Elsevier

Experimental Neurology

Volume 323, January 2020, 113086
Experimental Neurology

Review Article
Stem cell-based therapies for Duchenne muscular dystrophy

https://doi.org/10.1016/j.expneurol.2019.113086Get rights and content

Highlights

  • Myogenic progenitor cells could provide a source for cell therapy for DMD to regenerate and replace the diseased tissue

  • An orchestration of signaling molecules directing the lineage determination sets the basis for PSC differentiation in vitro

  • There is a need to develop more cell therapy options (e.g iPSC) for DMD due to limited success of current ones

  • The review describes the most recent studies of hiPSC muscle lineage specification and the potential of their application

Abstract

Muscular dystrophies are a group of genetic muscle disorders that cause progressive muscle weakness and degeneration. Within this group, Duchenne muscular dystrophy (DMD) is the most common and one of the most severe. DMD is an X chromosome linked disease that occurs to 1 in 3500 to 1 in 5000 boys. The cause of DMD is a mutation in the dystrophin gene, whose encoded protein provides both structural support and cell signaling capabilities. So far, there are very limited therapeutic options available and there is no cure for this disease. In this review, we discuss the existing cell therapy research, especially stem cell-based, which utilize myoblasts, satellite cells, bone marrow cells, mesoangioblasts and CD133+ cells. Finally, we focus on human pluripotent stem cells (hPSCs) which hold great potential in treating DMD. hPSCs can be used for autologous transplantation after being specified to a myogenic lineage. Over the last few years, there has been a rapid development of isolation, as well as differentiation, techniques in order to achieve effective transplantation results of myogenic cells specified from hPSCs. In this review, we summarize the current methods of hPSCs myogenic commitment/differentiation, and describe the current status of hPSC-derived myogenic cell transplantation.

Section snippets

Pathological features of DMD

DMD is due to mutations in the dystrophin gene. The dystrophin gene is translated into a 427 kDa protein, which is part of the Dystrophin Glycoprotein Complex (DGC) that provides a structural and signaling link between the cytoskeleton of the muscle fiber and the extracellular matrix (Ervasti, 2007). In healthy individuals, the dystrophin protein stabilizes the plasma membrane of the striated muscle fibers. However, in patients with DMD or the allelic Becker muscular dystrophy (BMD), mutations

Skeletal muscle determination in the vertebrate embryo

Muscle commitment and differentiation are mainly controlled by the regulated spatio-temporal expression of a set of four proteins (Myf5, MyoD1, Myogenin, and Mrf4) termed the muscle regulatory factors (MRFs) (Buckingham and Rigby, 2014). The MRFs are transcription factors that drive the expression of a multitude of genes regulating establishment and maintenance of the myogenic fate.

In embryonic development, myogenic cells originate from mesodermal precursors that colonize the paraxial mesoderm

Cell therapy for DMD

Cell therapy is based on the heterologous, or autologous, transplantation of cells, with the goal of regenerating the damaged tissue or organ of the patient, and replenishing specific stem cell populations. In the case of DMD, the main goal is to reconstitute the satellite cell pool with dystrophin competent cells, and thereby restore muscle function due to the presence of dystrophin expressing muscle fibers. The source of the therapeutic cells can be healthy, histo-compatible donors, or

Pluripotent stem cells

Vertebrate pluripotent stem cells (PSCs) retain their ability to differentiate into the three germ layers of the embryo: ectoderm, mesoderm, and endoderm. Typical PSCs are the embryonic stem cells (ESCs) (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998), and the induced PSC (iPSCs) (Takahashi et al., 2007; Takahashi and Yamanaka, 2006).

The generation of iPSCs opened up new avenues in stem cell therapy, and solved many problems associated with use of ESCs. For example, while human

Muscle linage specification systems

One of the strategies to achieve a direct myogenic specification of PSCs is to replicate in the culture dish the inductive stimuli which underlie the muscle determination in the developing embryos. To accomplish this goal, one approach is for monolayer PSCs to be treated in vitro with the specific cytokines and growth/morphogenetic factors that orchestrate the specification of the mesoderm in vivo, the somitogenesis, and the commitment of the early muscle progenitors (Chal et al., 2015). A

Genetic engineering of hiPSCs to restore functional dystrophin expression

In order to generate dystrophin expressing muscle fibers, hiPSCs derived from DMD patients can be genetically corrected to express functional dystrophin for autologous cell replacement therapy. CRISPR-Cas9 mediated gene editing is currently being investigated as a tool to perform such correction. It involves two components: a single guide RNA (sgRNA) and the Cas9 endonuclease. Cas9 endonuclease associates with the sgRNA at the genomic target sequence to create DNA double strand breaks leading

Potential limitations of using hiPSCs for DMD treatment

The use of hiPSCs for treatment of muscular dystrophies is very promising. However, before proceeding to clinical trials, four key limitations must be overcome, and potential safety issues addressed. (i) We have to identify the patients' best somatic cell type from which to generate the hiPSCs, and we have to improve the hiPSCs muscle commitment protocols, for example by generating the myogenic cells more quickly in vitro, and by using culture media free of animal factors. (ii) We have to

Future directions of the hiPSCs therapy development for the MDs

To develop clinically applicable hiPSCs-based therapies, researchers have focused on deriving cells that have high potency in terms of regenerating and self-renew, i.e. cells that have similar features to those of adult satellite cells (Incitti et al., 2019) (Fig. 1). Based on this, progenitor cells can acquire a higher clinical potential. As discussed earlier, hiPSCs-derived myogenic progenitors have a molecular profile that is similar to fetal-stage myoblasts. Therefore, one of the most

Conclusion

Stem cells, due to their advantageous regeneration capability, bring the promise for cell transplantation therapy (Fig. 1 summary of cells that can be used or tested for stem cell-based therapies for muscular dystrophies). hiPSCs that can be derived from patients open the avenue for autologous cell therapy. With the rapid development of serum-free lineage specification protocols, expandable myogenic progenitor cells can be differentiated from hiPSCs. This population of cells has similar

Funding

This work was supported by National Institutes of Health R01AR070751 (G.L.), the Maryland Stem Cell Research Fund (MSCRF; G.L. and C.S.), and the Muscular Dystrophy Association (MDA: G.L.)

References (158)

  • R.L. Davis et al.

    Expression of a single transfected cDNA converts fibroblasts to myoblasts

    Cell

    (1987)
  • C. Dell'Agnola et al.

    Hematopoietic stem cell transplantation does not restore dystrophin expression in Duchenne muscular dystrophy dogs

    Blood

    (2004)
  • A.E. Emery

    The muscular dystrophies

    Lancet

    (2002)
  • J.M. Ervasti

    Dystrophin, its interactions with other proteins, and implications for muscular dystrophy

    Biochim. Biophys. Acta

    (2007)
  • S.M. Garcia et al.

    High-yield purification, preservation, and serial transplantation of human satellite cells

    Stem Cell Rep.

    (2018)
  • C. Garcia-Doval et al.

    Molecular architectures and mechanisms of class 2 CRISPR-associated nucleases

    Curr. Opin. Struct. Biol.

    (2017)
  • M.F.M. Gerli et al.

    Combined Notch and PDGF signaling enhances migration and expression of stem cell markers while inducing perivascular cell features in muscle satellite cells

    Stem Cell Rep.

    (2019)
  • S. Goudenege et al.

    Myoblasts derived from normal hESCs and dystrophic hiPSCs efficiently fuse with existing muscle fibers following transplantation

    Mol. Ther.

    (2012)
  • S. Gunther et al.

    Myf5-positive satellite cells contribute to Pax7-dependent long-term maintenance of adult muscle stem cells

    Cell Stem Cell

    (2013)
  • A. Hamade et al.

    Retinoic acid activates myogenesis in vivo through Fgf8 signalling

    Dev. Biol.

    (2006)
  • J.E. Heredia et al.

    Type 2 innate signals stimulate fibro/adipogenic progenitors to facilitate muscle regeneration

    Cell

    (2013)
  • D. Kim et al.

    Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins

    Cell Stem Cell

    (2009)
  • J. Kim et al.

    Expansion and purification are critical for the therapeutic application of pluripotent stem cell-derived myogenic progenitors

    Stem Cell Rep.

    (2017)
  • S. Kuang et al.

    Asymmetric self-renewal and commitment of satellite stem cells in muscle

    Cell

    (2007)
  • H.L. Li et al.

    Precise correction of the dystrophin gene in duchenne muscular dystrophy patient induced pluripotent stem cells by TALEN and CRISPR-Cas9

    Stem Cell Rep.

    (2015)
  • K.M. Loh et al.

    Mapping the pairwise choices leading from Pluripotency to human bone, heart, and other mesoderm cell types

    Cell

    (2016)
  • S.M. Maffioletti et al.

    Three-dimensional human iPSC-derived artificial skeletal muscles model muscular dystrophies and enable multilineage tissue engineering

    Cell Rep.

    (2018)
  • A. Magli et al.

    PAX7 targets, CD54, integrin alpha9beta1, and SDC2, allow isolation of human ESC/iPSC-derived myogenic progenitors

    Cell Rep.

    (2017)
  • S. Mendjan et al.

    NANOG and CDX2 pattern distinct subtypes of human mesoderm during exit from pluripotency

    Cell Stem Cell

    (2014)
  • J. Meng et al.

    Human skeletal muscle-derived CD133(+) cells form functional satellite cells after intramuscular transplantation in immunodeficient host mice

    Mol. Ther.

    (2014)
  • L. Morandi et al.

    Lack of mRNA and dystrophin expression in DMD patients three months after myoblast transfer

    Neuromuscul. Disord.

    (1995)
  • E. Negroni et al.

    In vivo myogenic potential of human CD133+ muscle-derived stem cells: a quantitative study

    Mol. Ther.

    (2009)
  • R. Abujarour et al.

    Myogenic differentiation of muscular dystrophy-specific induced pluripotent stem cells for use in drug discovery

    Stem Cells Transl. Med.

    (2014)
  • K. Alvarez et al.

    Augmented synthesis and differential localization of heparan sulfate proteoglycans in Duchenne muscular dystrophy

    J. Cell. Biochem.

    (2002)
  • L. Amoasii et al.

    Gene editing restores dystrophin expression in a canine model of Duchenne muscular dystrophy

    Science

    (2018)
  • A. Aulehla et al.

    A beta-catenin gradient links the clock and wavefront systems in mouse embryo segmentation

    Nat. Cell Biol.

    (2008)
  • T. Barberi et al.

    Derivation of multipotent mesenchymal precursors from human embryonic stem cells

    PLoS Med.

    (2005)
  • T. Barberi et al.

    Derivation of engraftable skeletal myoblasts from human embryonic stem cells

    Nat. Med.

    (2007)
  • N.E. Bengtsson et al.

    Corrigendum: muscle-specific CRISPR/Cas9 dystrophin gene editing ameliorates pathophysiology in a mouse model for Duchenne muscular dystrophy

    Nat. Commun.

    (2017)
  • J. Chal et al.

    Differentiation of pluripotent stem cells to muscle fiber to model Duchenne muscular dystrophy

    Nat. Biotechnol.

    (2015)
  • J. Chal et al.

    Generation of human muscle fibers and satellite-like cells from human pluripotent stem cells in vitro

    Nat. Protoc.

    (2016)
  • R.B. Chalamalasetty et al.

    The Wnt3a/beta-catenin target gene Mesogenin1 controls the segmentation clock by activating a Notch signalling program

    Nat. Commun.

    (2011)
  • R.B. Chalamalasetty et al.

    Mesogenin 1 is a master regulator of paraxial presomitic mesoderm differentiation

    Development

    (2014)
  • C.N. Chang et al.

    Location, location, location: signals in muscle specification

    J Dev Biol

    (2018)
  • S.B. Charge et al.

    Cellular and molecular regulation of muscle regeneration

    Physiol. Rev.

    (2004)
  • G. Cossu et al.

    Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy

    EMBO Mol. Med.

    (2015)
  • T.J. Cunningham et al.

    Retinoic acid activity in undifferentiated neural progenitors is sufficient to fulfill its role in restricting Fgf8 expression for somitogenesis

    PLoS One

    (2015)
  • R. Darabi et al.

    Functional skeletal muscle regeneration from differentiating embryonic stem cells

    Nat. Med.

    (2008)
  • R. Darabi et al.

    Assessment of the myogenic stem cell compartment following transplantation of Pax3/Pax7-induced embryonic stem cell-derived progenitors

    Stem Cells

    (2011)
  • L. De Angelis et al.

    Skeletal myogenic progenitors originating from embryonic dorsal aorta coexpress endothelial and myogenic markers and contribute to postnatal muscle growth and regeneration

    J. Cell Biol.

    (1999)
  • Cited by (0)

    View full text