transcriptomes and signature genes sets were compared between the in vivo PSM and in vitro differentiated Msgnl-YFP+ cells (data not shown).
We confirm that Msgnl-YFP+ cells express a number of key paraxial mesoderm markers already validated by Q-PCR such as Msgnl and Tbx6 (Figure 3B). We noticed that, in contrast to the native PSM cells, the Msgnl -YFP+ population also activates cardiac and angiogenic markers, which are a signature of more ventral/lateral mesoderm derivatives. Also, we found that TGFp/BMP signaling tends to be up-regulated in Msgnl -YFP+. Unexpectedly, we found that the Msgnl -YFP+ cell population expresses BMP4 at a significant level whereas BMP4 is detected only at very low level in vivo in the PSM. This was problematic because BMP4 has been shown to prevent cells to acquire a paraxial mesoderm fate at the expenses of lateral plate fates (Pourquie O et al, 1996, Tonegawa A et al, 1997).
Noggin to counteract BMP4 signaling
To counteract this BMP4 activity, we tested the effect of addition of recombinant Noggin (Nog) protein known to inhibit BMP4, to the differentiation medium. We found that addition of Noggin from day 0 or day 1 of differentiation, Msgnl -YFP+ cells effectively repress BMP4 expression compared to cells cultured in differentiation medium lacking Noggin. We further defined both the optimal concentration and timing of Noggin addition. Noggin does not change the total number of Msgnl -YFP+ induced Paraxial Mesoderm progenitor (iPAM) cells (efficiency of production) or the total cell number in culture but rather changes the maturation of the Msgnl-YFP+ (efficiency of maturation). To better characterize the impact of Noggin on Msgnl -YFP+ cells, we performed microarrays on the Msgnl -YFP+ induced paraxial mesoderm progenitors (iPAM) population at day 3 and 4 of differentiation (Figure 11, compare conditions RDN and RD). - -
We show that adding Noggin to the medium represses BMP4 expression in the Msgnl-YFP+ cells, leading to the upregulation of several PSM specific markers including Tbx6, Pcdh8, Pax3, Foxcl, Raldh2 and Ripply2 (Figure 12B and data not shown). BMP inhibitors and BMP signaling
We detected strong upregulation of the BMP inhibitors Follistatin (Fst) and Cerberus (Cerl) in the mouse PSM microarray series and other BMP inhibitors such as Chordin, Noggin, and Gremlinl are known to be expressed by the adjacent tissues during development [McMahon JA et al, 1998; Stafford DA et al, D2011 and Scott IC et al, 2000]. This suggests that in vivo the PSM requires BMP inhibition to mature properly, and that in vitro BMP inhibition is also required to the proper maturation of iPAM cells.
We screened a set of BMP inhibitors, including recombinant proteins Noggin (Nog), Chordin (Chd), Chordin-like 1 (Chdll), Follistatin (Fst), Follistatin- like 1 (Fstll), Dan family protein including Cerberus 1 (Cerberus) and Gremlin 1 (Greml); at varying concentration range (10-200ng/mL), and various time-window (day 0-4, day 1-4) and analyzed the impact on BMP4 and the PSM marker Tbx6 expression. Additionally, we tested the chemical compound Dorsomorphin (Compound C), a specific BMP type I receptor inhibitor, at various concentration (0.1-1μΜ) and various time-window (day 0-4, day 1-4). Addition of the BMP inhibitors does not affect the number of induced paraxial mesoderm progenitors (iPAM) cells or the total cell number in culture (data not shown). Among the set of candidates tested, we identified in particular Noggin (Nog), Follistatin (Fst) and Dorsomorphin as promoting induced paraxial mesoderm progenitors (iPAM) maturation by counteracting BMP4 and activating Msgnl and Tbx6 expression (data not shown). To monitor the PSM identity of MsgnlRepV cells differentiated in vitro, we purified them by FACS after both 3 and 4 days of differentiation in the presence of R-spondin3 and Noggin. qRT-PCR analysis confirmed that the sorted population strongly expresses Msgnl and Tbx6, as expected for PSM cells (Figure 3B). For a more systematic analysis, gene signatures were generated for both time-points as described above. Comparison of Day3 differentiated ES gene signature to that of the PSM transcriptional domains revealed that the differentiated ES cells expressed a large number of genes of the posterior PSM including T, Rspo3 and Fgf8 (Figure 12A-B). Thus the cells expressing the Msgnl reporter bear a close molecular resemblance to their counterparts in vivo. - -
We then asked whether the maturation process could be pursued further in vitro. Strikingly, Day4 but not Day3 differentiated ES cells were found to activate genes specific for the anterior PSM such as Mesp2, Ripply2 or Foxcl, indicating that these cells have acquired an anterior PSM identity (Figure 12B). In particular, these Day4 cells significantly upregulated the Pax3 gene. Pax3 regulates the differentiation of the myogenic lineage and, recently, overexpression of Pax3 in ES cells was shown to induce muscle [Darabi, R et al, 2008]. This suggested that, over time, R-spondin3 and Noggin treatment can transform undifferentiated ES cells into bona fide Pax3 positive muscle progenitors without genetic manipulation.
In conclusion, these results demonstrate the potential use and the synergistic effect of an activator of the Wnt signaling pathway like R-spondin3 and of an inhibitor of the Bone Morphogenetic Protein (BMP) signaling pathway like Noggin to obtain induced paraxial mesoderm progenitor (iPAM) cells. Example 3: Generation of myogenic lineage in vivo.
To further drive the terminal differentiation of these precursors, we next took an in vivo approach. Pax3-positive cells obtained after 4 days of differentiation in medium containing R-spondin3 and Noggin were transduced with a GFP-expressing lentivirus to permanently label them. They were then injected into the Tibialis anterior muscle of adult Rag2-/-:yc-/- immunocompromised mice that had been injured by intramuscular injection of the snake venom cardiotoxin [Gayraud-Morel B. et al, 2012]. Examination of the transplanted muscles after 1 month showed that the grafted GFP-expressing cells reconstituted large areas filled with muscle fibers expressing laminin and dystrophin (n=3; Figure 13 A). The fibers expressed high levels of the differentiation marker Myogenin, had a small diameter and were not aligned as in adult muscle suggesting that they might correspond to embryonic primary fibers (Figure 13B) [Gayraud-Morel B. et al, 2009]. This was confirmed by showing that these fibers expressed embryonic and perinatal iso forms of myosin heavy chain (Figure 13C). Remarkably, a significant population of cells expressing the satellite cell-specific marker Pax7 was observed in the engrafted regenerating area suggesting that the differentiated ES cells were also able to produce a progenitor pool of muscle cells (Figure 13B). Thus, when transplanted in vivo in injured muscles, Pax3-positive cells derived in vitro are able to continue their differentiation toward the myogenic lineage. - -
REFERENCES:
Throughout this application, various references describe the state of the art to which this invention pertains. The disclosures of these references are hereby incorporated by reference into the present disclosure.
Barberi, T., Bradbury, M., Dincer, Z., Panagiotakos, G., Socci, N. D. and Studer, L. (2007). Derivation of engraftable skeletal myoblasts from human embryonic stem cells. Nat Med 13, 642-8.
Carmon, K. S., Gong, X., Lin, Q., Thomas, A. and Liu, Q. (2011). R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/{beta}-catenin signaling. Proc Natl Acad Sci U S A 108, 11452-7.
Chal, J. and Pourquie, O. (2009). Patterning and Differentiation of the Vertebrate Spine. In The Skeletal System, (ed. O. Pourquie), pp. 41-116: Cold Spring Harbor Laboratory Press.
Chambers, I. (2004). The molecular basis of pluripotency in mouse embryonic stem cells. Cloning Stem Cells 6, 386-91.
Chapman, D. L., Agulnik, I., Hancock, S., Silver, L. M. and Papaioannou, V. E. (1996). Tbx6, a mouse T-Box gene implicated in paraxial mesoderm formation at gastrulation. Dev Biol 180, 534-42.
Clevers, H. (2006). Wnt/beta-catenin signaling in development and disease. Cell 127, 469-80.
Cohen, P. and Goedert, M. (2004). GSK3 inhibitors: development and therapeutic potential. Nat Rev Drug Discov 3, 479-87.
Cuny GD, Yu PB, Laha JK, Xing X, Liu JF, Lai CS, Deng DY, Sachidanandan C,
Bloch KD, Peterson RT. Structure-activity relationship study of bone morphogenetic protein (BMP) signaling inhibitors. Bioorg Med Chem Lett. 2008 Aug l;18(15):4388-92. Epub 2008 Jun 27.
Darabi, R., Gehlbach, K., Bachoo, R. M., Kamath, S., Osawa, M., Kamm, K. E., Kyba, M. and Perlingeiro, R. C. (2008). Functional skeletal muscle regeneration from differentiating embryonic stem cells. Nat Med 14, 134-43.
Darabi, R., Santos, F. N., Filareto, A., Pan, W., Koene, R., Rudnicki, M. A., Kyba, M. and Perlingeiro, R. C. (2011). Assessment of the Myogenic Stem Cell Compartment - -
Following Transplantation of Pax3/Pax7-Induced Embryonic Stem Cell-Derived Progenitors. Stem Cells.
de Lau, W., Barker, N., Low, T. Y., Koo, B. K., Li, V. S., Teunissen, H., Kujala, P., Haegebarth, A., Peters, P. J., van de Wetering, M. et al. (2011). Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature.
Dekel, I., Magal, Y., Pearson- White, S., Emerson, C. P. and Shani, M. (1992). Conditional conversion of ES cells to skeletal muscle by an exogenous MyoDl gene. New Biol 4, 217-24.
Derynck Rik, 2008. The TGF-β Family. CSHL. Kohei Miyazono Editors).
Dimos, J. T., Rodolfa, K. T., Niakan, K. K., Weisenthal, L. M., Mitsumoto, FL, Chung, W., Croft, G. F., Saphier, G., Leibel, R., Goland, R. et al. (2008). Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science 321, 1218-21.
Dinsmore, J., Ratliff, J., Deacon, T., Pakzaban, P., Jacoby, D., Galpern, W. and Isacson, O. (1996). Embryonic stem cells differentiated in vitro as a novel source of cells for transplantation. Cell Transplant 5, 131-43.
Fukada, S., Higuchi, S., Segawa, M., Koda, K., Yamamoto, Y., Tsujikawa, K., Kohama, Y., Uezumi, A., Imamura, M., Miyagoe- Suzuki, Y. et al. (2004). Purification and cell- surface marker characterization of quiescent satellite cells from murine skeletal muscle by a novel monoclonal antibody. Exp Cell Res 296, 245-55.
Gayraud-Morel B. et al, J Cell Sci 125, 1738 (Apr 1, 2012).
Gayraud-Morel B., F. Chretien, S. Tajbakhsh, Regen Med 4, 293 (Mar, 2009).
Gayraud-Morel B. et al. J Cell Sci 125, 1738 (Apr 1, 2012).
Han, X. H., Jin, Y. R., Seto, M. and Yoon, J. K. (2011). A WNT/beta-catenin signaling activator, R-spondin, plays positive regulatory roles during skeletal myogenesis. J Biol Chem 286, 10649-59.
Hankenson, K. D., Sweetwyne, M. T., Shitaye, H. and Posey, K. L. (2010). Thrombospondins and novel TSR-containing proteins, R-spondins, regulate bone formation and remodeling. Curr Osteoporos Rep 8, 68-76.
Hirsinger, E., Jouve, C, Dubrulle, J. and Pourquie, O. (2000). Somite formation and patterning. Int Rev Cytol 198, 1-65.
Hollnagel A, Oehlmann V, Heymer J, Riither U, Nordheim A. Id genes are direct targets of bone morphogenetic protein induction in embryonic stem cells. J Biol Chem. 1999 Jul 9;274(28): 19838-45. - -
Jin, Y. R., Turcotte, T. J., Crocker, A. L., Han, X. H. and Yoon, J. K. (2011). The canonical Wnt signaling activator, R-spondin2, regulates craniofacial patterning and morphogenesis within the branchial arch through ectodermal-mesenchymal interaction. Dev Biol 352, 1-13.
Kazanskaya, O., Glinka, A., del Barco Barrantes, I., Stannek, P., Niehrs, C. and Wu,
W. (2004). R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling and is required for Xenopus myogenesis. Dev Cell 7, 525-34.
Kazanskaya, O., Ohkawara, B., Heroult, M., Wu, W., Maltry, N., Augustin, H. G. and Niehrs, C. (2008). The Wnt signaling regulator R-spondin 3 promotes angioblast and vascular development. Development 135, 3655-64.
Kennedy, K. A., Porter, T., Mehta, V., Ryan, S. D., Price, F., Peshdary, V., Karamboulas, C, Savage, J., Drysdale, T. A., Li, S. C. et al. (2009). Retinoic acid enhances skeletal muscle progenitor formation and bypasses inhibition by bone morphogenetic protein 4 but not dominant negative beta-catenin. BMC Biol 7, 67.
Kim, K. A., Wagle, M., Tran, K, Zhan, X., Dixon, M. A., Liu, S., Gros, D., Korver,
W., Yonkovich, S., Tomasevic, N. et al. (2008). R-Spondin family members regulate the Wnt pathway by a common mechanism. Mol Biol Cell 19, 2588-96.
Krol AJ, Roellig D, Dequeant ML, Tassy O, Glynn E, Hattem G, Mushegian A, Oates AC, Pourquie O. Evolutionary plasticity of segmentation clock networks, lopment. 201 1 Jul;138(13):2783-92.
Loser, P., Schirm, J., Guhr, A., Wobus, A. M. and Kurtz, A. (2010). Human embryonic stem cell lines and their use in international research. Stem Cells 28, 240-6.
McBurney, M. W., Jones- Villeneuve, E. M., Edwards, M. K. and Anderson, P. J. (1982). Control of muscle and neuronal differentiation in a cultured embryonal carcinoma cell line. Nature 299, 165-7.
McMahon JA, Takada S, Zimmerman LB, Fan CM, Harland RM, McMahon AP. Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite. Genes Dev. 1998 May 15; 12(10): 1438-52.
Metallo, C. M., Mohr, J. C, Detzel, C. J., de Pablo, J. J., Van Wie, B. J. and Palecek, S. P. (2007). Engineering the stem cell micro environment. Biotechnol Prog 23, 18-23.
Mizuno, Y., Chang, H., Umeda, K, Niwa, A., Iwasa, T., Awaya, T., Fukada, S., Yamamoto, H., Yamanaka, S., Nakahata, T. et al. (2010). Generation of skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells. FASEB J 24, 2245-53. - -
Montcouquiol, M., Crenshaw, E. B., 3rd and Kelley, M. W. (2006). Noncanonical Wnt signaling and neural polarity. Annu Rev Neurosci 29, 363-86.
Nam, J. S., Turcotte, T. J., Smith, P. F., Choi, S. and Yoon, J. K. (2006). Mouse cristin/Pv-spondin family proteins are novel ligands for the Frizzled 8 and LRP6 receptors and activate beta-catenin-dependent gene expression. J Biol Chem 281, 13247-57.
Nam, J. S., Turcotte, T. J. and Yoon, J. K. (2007). Dynamic expression of R-spondin family genes in mouse development. Gene Expr Patterns 7, 306-12.
Ohkawara, B., Glinka, A. and Niehrs, C. (2011). Rspo3 binds syndecan 4 and induces Wnt/PCP signaling via clathrin-mediated endocytosis to promote morphogenesis. Dev Cell 20, 303-14.
Park, I. H., Arora, N., Huo, H., Maherali, N., Ahfeldt, T., Shimamura, A., Lensch, M. W., Cowan, C, Hochedlinger, K. and Daley, G. Q. (2008a). Disease-specific induced pluripotent stem cells. Cell 134, 877-86.
Park, I. H., Zhao, R., West, J. A., Yabuuchi, A., Huo, H., Ince, T. A., Lerou, P. H., Lensch, M. W. and Daley, G. Q. (2008b). Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, 141-6.
Pourquie O, Fan CM, Coltey M, Hirsinger E, Watanabe Y, Breant C, Francis- West P, Brickell P, Tessier-Lavigne M, Le Douarin NM. Lateral and axial signals involved in avian somite patterning: a role for BMP4. Cell. 1996 Feb 9;84(3):461-71.
Prelle, K., Wobus, A. M., Krebs, O., Blum, W. F. and Wolf, E . (2000) .
Overexpression of insulin-like growth factor-II in mouse embryonic stem cells promotes myogenic differentiation. Biochem Biophys Res Comrnun 277, 631-8.
Reshef R, Maroto M, Lassar AB. Regulation of dorsal somitic cell fates: BMPs and Noggin control the timing and pattern of myogenic regulator expression. Genes Dev. 1998 Feb l;12(3):290-303.
Ring, D. B., Johnson, K. W., Henriksen, E. J., Nuss, J. M., Goff, D., Kinnick, T. R., Ma, S. T., Reeder, J. W., Samuels, I., Slabiak, T. et al. (2003). Selective glycogen synthase kinase 3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo. Diabetes 52, 588-95.
Rohwedel, J., Maltsev, V., Bober, E., Arnold, H. FL, Hescheler, J. and Wobus, A. M.
(1994). Muscle cell differentiation of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents. Dev Biol 164, 87-101. - -
Sakurai, H., Inami, Y., Tamamura, Y., Yoshikai, T., Sehara-Fujisawa, A. and Isobe, K. (2009). Bidirectional induction toward paraxial mesodermal derivatives from mouse ES cells in chemically defined medium. Stem Cell Res 3, 157-69.
Sakurai, H., Okawa, Y., Inami, Y., Nishio, N. and Isobe, K. (2008). Paraxial mesodermal progenitors derived from mouse embryonic stem cells contribute to muscle regeneration via differentiation into muscle satellite cells. Stem Cells 26, 1865-73.
Sakurai H, Era T, Jakt LM, Okada M, Nakai S, Nishikawa S, Nishikawa S. In vitro modeling of paraxial and lateral mesoderm differentiation reveals early reversibility. Stem Cells. 2006 Mar;24(3):575-86. Epub 2005 Dec 9.
Sato, N., Meijer, L., Skaltsounis, L., Greengard, P. and Brivanlou, A. H. (2004).
Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat Med 10, 55-63.
Schlessinger, K., Hall, A. and Tolwinski, N. (2009). Wnt signaling pathways meet Rho GTPases. Genes Dev 23, 265-77.
Scott IC, Steiglitz BM, Clark TG, Pappano WN, Greenspan DS. Spatiotemporal expression patterns of mammalian chordin during postgastrulation embryogenesis and in postnatal brain. Dev Dyn. 2000 Apr;217(4):449-56.
Shani, M., Faerman, A., Emerson, C. P., Pearson- White, S., Dekel, I. and Magal, Y. (1992). The consequences of a constitutive expression of MyoDl in ES cells and mouse embryos. Symp Soc Exp Biol 46, 19-36.
Skerjanc, I. S. (1999). Cardiac and skeletal muscle development in P19 embryonal carcinoma cells. Trends Cardiovasc Med 9, 139-43.
Stafford DA, Brunei LJ, Khokha MK, Economides AN, Harland RM. Cooperative activity of noggin and gremlin 1 in axial skeleton development. Development. 2011 Mar;138(5): 1005-14.
Taelman, V. F., Dobrowolski, R., Plouhinec, J. L., Fuentealba, L. C, Vorwald, P. P., Gumper, I., Sabatini, D. D. and De Robertis, E. M. (2010). Wnt signaling requires sequestration of glycogen synthase kinase 3 inside multivesicular endosomes. Cell 143, 1136- 48.
Takahashi, K., Tanabe, K., Ohnuki, M., Narita, M., Ichisaka, T., Tomoda, K. and
Yamanaka, S. (2007). Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861-72.
Takahashi, K. and Yamanaka, S. (2006). Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-76. - -
Takebe A, Era T, Okada M, Martin Jakt L, Kuroda Y, Nishikawa S. Microarray analysis of PDGFR alpha+ populations in ES cell differentiation culture identifies genes involved in differentiation of mesoderm and mesenchyme including ARID3b that is essential for development of embryonic mesenchymal cells. Dev Biol. 2006 May l ;293(l):25-37. Epub 2006 Mar 13.
Tonegawa A, Funayama N, Ueno N, Takahashi Y. Mesodermal subdivision along the medio lateral axis in chicken controlled by different concentrations of BMP-4. Development. 1997 May;124(10): 1975-84.
Wijgerde M, Karp S, McMahon J, McMahon AP. Noggin antagonism of BMP4 signaling controls development of the axial skeleton in the mouse. Dev Biol. 2005 Oct l;286(l): 149-57.
Wittier, L., Shin, E. H., Grote, P., Kispert, A., Beckers, A., Gossler, A., Werber, M. and Herrmann, B. G. (2007). Expression of Msgnl in the presomitic mesoderm is controlled by synergism of WNT signalling and Tbx6. EMBO Rep 8, 784-9.
Wu, D. and Pan, W. (2010). GSK3 : a multifaceted kinase in Wnt signaling. Trends
Biochem Sci 35, 161-8.
Yoon, J. K. and Wold, B. (2000). The bHLH regulator pMesogeninl is required for maturation and segmentation of paraxial mesoderm. Genes Dev 14, 3204-14.
Yu, J., Vodyanik, M. A., Smuga-Otto, K., Antosiewicz-Bourget, J., Frane, J. L., Tian, S., Nie, J., Jonsdottir, G. A., Ruotti, V., Stewart, R. et al. (2007). Induced pluripotent stem cell lines derived from human somatic cells. Science 318, 1917-20.
Yu PB, Hong CC, Sachidanandan C, Babitt JL, Deng DY, Hoyng SA, Lin HY, Bloch KD, Peterson RT. Dorsomorphin inhibits BMP signals required for embryogenesis and iron metabolism. Nat Chem Biol. 2008 Jan;4(l):33-41. Epub 2007 Nov 18.

Claims

CLAIMS:
1. An ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signalling pathway.
2. An ex vivo method for preparing induced paraxial mesoderm progenitor (iPAM) cells, said method comprising the step of culturing pluripotent cells in an appropriate culture medium comprising an effective amount of an activator of the Wnt signaling pathway and an effective amount of an inhibitor of the Bone Morpho genetic Protein (BMP) signaling pathway.
3. An ex vivo method according to claim 1 or 2, wherein said Wnt signaling pathway is the canonical Wnt/beta catenin signaling pathway and/or the Wnt/PCP signaling pathway.
4. An ex vivo method according to one of the claims 1 to 3, wherein the activator of said Wnt signaling pathway is a member of the R-spondin family.
5. An ex vivo method according to the claim 4 wherein the said member of the R-spondin family is selected in the group consisting of R-spondin 3, R-spondin2, or a combination of said R-spondin 3 and R-spondin 2.
6. An ex vivo method according to claim 5, characterized in that said R-spondin-3 is the human R-spondin-3 of sequence SEQ ID NO 1 or the human R-spondin-3 isoform 2 of sequence SEQ ID NO 5.
7. An ex vivo method according to claim 5, characterized in that said R-spondin-2 is the human R-spondin-2 of sequence SEQ ID No 3, the human R-spondin-2 isoform 2 of sequence SEQ ID NO 6, or the human R-spondin-2 isoform 3 of sequence SEQ ID NO 7.
8. An ex vivo method according to claim 2, characterized in that said inhibitor of the BMP signaling pathway is selected from the group consisting of Noggin, Chordin, Chordin-like 1-3, Follistatin, Follistatin-like 1-5, a member of the Dan family and variants and fragments thereof which inhibit the BMP signaling pathway.
9. An ex vivo method according to claim 8, characterized in that said inhibitor of BMP signaling pathway is Noggin.
10. An ex vivo method according to claim 8, characterized in that said inhibitor of the BMP signaling pathway is a chemical inhibitor of BMP signaling such as Dorsomorphin.
11. An ex vivo method according to one of the claims 1 to 2, wherein the activator of said Wnt signaling pathway is an inhibitor of GSK-3B.
12. An ex vivo method according to one of claim 1 to 11 wherein said appropriate culture medium further comprises DMSO, or an equivalent.
13. The method according to one of the claims 1 to 12 wherein the pluripotent stem cells are mouse or human embryonic stem cells or iPS cells.
14. A culture medium which comprises a Wnt activator and an inhibitor of BMP signalling pathway to improve the differentiation of pluripotent cells into induced Paraxial Mesoderm progenitor (iPAM) cells.
15. A culture medium according to claim 14 wherein the Wnt activator is R-spondin3 and the BMP signalling pathway is Noggin.
16. The culture medium according to claim 15 which further comprise DMSO.
17. A population comprising induced paraxial mesoderm progenitor (iPAM) cells obtainable from the method according to claims 1 to 13.
18. A population according to claim 17 characterized in that at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80% and preferably 90% of the cells in said population, exhibit a high expression of biomarker characteristic of paraxial mesoderm progenitor cells, for example, Msgnl gene product.
19. A method for preparing populations comprising skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages, said method comprising the steps of
(a) providing a population comprising induced paraxial mesoderm progenitor (iPAM) cells prepared according to claims 1 to 13; and, culturing said population comprising induced paraxial mesoderm progenitor (iPAM) cells, under appropriate conditions for their differentiation into the desired cell lineages selected among the paraxial mesoderm derivatives which include skeletal muscle, bone, cartilage, dermal cell, adipocytes or endothelial cells lineages.
20. The method according to claim 19, for preparing populations comprising skeletal muscle cell lineages, said method comprising the steps of
(a) providing a population comprising induced paraxial mesoderm progenitor (iPAM) cells prepared according to claims 1 to 13;
(b) culturing said population comprising induced paraxial mesoderm progenitor (iPAM) cells in the presence of a differentiation medium comprising at least the following components:
i. an extracellular matrix material; and,
ii. compounds activating or inhibiting the signaling pathways known to control of the differentiation of said lineages which include but are not restricted to retinoic acid, BMP, TGFB (Transforming Growth FactorB), Hedgehog, Notch, FGF, Wnt, myostatin, insulin, PDGF, VEGF, MAPK, PI3K; and,
(c) optionally, culturing said population obtained from step (b) in a second differentiation medium comprising at least one or more compounds activating or inhibiting the Wnt, FGF, HGF (Hepatocyte growth factor), Activin, EGF (Epidermal growth factor), insulin, and IGF signaling pathways or compounds known to promote myogenic differentiation such as horse serum or transferrin,
thereby obtaining a population comprising skeletal muscle cell lineages, that can be identified by markers such as Desmin, or Myosin Heavy Chain.
21. The method according to claim 19 for preparing a population comprising dermal cell lineages, said method comprising the steps of culturing a population comprising induced paraxial mesoderm progenitor (iPAM) cells prepared according to claims 1 to 13 in the presence of an efficient amount of at least one or more compounds activating or inhibiting the BMP, TGFB, Wnt, FGF, EGF, retinoic acid, Notch and Hedgehog pathways.
22. The method according to claim 19 for preparing a population comprising bone or cartilage cell lineages, comprising the step of culturing a population comprising induced paraxial mesoderm progenitor (iPAM) cells prepared according to claims 1 to 13 in the presence of an efficient amount of at least one or more compounds activating or inhibiting retinoic acid, Wnt, Hedgehog, pTHRP, TGFB, BMP pathways, or compounds known to promote bone or cartilage differentiation such as dexamethasone, ascorbic acid, vitamin D3, and beta-glycerophosphate.
PCT/EP2012/066793 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use WO2013030243A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP12753961.7A EP2756075B1 (en) 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
DK12753961T DK2756075T3 (en) 2011-08-29 2012-08-29 Process for Preparation of Induced Paraxial Mesodermal Progenitor (IPAM) Cells and their Use
ES12753961T ES2761859T3 (en) 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor cells (iPAM) and their use
EP19200815.9A EP3611257A1 (en) 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
JP2014527647A JP6570833B2 (en) 2011-08-29 2012-08-29 Method for preparing artificial paraxial mesoderm progenitor (iPAM) cells and use thereof
CA2847325A CA2847325C (en) 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
US14/342,251 US10240123B2 (en) 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor (IPAM) cells and their use
IL231205A IL231205B (en) 2011-08-29 2014-02-27 Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US201161528348P 2011-08-29 2011-08-29
US61/528,348 2011-08-29
EP11306080.0 2011-08-29
EP11306080A EP2565264A1 (en) 2011-08-29 2011-08-29 Method for preparing induced paraxial mesoderm progenitor (iPAM) cells and their use
US201261654120P 2012-06-01 2012-06-01
EP12305610.3 2012-06-01
US61/654,120 2012-06-01
EP12305610 2012-06-01

Publications (1)

Publication Number Publication Date
WO2013030243A1 true WO2013030243A1 (en) 2013-03-07

Family

ID=47755366

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/066793 WO2013030243A1 (en) 2011-08-29 2012-08-29 Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use

Country Status (7)

Country Link
US (1) US10240123B2 (en)
EP (2) EP2756075B1 (en)
JP (1) JP6570833B2 (en)
CA (1) CA2847325C (en)
DK (1) DK2756075T3 (en)
ES (1) ES2761859T3 (en)
WO (1) WO2013030243A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20150140724A (en) * 2013-04-05 2015-12-16 유니버시티 헬스 네트워크 Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
WO2017072590A1 (en) 2015-10-28 2017-05-04 Crispr Therapeutics Ag Materials and methods for treatment of duchenne muscular dystrophy
WO2019092507A2 (en) 2017-11-09 2019-05-16 Crispr Therapeutics Ag Crispr/cas systems for treatment of dmd
WO2019129768A1 (en) 2017-12-29 2019-07-04 Anagenesis Biotechnologies S.A.S. Method for preparing bap or ba cells
CN111282018A (en) * 2020-05-13 2020-06-16 优赛生命科技有限公司 Adipose-derived stem cell composition for wound repair
WO2020225606A1 (en) 2019-05-08 2020-11-12 Crispr Therapeutics Ag Crispr/cas all-in-two vector systems for treatment of dmd
EP3565887A4 (en) * 2017-01-06 2020-12-02 The Regents of The University of California Methods for generating skeletal muscle progenitor cells
US11390850B2 (en) 2016-06-24 2022-07-19 The Brigham And Women's Hospital, Inc. Generation of brown adipose tissue from pluripotent stem cells in vitro
WO2022201147A1 (en) * 2021-03-22 2022-09-29 Ramot At Tel Aviv University Ltd. Methods for culturing and differentiating pluripotent cells into progenitor or mature muscle cells and composition comprising said cells

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2014268197A1 (en) * 2014-02-11 2015-08-27 Monash University Muscle cell production
EP3259348A4 (en) 2015-02-17 2018-07-18 University Health Network Methods for making and using sinoatrial node-like pacemaker cardiomyocytes and ventricular-like cardiomyocytes
WO2017100498A1 (en) 2015-12-11 2017-06-15 The Johns Hopkins University Isolation of fusion-competent myoblasts and therapeutic applications thereof related to muscular dystrophy
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

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080038820A1 (en) * 2004-06-22 2008-02-14 Rudy-Reil Diane E Induction of pluripotent stem cells into mesodermal lineages

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPWO2004100972A1 (en) * 2003-05-16 2006-07-13 協和醗酵工業株式会社 Preventive and / or therapeutic agent for diseases associated with tissue destruction
WO2007010858A1 (en) * 2005-07-15 2007-01-25 Kyoto University Pluripotent stem cell cloned from single cell derived from skeletal muscle tissue
WO2010008100A1 (en) 2008-07-17 2010-01-21 国立大学法人名古屋大学 Method for induction of skeletal muscle cell or osteocyte
PL3441458T3 (en) * 2009-02-03 2023-11-06 Koninklijke Nederlandse Akademie Van Wetenschappen Culture medium for epithelial stem cells and organoids comprising said stem cells
JP5846558B2 (en) 2009-07-09 2016-01-20 国立大学法人京都大学 Method for inducing differentiation from pluripotent stem cells to skeletal muscle progenitor cells
KR101193541B1 (en) 2009-08-24 2012-10-22 한국과학기술원 Method of inducing differentiation of human embryonic stem cells into mesenchymal stem cells
GB201412554D0 (en) 2014-07-15 2014-08-27 Cambridge Entpr Ltd In vitro mesodermal differentiation
US10787640B2 (en) 2015-03-03 2020-09-29 The Board Of Trustees Of The Leland Stanford Junior University Producing mesodermal cell types and methods of using the same

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080038820A1 (en) * 2004-06-22 2008-02-14 Rudy-Reil Diane E Induction of pluripotent stem cells into mesodermal lineages

Non-Patent Citations (72)

* Cited by examiner, † Cited by third party
Title
BARBERI, T.; BRADBURY, M.; DINCER, Z.; PANAGIOTAKOS, G.; SOCCI, N. D.; STUDER, L.: "Derivation of engraftable skeletal myoblasts from human embryonic stem cells", NAT MED, vol. 13, 2007, pages 642 - 8
CARMON KENDRA S ET AL: "R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, vol. 108, no. 28, July 2011 (2011-07-01), pages 11452 - 11457, XP002685660, ISSN: 0027-8424 *
CARMON, K. S.; GONG, X.; LIN, Q.; THOMAS, A.; LIU, Q.: "R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/ {beta}-catenin signaling", PROC NATL ACAD SCI USA, vol. 108, 2011, pages 11452 - 7
CHAL, J.; POURQUIE, O.: "The Skeletal System", 2009, COLD SPRING HARBOR LABORATORY PRESS, article "Patterning and Differentiation of the Vertebrate Spine", pages: 41 - 116
CHAMBERS, 1.: "The molecular basis of pluripotency in mouse embryonic stem cells", CLONING STEM CELLS, vol. 6, 2004, pages 386 - 91
CHAPMAN, D. L.; AGULNIK, I.; HANCOCK, S.; SILVER, L. M.; PAPAIOANNOU, V. E.: "Tbx6, a mouse T-Box gene implicated in paraxial mesoderm formation at gastrulation", DEV BIOL, vol. 180, 1996, pages 534 - 42
CLEVERS, H.: "Wnt/beta-catenin signaling in development and disease", CELL, vol. 127, 2006, pages 469 - 80
COHEN, P.; GOEDERT, M.: "GSK3 inhibitors: development and therapeutic potential.", NAT REV DRUG DISCOV, vol. 3, 2004, pages 479 - 87
CUNY GD; YU PB; LAHA JK; XING X; LIU JF; LAI CS; DENG DY; SACHIDANANDAN C; BLOCH KD; PETERSON RT: "Structure-activity relationship study of bone morphogenetic protein (BMP) signaling inhibitors", BIOORG MED CHEM LETT., vol. 18, no. 15, 27 June 2008 (2008-06-27), pages 4388 - 92
DARABI, R.; GEHLBACH, K.; BACHOO, R. M.; KAMATH, S.; OSAWA, M.; KAMM, K. E.; KYBA, M.; PERLINGEIRO, R. C.: "Functional skeletal muscle regeneration from differentiating embryonic stem cells", NAT MED, vol. 14, 2008, pages 134 - 43
DARABI, R.; SANTOS, F. N.; FILARETO, A.; PAN, W.; KOENE, R.; RUDNICKI, M. A.; KYBA, M.; PERLINGEIRO, R. C.: "Assessment of the Myogenic Stem Cell Compartment Following Transplantation of Pax3/Pax7-Induced Embryonic Stem Cell-Derived Progenitors", STEM CELLS, 2011
DE LAU, W.; BARKER, N.; LOW, T. Y.; KOO, B. K.; LI, V. S.; TEUNISSEN, H.; KUJALA, P.; HAEGEBARTH, A.; PETERS, P. J.; VAN DE WETERI: "Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling", NATURE, 2011
DEKEL, I.; MAGAL, Y.; PEARSON-WHITE, S.; EMERSON, C. P.; SHANI, M.: "Conditional conversion of ES cells to skeletal muscle by an exogenous MyoDl gene", NEW BIOL, vol. 4, 1992, pages 217 - 24
DERYNCK RIK: "The TGF-0 Family", 2008, CSHL
DIMOS, J. T.; RODOLFA, K. T.; NIAKAN, K. K.; WEISENTHAL, L. M.; MITSUMOTO, H.; CHUNG, W.; CROFT, G. F.; SAPHIER, G.; LEIBEL, R.; G: "Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons", SCIENCE, vol. 321, 2008, pages 1218 - 21
DINSMORE, J.; RATLIFF, J.; DEACON, T.; PAKZABAN, P.; JACOBY, D.; GALPERN, W.; ISACSON, O.: "Embryonic stem cells differentiated in vitro as a novel source of cells for transplantation", CELL TRANSPLANT, vol. 5, 1996, pages 131 - 43
E. MEYERS; W. MILLER, COMPUT. APPL. BIOSCI., vol. 4, 1988, pages 11 - 17
FUKADA, S.; HIGUCHI, S.; SEGAWA, M.; KODA, K.; YAMAMOTO, Y.; TSUJIKAWA, K.; KOHAMA, Y.; UEZUMI, A.; IMAMURA, M.; MIYAGOE-SUZUKI, Y: "Purification and cell-surface marker characterization of quiescent satellite cells from murine skeletal muscle by a novel monoclonal antibody", EXP CELL RES, vol. 296, 2004, pages 245 - 55
GAYRAUD-MOREL B. ET AL., J CELL SCI, vol. 125, 1 April 2012 (2012-04-01), pages 1738
GAYRAUD-MOREL B.; F. CHRETIEN; S. TAJBAKHSH, REGEN MED, vol. 4, March 2009 (2009-03-01), pages 293
HAN, X. H.; JIN, Y. R.; SETO, M.; YOON, J. K.: "A WNT/beta-catenin signaling activator, R-spondin, plays positive regulatory roles during skeletal myogenesis", J BIOL CHEM, vol. 286, 2011, pages 10649 - 59
HANKENSON, K. D.; SWEETWYNE, M. T.; SHITAYE, H.; POSEY, K. L.: "Thrombospondins and novel TSR-containing proteins, R-spondins, regulate bone formation and remodeling", CURR OSTEOPOROS REP, vol. 8, 2010, pages 68 - 76
HIDETOSHI SAKURAI ET AL: "In Vitro Modeling of Paraxial and Lateral Mesoderm Differentiation Reveals Early Reversibility", STEM CELLS, vol. 24, no. 3, 1 March 2006 (2006-03-01), pages 575 - 586, XP055018939, ISSN: 1066-5099, DOI: 10.1634/stemcells.2005-0256 *
HIDETOSHI SAKURAI ET AL: "Paraxial Mesodermal Progenitors Derived from Mouse Embryonic Stem Cells Contribute to Muscle Regeneration via Differentiation into Muscle Satellite Cells", STEM CELLS, vol. 26, no. 7, 1 July 2008 (2008-07-01), pages 1865 - 1873, XP055018969, ISSN: 1066-5099, DOI: 10.1634/stemcells.2008-0173 *
HIRSINGER, E.; JOUVE, C.; DUBRULLE, J.; POURQUIE, O.: "Somite formation and patterning", INT REV CYTOL, vol. 198, 2000, pages 1 - 65
HOLLNAGEL A; OEHLMANN V; HEYMER J; RUTHER U; NORDHEIM A: "Id genes are direct targets of bone morphogenetic protein induction in embryonic stem cells", J BIOL CHEM., vol. 274, no. 28, 9 July 1999 (1999-07-09), pages 19838 - 45
JIN, Y. R.; TURCOTTE, T. J.; CROCKER, A. L.; HAN, X. H.; YOON, J. K.: "The canonical Wnt signaling activator, R-spondin2, regulates craniofacial patterning and morphogenesis within the branchial arch through ectodermal-mesenchymal interaction", DEV BIOL, vol. 352, 2011, pages 1 - 13
KAZANSKAYA, 0.; GLINKA, A.; DEL BARCO BARRANTES, I.; STANNEK, P.; NIEHRS, C.; WU, W.: "R-Spondin2 is a secreted activator of Wnt/beta-catenin signaling and is required for Xenopus myogenesis", DEV CELL, vol. 7, 2004, pages 525 - 34
KAZANSKAYA, 0.; OHKAWARA, B.; HEROULT, M.; WU, W.; MALTRY, N.; AUGUSTIN, H. G.; NIEHRS, C.: "The Wnt signaling regulator R-spondin 3 promotes angioblast and vascular development", DEVELOPMENT, vol. 135, 2008, pages 3655 - 64
KENNEDY, K. A.; PORTER, T.; MEHTA, V.; RYAN, S. D.; PRICE, F.; PESHDARY, V.; KARAMBOULAS, C.; SAVAGE, J.; DRYSDALE, T. A.; LI, S.: "Retinoic acid enhances skeletal muscle progenitor formation and bypasses inhibition by bone morphogenetic protein 4 but not dominant negative beta-catenin", BMC BIOL, vol. 7, 2009, pages 67
KIM, K. A.; WAGLE, M.; TRAN, K.; ZHAN, X.; DIXON, M. A.; LIU, S.; GROS, D.; KORVER, W.; YONKOVICH, S.; TOMASEVIC, N. ET AL.: "R-Spondin family members regulate the Wnt pathway by a common mechanism", MOL BIOL CELL, vol. 19, 2008, pages 2588 - 96
KROL AJ; ROELLIG D; DEQUEANT ML; TASSY 0; GLYNN E; HATTEM G; MUSHEGIAN A; OATES AC; POURQUIE O: "Evolutionary plasticity of segmentation clock networks", LOPMENT, vol. 138, no. 13, July 2011 (2011-07-01), pages 2783 - 92
LARS WITTLER ET AL: "Expression of Msgn1 in the presomitic mesoderm is controlled by synergism of WNT signalling and Tbx6", EMBO REPORTS, vol. 8, no. 8, 1 August 2007 (2007-08-01), pages 784 - 789, XP055018779, ISSN: 1469-221X, DOI: 10.1038/sj.embor.7401030 *
LOSER, P.; SCHIRM, J.; GUHR, A.; WOBUS, A. M.; KURTZ, A.: "Human embryonic stem cell lines and their use in international research", STEM CELLS, vol. 28, 2010, pages 240 - 6
MCBURNEY, M. W.; JONES-VILLENEUVE, E. M.; EDWARDS, M. K.; ANDERSON, P. J.: "Control of muscle and neuronal differentiation in a cultured embryonal carcinoma cell line", NATURE, vol. 299, 1982, pages 165 - 7
MCMAHON JA; TAKADA S; ZIMMERMAN LB; FAN CM; HARLAND RM; MCMAHON AP: "Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite", GENES DEV., vol. 12, no. 10, 15 May 1998 (1998-05-15), pages 1438 - 52
METALLO, C. M.; MOHR, J. C.; DETZEL, C. J.; DE PABLO, J. J.; VAN WIE, B. J.; PALECEK, S. P.: "Engineering the stem cell microenvironment", BIOTECHNOL PROG, vol. 23, 2007, pages 18 - 23
MIZUNO, Y.; CHANG, H.; UMEDA, K.; NIWA, A.; IWASA, T.; AWAYA, T.; FUKADA, S.; YAMAMOTO, H.; YAMANAKA, S.; NAKAHATA, T. ET AL.: "Generation of skeletal muscle stem/progenitor cells from murine induced pluripotent stem cells", FASEB J, vol. 24, 2010, pages 2245 - 53
MONTCOUQUIOL, M.; CRENSHAW, E. B., 3RD; KELLEY, M. W.: "Noncanonical Wnt signaling and neural polarity", ANNU REV NEUROSCI, vol. 29, 2006, pages 363 - 86
NAM, J. S.; TURCOTTE, T. J.; SMITH, P. F.; CHOI, S.; YOON, J. K.: "Mouse cristin/R-spondin family proteins are novel ligands for the Frizzled 8 and LRP6 receptors and activate beta-catenin-dependent gene expression", J BIOL CHEM, vol. 281, 2006, pages 13247 - 57
NAM, J. S.; TURCOTTE, T. J.; YOON, J. K.: "Dynamic expression of R-spondin family genes in mouse development", GENE EXPR PATTERNS, vol. 7, 2007, pages 306 - 12
OHKAWARA, B.; GLINKA, A.; NIEHRS, C.: "Rspo3 binds syndecan 4 and induces Wnt/PCP signaling via clathrin-mediated endocytosis to promote morphogenesis", DEV CELL, vol. 20, 2011, pages 303 - 14
PARK, 1. H.; ARORA, N.; HUO, H.; MAHERALI, N.; AHFELDT, T.; SHIMAMURA, A.; LENSCH, M. W.; COWAN, C.; HOCHEDLINGER, K.; DALEY, G. Q: "Disease-specific induced pluripotent stem cells", CELL, vol. 134, 2008, pages 877 - 86
PARK, 1. H.; ZHAO, R.; WEST, J. A.; YABUUCHI, A.; HUO, H.; INCE, T. A.; LEROU, P. H.; LENSCH, M. W.; DALEY, G. Q.: "Reprogramming of human somatic cells to pluripotencywith defined factors", NATURE, vol. 451, 2008, pages 141 - 6
POURQUIE 0; FAN CM; COLTEY M; HIRSINGER E; WATANABE Y; BREANT C; FRANCIS-WEST P; BRICKELL P; TESSIER-LAVIGNE M; LE DOUARIN NM: "Lateral and axial signals involved in avian somite patterning: a role for BMP4", CELL, vol. 84, no. 3, 9 February 1996 (1996-02-09), pages 461 - 71
PRELLE, K.; WOBUS, A. M.; KREBS, 0.; BLUM, W. F.; WOLF, E.: "Overexpression of insulin-like growth factor-11 in mouse embryonic stem cells promotes myogenic differentiation", BIOCHEM BIOPHYS RES COMMUN, vol. 277, 2000, pages 631 - 8
RESHEF R; MAROTO M; LASSAR AB: "Regulation of dorsal somitic cell fates: BMPs and Noggin control the timing and pattern of myogenic regulator expression", GENES DEV., vol. 12, no. 3, 1 February 1998 (1998-02-01), pages 290 - 303
RING, D. B.; JOHNSON, K. W.; HENRIKSEN, E. J.; NUSS, J. M.; GOFF, D.; KINNICK, T. R.; MA, S. T.; REEDER, J. W.; SAMUELS, I.; SLABI: "Selective glycogen synthase kinase 3 inhibitors potentiate insulin activation of glucose transport and utilization in vitro and in vivo", DIABETES, vol. 52, 2003, pages 588 - 95
ROHWEDEL, J.; MALTSEV, V.; BOBER, E.; ARNOLD, H. H.; HESCHELER, J.; WOBUS, A. M.: "Muscle cell differentiation of embryonic stem cells reflects myogenesis in vivo: developmentally regulated expression of myogenic determination genes and functional expression of ionic currents", DEV BIOL, vol. 164, 1994, pages 87 - 101
S.J.MATHEW, DEV, vol. 138, 2011, pages 371
SAKURAI H; ERA T; JAKT LM; OKADA M; NAKAI S; NISHIKAWA S; NISHIKAWA S.: "In vitro modeling of paraxial and lateral mesoderm differentiation reveals early reversibility", STEM CELLS, vol. 24, no. 3, 9 December 2005 (2005-12-09), pages 575 - 86
SAKURAI, H.; INAMI, Y.; TAMAMURA, Y.; YOSHIKAI, T.; SEHARA-FUJISAWA, A.; ISOBE, K.: "Bidirectional induction toward paraxial mesodermal derivatives from mouse ES cells in chemically defined medium", STEM CELL RES, vol. 3, 2009, pages 157 - 69
SAKURAI, H.; OKAWA, Y.; INAMI, Y.; NISHIO, N.; ISOBE, K.: "Paraxial mesodermal progenitors derived from mouse embryonic stem cells contribute to muscle regeneration via differentiation into muscle satellite cells", STEM CELLS, vol. 26, 2008, pages 1865 - 73
SATO, N.; MEIJER, L.; SKALTSOUNIS, L.; GREENGARD, P.; BRIVANLOU, A. H.: "Maintenance of pluripotency in human and mouse embryonic stem cells through activation of Wnt signaling by a pharmacological GSK-3-specific inhibitor", NAT MED, vol. 10, 2004, pages 55 - 63
SCHLESSINGER, K.; HALL, A.; TOLWINSKI, N.: "Wnt signaling pathways meet Rho GTPases", GENES DEV, vol. 23, 2009, pages 265 - 77
SCHRÖDER, ARNOLD GÜNTER: "Differenzierung embryonaler Stammzellen der Maus zu paraxialem Mesoderm in vitro", 6 June 2011 (2011-06-06), pages Frontpg. - 105, XP002685659, Retrieved from the Internet <URL:http://www.diss.fu-berlin.de/diss/receive/FUDISS_thesis_000000023130> [retrieved on 20121018] *
SCOTT IC; STEIGLITZ BM; CLARK TG; PAPPANO WN; GREENSPAN DS: "Spatiotemporal expression patterns of mammalian chordin during postgastrulation embryogenesis and in postnatal brain", DEV DYN., vol. 217, no. 4, April 2000 (2000-04-01), pages 449 - 56
SHANI, M.; FAERMAN, A.; EMERSON, C. P.; PEARSON-WHITE, S.; DEKEL, 1.; MAGAL, Y.: "The consequences of a constitutive expression of MyoD1 in ES cells and mouse embryos", SYMP SOC EXP BIOL, vol. 46, 1992, pages 19 - 36
SKERJANC, I. S.: "Cardiac and skeletal muscle development in P19 embryonal carcinoma cells", TRENDS CARDIOVASC MED, vol. 9, 1999, pages 139 - 43
STAFFORD DA; BRUNET LJ; KHOKHA MK; ECONOMIDES AN; HARLAND RM: "Cooperative activity of noggin and gremlin 1 in axial skeleton development", DEVELOPMENT, vol. 138, no. 5, March 2011 (2011-03-01), pages 1005 - 14
TAELMAN, V. F.; DOBROWOLSKI, R.; PLOUHINEC, J. L.; FUENTEALBA, L. C.; VORWALD, P. P.; GUMPER, I.; SABATINI, D. D.; DE ROBERTIS, E.: "Wnt signaling requires sequestration of glycogen synthase kinase 3 inside multivesicular endosomes", CELL, vol. 143, 2010, pages 1136 - 48
TAKAHASHI, K.; 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, 2007, pages 861 - 72
TAKAHASHI, K.; YAMANAKA, S.: "Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors", CELL, vol. 126, 2006, pages 663 - 76
TAKEBE A; ERA T; OKADA M; MARTIN JAKT L; KURODA Y; NISHIKAWA S: "Microarray analysis of PDGFR alpha+ populations in ES cell differentiation culture identifies genes involved in differentiation of mesoderm and mesenchyme including ARID3b that is essential for development of embryonic mesenchymal cells", DEV BIOL., vol. 293, no. 1, 1 May 2006 (2006-05-01), pages 25 - 37
TONEGAWA A; FUNAYAMA N; UENO N; TAKAHASHI Y: "Mesodermal subdivision along the mediolateral axis in chicken controlled by different concentrations of BMP-4", DEVELOPMENT, vol. 124, no. 10, May 1997 (1997-05-01), pages 1975 - 84
WIJGERDE M; KARP S; MCMAHON J; MCMAHON AP: "Noggin antagonism of BMP4 signaling controls development of the axial skeleton in the mouse", DEV BIOL., vol. 286, no. 1, 1 October 2005 (2005-10-01), pages 149 - 57
WITTLER, L.; SHIN, E. H.; GROTE, P.; KISPERT, A.; BECKERS, A.; GOSSLER, A.; WERBER, M.; HERRMANN, B. G.: "Expression of Msgnl in the presomitic mesoderm is controlled by synergism of WNT signalling and Tbx6", EMBO REP, vol. 8, 2007, pages 784 - 9
WU, D.; PAN, W.: "GSK3: a multifaceted kinase in Wnt signaling", TRENDS BIOCHEM SCI, vol. 35, 2010, pages 161 - 8
YOON, J. K.; WOLD, B.: "The bHLH regulator pMesogenin1 is required for maturation and segmentation of paraxial mesoderm", GENES DEV, vol. 14, 2000, pages 3204 - 14
YU ET AL., NAT CHEM BIO1., 2008
YU PB; HONG CC; SACHIDANANDAN C; BABITT JL; DENG DY; HOYNG SA; LIN HY; BLOCH KD; PETERSON RT: "Dorsomorphin inhibits BMP signals required for embryogenesis and iron metabolism", NAT CHEM BIOL., vol. 4, no. 1, 18 November 2007 (2007-11-18), pages 33 - 41
YU, J.; VODYANIK, M. A.; SMUGA-OTTO, K.; ANTOSIEWICZ-BOURGET, J.; FRANE, J. L.; TIAN, S.; NIE, J.; JONSDOTTIR, G. A.; RUOTTI, V.;: "Induced pluripotent stem cell lines derived from human somatic cells", SCIENCE, vol. 318, 2007, pages 1917 - 20

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102393716B1 (en) 2013-04-05 2022-05-02 유니버시티 헬스 네트워크 Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
JP2016515825A (en) * 2013-04-05 2016-06-02 ユニバーシティー ヘルス ネットワーク Methods and compositions for producing chondrocyte lineage cells and / or cartilage-like tissue
US9993504B2 (en) 2013-04-05 2018-06-12 University Health Network Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
KR20150140724A (en) * 2013-04-05 2015-12-16 유니버시티 헬스 네트워크 Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
US10736923B2 (en) 2013-04-05 2020-08-11 University Health Network Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
KR102393715B1 (en) 2013-04-05 2022-05-02 유니버시티 헬스 네트워크 Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
KR20210138802A (en) * 2013-04-05 2021-11-19 유니버시티 헬스 네트워크 Methods and compositions for generating chondrocyte lineage cells and/or cartilage like tissue
WO2017072590A1 (en) 2015-10-28 2017-05-04 Crispr Therapeutics Ag Materials and methods for treatment of duchenne muscular dystrophy
EP4279084A1 (en) 2015-10-28 2023-11-22 Vertex Pharmaceuticals Inc. Materials and methods for treatment of duchenne muscular dystrophy
US11390850B2 (en) 2016-06-24 2022-07-19 The Brigham And Women's Hospital, Inc. Generation of brown adipose tissue from pluripotent stem cells in vitro
EP3565887A4 (en) * 2017-01-06 2020-12-02 The Regents of The University of California Methods for generating skeletal muscle progenitor cells
WO2019092507A2 (en) 2017-11-09 2019-05-16 Crispr Therapeutics Ag Crispr/cas systems for treatment of dmd
WO2019129768A1 (en) 2017-12-29 2019-07-04 Anagenesis Biotechnologies S.A.S. Method for preparing bap or ba cells
WO2020225606A1 (en) 2019-05-08 2020-11-12 Crispr Therapeutics Ag Crispr/cas all-in-two vector systems for treatment of dmd
CN111282018A (en) * 2020-05-13 2020-06-16 优赛生命科技有限公司 Adipose-derived stem cell composition for wound repair
WO2022201147A1 (en) * 2021-03-22 2022-09-29 Ramot At Tel Aviv University Ltd. Methods for culturing and differentiating pluripotent cells into progenitor or mature muscle cells and composition comprising said cells

Also Published As

Publication number Publication date
US20140363886A1 (en) 2014-12-11
ES2761859T3 (en) 2020-05-21
EP2756075B1 (en) 2019-10-02
CA2847325C (en) 2020-01-14
EP3611257A1 (en) 2020-02-19
DK2756075T3 (en) 2019-12-09
JP2014525253A (en) 2014-09-29
JP6570833B2 (en) 2019-09-04
EP2756075A1 (en) 2014-07-23
US10240123B2 (en) 2019-03-26
CA2847325A1 (en) 2013-03-07

Similar Documents

Publication Publication Date Title
EP2756075B1 (en) Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
US20130052729A1 (en) Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
JP7464652B2 (en) Colon organoids and methods for producing and using same
Bajpai et al. Functional vascular smooth muscle cells derived from human induced pluripotent stem cells via mesenchymal stem cell intermediates
DK2898063T3 (en) PANCREATIC DIFFERENTIALIZATION OF PLURIPOTENT PATTERN CELLS IN VITRO
EP2596096B1 (en) Method for inducing differentiation of human pluripotent stem cell into intermediate mesoderm cell
JP2022188229A (en) Method for inducing differentiation from intermediate mesoderm cells to renal progenitor cells, and method for inducing differentiation from pluripotent stem cells to renal progenitor cells
US10767162B2 (en) Methods for differentiation
Kidwai et al. Lineage-specific differentiation of osteogenic progenitors from pluripotent stem cells reveals the FGF1-RUNX2 association in neural crest-derived osteoprogenitors
Li et al. Ectodermal progenitors derived from epiblast stem cells by inhibition of Nodal signaling
WO2023127824A1 (en) Neural crest cell culturing method and production method
CN113166219A (en) Stem cell derived human microglia, methods of making, and methods of use
CA2908225A1 (en) Method for culturing hepatoblast-like cells and culture product thereof
Mitsumoto et al. Improved methods for the differentiation of hypothalamic vasopressin neurons using mouse induced pluripotent stem cells
US20200362309A1 (en) Method for preparing BAP or BA cells
EP2565264A1 (en) Method for preparing induced paraxial mesoderm progenitor (iPAM) cells and their use
JP2019004903A (en) Method for preparing induced paraxial mesoderm progenitor (ipam) cells and their use
Wei et al. Lateral Mesoderm-derived mesenchymal stem cells with robust osteochondrogenic potential and hematopoiesis-supporting ability
WO2016009196A1 (en) In vitro mesodermal differentiation
US11857697B2 (en) Compositions and methods for obtaining 3-dimensional lung-like epithelium and related uses thereof
EP4342980A1 (en) Method for producing brain capillary endothelial-like cells, and use therefor
Kuboth et al. Chondrogenic differentiation in vitro of murine two-factor induced pluripotent stem cells is comparable to murine embryonic stem cells
Pedroso Definitive Endoderm Induction of Human Induced Pluripotent Stem Cells
Karmach Epigenetic and Environmental Regulators of Early Bone Development
Lin Investigating the role of Neuronatin in neural development

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: 12753961

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2014527647

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 231205

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2847325

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 14342251

Country of ref document: US