1
|
Price FD, Matyas MN, Gehrke AR, Chen W, Wolin EA, Holton KM, Gibbs RM, Lee A, Singu PS, Sakakeeny JS, Poteracki JM, Goune K, Pfeiffer IT, Boswell SA, Sorger PK, Srivastava M, Pfaff KL, Gussoni E, Buchanan SM, Rubin LL. Organoid culture promotes dedifferentiation of mouse myoblasts into stem cells capable of complete muscle regeneration. Nat Biotechnol 2024:10.1038/s41587-024-02344-7. [PMID: 39261590 DOI: 10.1038/s41587-024-02344-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/27/2024] [Indexed: 09/13/2024]
Abstract
Experimental cell therapies for skeletal muscle conditions have shown little success, primarily because they use committed myogenic progenitors rather than true muscle stem cells, known as satellite cells. Here we present a method to generate in vitro-derived satellite cells (idSCs) from skeletal muscle tissue. When transplanted in small numbers into mouse muscle, mouse idSCs fuse into myofibers, repopulate the satellite cell niche, self-renew, support multiple rounds of muscle regeneration and improve force production on par with freshly isolated satellite cells in damaged skeletal muscle. We compared the epigenomic and transcriptional signatures between idSCs, myoblasts and satellite cells and used these signatures to identify core signaling pathways and genes that confer idSC functionality. Finally, from human muscle biopsies, we successfully generated satellite cell-like cells in vitro. After further development, idSCs may provide a scalable source of cells for the treatment of genetic muscle disorders, trauma-induced muscle damage and age-related muscle weakness.
Collapse
Affiliation(s)
- Feodor D Price
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA.
| | - Mark N Matyas
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Andrew R Gehrke
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - William Chen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Erica A Wolin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kristina M Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rebecca M Gibbs
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Alice Lee
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Pooja S Singu
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Jeffrey S Sakakeeny
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - James M Poteracki
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Kelsey Goune
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Isabella T Pfeiffer
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Sarah A Boswell
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Peter K Sorger
- Department of Systems Biology and Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Mansi Srivastava
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Kathleen Lindahl Pfaff
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Emanuela Gussoni
- Division of Genetics and Genomics and the Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Sean M Buchanan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Lee L Rubin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Ozturk T, Mignot J, Gattazzo F, Gervais M, Relaix F, Rouard H, Didier N. Dual inhibition of P38 MAPK and JNK pathways preserves stemness markers and alleviates premature activation of muscle stem cells during isolation. Stem Cell Res Ther 2024; 15:179. [PMID: 38902774 PMCID: PMC11191274 DOI: 10.1186/s13287-024-03795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/08/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Adult skeletal muscle contains resident muscle stem cells (MuSC) with high myogenic and engraftment potentials, making them suitable for cell therapy and regenerative medicine approaches. However, purification process of MuSC remains a major hurdle to their use in the clinic. Indeed, muscle tissue enzymatic dissociation triggers a massive activation of stress signaling pathways, among which P38 and JNK MAPK, associated with a premature loss of MuSC quiescence. While the role of these pathways in the myogenic progression of MuSC is well established, the extent to which their dissociation-induced activation affects the functionality of these cells remains unexplored. METHODS We assessed the effect of P38 and JNK MAPK induction on stemness marker expression and MuSC activation state during isolation by pharmacological approaches. MuSC functionality was evaluated by in vitro assays and in vivo transplantation experiments. We performed a comparative analysis of the transcriptome of human MuSC purified with pharmacological inhibitors of P38 and JNK MAPK (SB202190 and SP600125, respectively) versus available RNAseq resources. RESULTS We monitored PAX7 protein levels in murine MuSC during muscle dissociation and demonstrated a two-step decline partly dependent on P38 and JNK MAPK activities. We showed that simultaneous inhibition of these pathways throughout the MuSC isolation process preserves the expression of stemness markers and limits their premature activation, leading to improved survival and amplification in vitro as well as increased engraftment in vivo. Through a comparative RNAseq analysis of freshly isolated human MuSC, we provide evidence that our findings in murine MuSC could be relevant to human MuSC. Based on these findings, we implemented a purification strategy, significantly improving the recovery yields of human MuSC. CONCLUSION Our study highlights the pharmacological limitation of P38 and JNK MAPK activities as a suitable strategy to qualitatively and quantitatively ameliorate human MuSC purification process, which could be of great interest for cell-based therapies.
Collapse
Affiliation(s)
- Teoman Ozturk
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | - Julien Mignot
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | | | - Marianne Gervais
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
| | - Frédéric Relaix
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
- EnvA, IMRB, 94700, Maisons-Alfort, France
- AP-HP, Hopital Mondor, Service d'histologie, 94010, Creteil, France
| | - Hélène Rouard
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France
- AP-HP, Hopital Mondor, Service d'histologie, 94010, Creteil, France
| | - Nathalie Didier
- Univ Paris Est Creteil, INSERM, EFS, IMRB, 94010, Creteil, France.
| |
Collapse
|
3
|
Hicks MR, Saleh KK, Clock B, Gibbs DE, Yang M, Younesi S, Gane L, Gutierrez-Garcia V, Xi H, Pyle AD. Regenerating human skeletal muscle forms an emerging niche in vivo to support PAX7 cells. Nat Cell Biol 2023; 25:1758-1773. [PMID: 37919520 PMCID: PMC10709143 DOI: 10.1038/s41556-023-01271-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 09/26/2023] [Indexed: 11/04/2023]
Abstract
Skeletal muscle stem and progenitor cells including those derived from human pluripotent stem cells (hPSCs) offer an avenue towards personalized therapies and readily fuse to form human-mouse myofibres in vivo. However, skeletal muscle progenitor cells (SMPCs) inefficiently colonize chimeric stem cell niches and instead associate with human myofibres resembling foetal niches. We hypothesized competition with mouse satellite cells (SCs) prevented SMPC engraftment into the SC niche and thus generated an SC ablation mouse compatible with human engraftment. Single-nucleus RNA sequencing of SC-ablated mice identified the absence of a transient myofibre subtype during regeneration expressing Actc1. Similarly, ACTC1+ human myofibres supporting PAX7+ SMPCs increased in SC-ablated mice, and after re-injury we found SMPCs could now repopulate into chimeric niches. To demonstrate ACTC1+ myofibres are essential to supporting PAX7 SMPCs, we generated caspase-inducible ACTC1 depletion human pluripotent stem cells, and upon SMPC engraftment we found a 90% reduction in ACTC1+ myofibres and a 100-fold decrease in PAX7 cell numbers compared with non-induced controls. We used spatial RNA sequencing to identify key factors driving emerging human niche formation between ACTC1+ myofibres and PAX7+ SMPCs in vivo. This revealed that transient regenerating human myofibres are essential for emerging niche formation in vivo to support PAX7 SMPCs.
Collapse
Affiliation(s)
- Michael R Hicks
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Physiology and Biophysics, University of California, Irvine, CA, USA.
| | - Kholoud K Saleh
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, CA, USA
| | - Ben Clock
- Physiology and Biophysics, University of California, Irvine, CA, USA
| | - Devin E Gibbs
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Mandee Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Shahab Younesi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Lily Gane
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | | | - Haibin Xi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - April D Pyle
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA.
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, USA.
- Jonnson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Lazure F, Blackburn DM, Soleimani VD. Transcriptional Profiling of Skeletal Muscle Stem Cells After In Vivo Engraftment into a Heterologous Niche Environment. Curr Protoc 2023; 3:e877. [PMID: 37638781 DOI: 10.1002/cpz1.877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2023]
Abstract
Adult stem cells play a critical role in the maintenance and repair of the organs in which they reside. However, their function is highly dependent on the crosstalk with their niche environment that changes during development and in disease states. The niche provides signals to stem cells to activate, proliferate, self-renew, or remain in quiescence. In skeletal muscle, the niche is perturbed in disease contexts such as aging, muscular dystrophies, and cachexia. Therefore, it is important to develop methods that permit the decoupling of niche-mediated from cell-intrinsic changes that occur in muscle stem cells (MuSCs) in development and disease contexts. With the purpose of determining the effect of the niche environment on the MuSC transcriptome, function, or health, we have coupled an allogeneic stem cell transplantation system, meaning the transplantation of MuSCs from a donor mouse into a recipient host mouse, with Switching Mechanism at 5' End of RNA Template (SMART-Seq) to quantify the effects of the niche on the MuSC transcriptome in vivo. Briefly, MuSCs are isolated from a GFP reporter donor mouse (Pax7-nGFP) and transplanted into the irradiated muscles of immunocompromised allogeneic hosts. The MuSCs are re-isolated by fluorescence-activated cell sorting (FACS) after three weeks of inhabiting the heterologous niche, defined as a niche that is different from their originating niche, and sequencing-ready libraries are created. This method allows for the direct comparison of the transcriptome of stem cells before and after transplantation into a host of a different age, disease status, or genetic background. This method can be used to accurately quantify the direct effect of the niche environment on the stem cell gene expression profile and to decouple cell-intrinsic versus niche-mediated alterations in the stem cell transcriptome. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Allogeneic muscle stem cell transplantation.
Collapse
Affiliation(s)
- Felicia Lazure
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Quebec, Canada
| | - Darren M Blackburn
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Quebec, Canada
| | - Vahab D Soleimani
- Department of Human Genetics, McGill University, Montréal, Quebec, Canada
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Quebec, Canada
- Present address: Department of Biochemistry, Microbiology & Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| |
Collapse
|
5
|
Sesillo FB, Rajesh V, Wong M, Duran P, Rudell JB, Rundio CP, Baynes BB, Laurent LC, Sacco A, Christman KL, Alperin M. Muscle stem cells and fibro-adipogenic progenitors in female pelvic floor muscle regeneration following birth injury. NPJ Regen Med 2022; 7:72. [PMID: 36526635 PMCID: PMC9758192 DOI: 10.1038/s41536-022-00264-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Pelvic floor muscle (PFM) injury during childbirth is a key risk factor for pelvic floor disorders that affect millions of women worldwide. Muscle stem cells (MuSCs), supported by the fibro-adipogenic progenitors (FAPs) and immune cells, are indispensable for the regeneration of injured appendicular skeletal muscles. However, almost nothing is known about their role in PFM regeneration following birth injury. To elucidate the role of MuSCs, FAPs, and immune infiltrate in this context, we used radiation to perturb cell function and followed PFM recovery in a validated simulated birth injury (SBI) rat model. Non-irradiated and irradiated rats were euthanized at 3,7,10, and 28 days post-SBI (dpi). Twenty-eight dpi, PFM fiber cross-sectional area (CSA) was significantly lower and the extracellular space occupied by immune infiltrate was larger in irradiated relative to nonirradiated injured animals. Following SBI in non-irradiated animals, MuSCs and FAPs expanded significantly at 7 and 3 dpi, respectively; this expansion did not occur in irradiated animals at the same time points. At 7 and 10 dpi, we observed persistent immune response in PFMs subjected to irradiation compared to non-irradiated injured PFMs. CSA of newly regenerated fibers was also significantly smaller following SBI in irradiated compared to non-irradiated injured PFMs. Our results demonstrate that the loss of function and decreased expansion of MuSCs and FAPs after birth injury lead to impaired PFM recovery. These findings form the basis for further studies focused on the identification of novel therapeutic targets to counteract postpartum PFM dysfunction and the associated pelvic floor disorders.
Collapse
Affiliation(s)
- Francesca Boscolo Sesillo
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California, San Diego, San Diego, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Varsha Rajesh
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92161, USA
| | - Michelle Wong
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California, San Diego, San Diego, CA, 92037, USA
| | - Pamela Duran
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - John B Rudell
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California, San Diego, San Diego, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Courtney P Rundio
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California, San Diego, San Diego, CA, 92037, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
| | - Brittni B Baynes
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California, San Diego, San Diego, CA, 92037, USA
| | - Louise C Laurent
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Maternal-Fetal Medicine, University of San Diego, La Jolla, CA, 92037, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Karen L Christman
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Marianna Alperin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Division of Female Pelvic Medicine and Reconstructive Surgery, University of California, San Diego, San Diego, CA, 92037, USA.
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
6
|
Hekmatnejad B, Rudnicki MA. Transplantation to study satellite cell heterogeneity in skeletal muscle. Front Cell Dev Biol 2022; 10:902225. [PMID: 36092722 PMCID: PMC9448869 DOI: 10.3389/fcell.2022.902225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/26/2022] [Indexed: 11/15/2022] Open
Abstract
Skeletal muscle has a remarkable capacity to regenerate throughout life, which is mediated by its resident muscle stem cells, also called satellite cells. Satellite cells, located periphery to the muscle fibers and underneath the basal lamina, are an indispensable cellular source for muscle regeneration. Satellite cell transplantation into regenerating muscle contributes robustly to muscle repair, thereby indicating that satellite cells indeed function as adult muscle stem cells. Moreover, satellite cells are a heterogenous population in adult tissue, with subpopulations that can be distinguished based on gene expression, cell-cycle progression, ability to self-renew, and bi-potential ability. Transplantation assays provide a powerful tool to better understand satellite cell function in vivo enabling the separation of functionally distinct satellite cell subpopulations. In this review, we focus on transplantation strategies to explore satellite cells’ functional heterogeneity, approaches targeting the recipient tissue to improve transplantation efficiency, and common strategies to monitor the behaviour of the transplanted cells. Lastly, we discuss some recent approaches to overcome challenges to enhance the transplantation potential of muscle stem cells.
Collapse
Affiliation(s)
- Bahareh Hekmatnejad
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A. Rudnicki
- The Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- *Correspondence: Michael A. Rudnicki,
| |
Collapse
|
7
|
Meng J, Moore M, Counsell J, Muntoni F, Popplewell L, Morgan J. Optimized lentiviral vector to restore full-length dystrophin via a cell-mediated approach in a mouse model of Duchenne muscular dystrophy. Mol Ther Methods Clin Dev 2022; 25:491-507. [PMID: 35615709 PMCID: PMC9121076 DOI: 10.1016/j.omtm.2022.04.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 04/28/2022] [Indexed: 11/16/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the DMD gene. Restoration of full-length dystrophin protein in skeletal muscle would have therapeutic benefit, but lentivirally mediated delivery of such a large gene in vivo has been hindered by lack of tissue specificity, limited transduction, and insufficient transgene expression. To address these problems, we developed a lentiviral vector, which contains a muscle-specific promoter and sequence-optimized full-length dystrophin, to constrain dystrophin expression to differentiated myotubes/myofibers and enhance the transgene expression. We further explored the efficiency of restoration of full-length dystrophin in vivo, by grafting DMD myoblasts that had been corrected by this optimized lentiviral vector intramuscularly into an immunodeficient DMD mouse model. We show that these lentivirally corrected DMD myoblasts effectively reconstituted full-length dystrophin expression in 93.58% ± 2.17% of the myotubes in vitro. Moreover, dystrophin was restored in 64.4% ± 2.87% of the donor-derived regenerated muscle fibers in vivo, which were able to recruit members of the dystrophin-glycoprotein complex at the sarcolemma. This study represents a significant advance over existing cell-mediated gene therapy strategies for DMD that aim to restore full-length dystrophin expression in skeletal muscle.
Collapse
Affiliation(s)
- Jinhong Meng
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Marc Moore
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham TW20 0EX, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - John Counsell
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- UCL Division of Surgery and Interventional Science, Charles Bell House, 43-45 Foley Street, London W1W 7TY, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| | - Linda Popplewell
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham TW20 0EX, UK
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, UK
| |
Collapse
|
8
|
Primary cilia on muscle stem cells are critical to maintain regenerative capacity and are lost during aging. Nat Commun 2022; 13:1439. [PMID: 35301320 PMCID: PMC8931095 DOI: 10.1038/s41467-022-29150-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/02/2022] [Indexed: 12/11/2022] Open
Abstract
During aging, the regenerative capacity of muscle stem cells (MuSCs) decreases, diminishing the ability of muscle to repair following injury. We found that the ability of MuSCs to regenerate is regulated by the primary cilium, a cellular protrusion that serves as a sensitive sensory organelle. Abolishing MuSC cilia inhibited MuSC proliferation in vitro and severely impaired injury-induced muscle regeneration in vivo. In aged muscle, a cell intrinsic defect in MuSC ciliation was associated with the decrease in regenerative capacity. Exogenous activation of Hedgehog signaling, known to be localized in the primary cilium, promoted MuSC expansion, both in vitro and in vivo. Delivery of the small molecule Smoothened agonist (SAG1.3) to muscles of aged mice restored regenerative capacity leading to increased strength post-injury. These findings provide fresh insights into the signaling dysfunction in aged MuSCs and identify the ciliary Hedgehog signaling pathway as a potential therapeutic target to counter the loss of muscle regenerative capacity which accompanies aging. Repair of muscle damage requires muscle stem cells, which lose regenerative capacity with aging. Here, the authors show that a sensory organelle, the primary cilium, is critical for muscle stem cell proliferation during regeneration and lost with aging.
Collapse
|
9
|
Rocheteau P, Warot G, Chapellier M, Zampaolo M, Chretien F, Piquemal F. Cryopreserved Stem Cells Incur Damages Due To Terrestrial Cosmic Rays Impairing Their Integrity Upon Long-Term Storage. Cell Transplant 2022; 31:9636897211070239. [PMID: 35170351 PMCID: PMC8855380 DOI: 10.1177/09636897211070239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Stem cells have the capacity to ensure the renewal of tissues and organs. They
could be used in the future for a wide range of therapeutic purposes and are
preserved at liquid nitrogen temperature to prevent any chemical or biological
activity up to several decades before their use. We show that the cryogenized
cells accumulate damages coming from natural radiations, potentially inducing
DNA double-strand breaks (DSBs). Such DNA damage in stem cells could lead to
either mortality of the cells upon thawing or a mutation diminishing the
therapeutic potential of the treatment. Many studies show how stem cells react
to different levels of radiation; the effect of terrestrial cosmic rays being
key, it is thus also important to investigate the effect of the natural
radiation on the cryopreserved stem cell behavior over time. Our study showed
that the cryostored stem cells totally shielded from cosmic rays had less DSBs
upon long-term storage. This could have important implications on the long-term
cryostorage strategy and quality control of different cell banks.
Collapse
Affiliation(s)
- P Rocheteau
- Human Histopathology and Animal Models, Department of Infection & Epidemiology, Institut Pasteur, Paris, France
| | - G Warot
- Laboratoire de Physique Subatomique et Corpusculaire, UMR 5821, Université Grenoble Alpes, Centre National de la Recherche Scientifique, Grenoble Institute of Technology (Institute of Engineering University Grenoble Alpes), LPSC-IN2P3, Grenoble, France
| | - M Chapellier
- Laboratoire de Physique Subatomique et Corpusculaire, UMR 5821, Université Grenoble Alpes, Centre National de la Recherche Scientifique, Grenoble Institute of Technology (Institute of Engineering University Grenoble Alpes), LPSC-IN2P3, Grenoble, France
| | - M Zampaolo
- Laboratoire de Physique Subatomique et Corpusculaire, UMR 5821, Université Grenoble Alpes, Centre National de la Recherche Scientifique, Grenoble Institute of Technology (Institute of Engineering University Grenoble Alpes), LPSC-IN2P3, Grenoble, France
| | - F Chretien
- Human Histopathology and Animal Models, Department of Infection & Epidemiology, Institut Pasteur, Paris, France
| | - F Piquemal
- Centre d'Etudes Nucléaires de Bordeaux Gradignan, UMR 5797, Centre National de la Recherche Scientifique and Université de Bordeaux, Gradignan, France
| |
Collapse
|
10
|
Flück M, Kasper S, Benn MC, Clement Frey F, von Rechenberg B, Giraud MN, Meyer DC, Wieser K, Gerber C. Transplant of Autologous Mesenchymal Stem Cells Halts Fatty Atrophy of Detached Rotator Cuff Muscle After Tendon Repair: Molecular, Microscopic, and Macroscopic Results From an Ovine Model. Am J Sports Med 2021; 49:3970-3980. [PMID: 34714701 PMCID: PMC8649427 DOI: 10.1177/03635465211052566] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/13/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND The injection of mesenchymal stem cells (MSCs) mitigates fat accumulation in released rotator cuff muscle after tendon repair in rodents. PURPOSE To investigate whether the injection of autologous MSCs halts muscle-to-fat conversion after tendon repair in a large animal model for rotator cuff tendon release via regional effects on extracellular fat tissue and muscle fiber regeneration. STUDY DESIGN Controlled laboratory study. METHODS Infraspinatus (ISP) muscles of the right shoulder of Swiss Alpine sheep (n = 14) were released by osteotomy and reattached 16 weeks later without (group T; n = 6) or with (group T-MSC; n = 8) electropulse-assisted injection of 0.9 Mio fluorescently labeled MSCs as microtissues with media in demarcated regions; animals were allowed 6 weeks of recovery. ISP volume and composition were documented with computed tomography and magnetic resonance imaging. Area percentages of muscle fiber types, fat, extracellular ground substance, and fluorescence-positive tissue; mean cross-sectional area (MCSA) of muscle fibers; and expression of myogenic (myogenin), regeneration (tenascin-C), and adipogenic markers (peroxisome proliferator-activated receptor gamma [PPARG2]) were quantified in injected and noninjected regions after recovery. RESULTS At 16 weeks after tendon release, the ISP volume was reduced and the fat fraction of ISP muscle was increased in group T (137 vs 185 mL; 49% vs 7%) and group T-MSC (130 vs 166 mL; 53% vs 10%). In group T-MSC versus group T, changes during recovery after tendon reattachment were abrogated for fat-free mass (-5% vs -29%, respectively; P = .018) and fat fraction (+1% vs +24%, respectively; P = .009%). The area percentage of fat was lower (9% vs 20%; P = .018) and the percentage of the extracellular ground substance was higher (26% vs 20%; P = .007) in the noninjected ISP region for group T-MSC versus group T, respectively. Regionally, MCS injection increased tenascin-C levels (+59%) and the water fraction, maintaining the reduced PPARG2 levels but not the 29% increased fiber MCSA, with media injection. CONCLUSION In a sheep model, injection of autologous MSCs in degenerated rotator cuff muscle halted muscle-to-fat conversion during recovery from tendon repair by preserving fat-free mass in association with extracellular reactions and stopping adjuvant-induced muscle fiber hypertrophy. CLINICAL RELEVANCE A relatively small dose of MSCs is therapeutically effective to halt fatty atrophy in a large animal model.
Collapse
Affiliation(s)
- Martin Flück
- Laboratory of Muscle Plasticity,
Department of Orthopedics, University of Zurich, Balgrist Campus, Zürich,
Switzerland
| | - Stephanie Kasper
- Laboratory of Muscle Plasticity,
Department of Orthopedics, University of Zurich, Balgrist Campus, Zürich,
Switzerland
| | - Mario C. Benn
- Musculoskeletal Research Unit, Center
for Applied Biotechnology and Molecular Medicine, Department of Molecular
Mechanisms, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
| | - Flurina Clement Frey
- Musculoskeletal Research Unit, Center
for Applied Biotechnology and Molecular Medicine, Department of Molecular
Mechanisms, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
| | - Brigitte von Rechenberg
- Musculoskeletal Research Unit, Center
for Applied Biotechnology and Molecular Medicine, Department of Molecular
Mechanisms, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland
| | - Marie-Noëlle Giraud
- Cardiology, Faculty of Sciences and
Medicine, University of Fribourg, Fribourg, Switzerland
| | - Dominik C. Meyer
- Author deceased
- Laboratory of Muscle Plasticity,
Department of Orthopedics, University of Zurich, Balgrist Campus, Zürich,
Switzerland
- University Hospital Balgrist,
Department of Orthopedics, University of Zurich, Zürich, Switzerland
| | - Karl Wieser
- University Hospital Balgrist,
Department of Orthopedics, University of Zurich, Zürich, Switzerland
| | - Christian Gerber
- University Hospital Balgrist,
Department of Orthopedics, University of Zurich, Zürich, Switzerland
| |
Collapse
|
11
|
Yang BA, Castor-Macias J, Fraczek P, Cornett A, Brown LA, Kim M, Brooks SV, Lombaert IMA, Lee JH, Aguilar CA. Sestrins regulate muscle stem cell metabolic homeostasis. Stem Cell Reports 2021; 16:2078-2088. [PMID: 34388363 PMCID: PMC8452514 DOI: 10.1016/j.stemcr.2021.07.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/20/2023] Open
Abstract
The health and homeostasis of skeletal muscle are preserved by a population of tissue-resident muscle stem cells (MuSCs) that maintain a state of mitotic and metabolic quiescence in adult tissues. The capacity of MuSCs to preserve the quiescent state declines with aging and metabolic insults, promoting premature activation and stem cell exhaustion. Sestrins are a class of stress-inducible proteins that act as antioxidants and inhibit the activation of the mammalian target of rapamycin complex 1 (mTORC1) signaling complex. Despite these pivotal roles, the role of Sestrins has not been explored in adult stem cells. We show that SESTRIN1,2 loss results in hyperactivation of the mTORC1 complex, increased propensity to enter the cell cycle, and shifts in metabolic flux. Aged SESTRIN1,2 knockout mice exhibited loss of MuSCs and a reduced ability to regenerate injured muscle. These findings demonstrate that Sestrins help maintain metabolic pathways in MuSCs that protect quiescence against aging.
Collapse
Affiliation(s)
- Benjamin A Yang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jesus Castor-Macias
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Paula Fraczek
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ashley Cornett
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lemuel A Brown
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Susan V Brooks
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Isabelle M A Lombaert
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA; Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carlos A Aguilar
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
12
|
Arpke RW, Shams AS, Collins BC, Larson AA, Lu N, Lowe DA, Kyba M. Preservation of satellite cell number and regenerative potential with age reveals locomotory muscle bias. Skelet Muscle 2021; 11:22. [PMID: 34481522 PMCID: PMC8418011 DOI: 10.1186/s13395-021-00277-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/24/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Although muscle regenerative capacity declines with age, the extent to which this is due to satellite cell-intrinsic changes vs. environmental changes has been controversial. The majority of aging studies have investigated hindlimb locomotory muscles, principally the tibialis anterior, in caged sedentary mice, where those muscles are abnormally under-exercised. METHODS We analyze satellite cell numbers in 8 muscle groups representing locomotory and non-locomotory muscles in young and 2-year-old mice and perform transplantation assays of low numbers of hind limb satellite cells from young and old mice. RESULTS We find that satellite cell density does not decline significantly by 2 years of age in most muscles, and one muscle, the masseter, shows a modest but statistically significant increase in satellite cell density with age. The tibialis anterior and extensor digitorum longus were clear exceptions, showing significant declines. We quantify self-renewal using a transplantation assay. Dose dilution revealed significant non-linearity in self-renewal above a very low threshold, suggestive of competition between satellite cells for space within the pool. Assaying within the linear range, i.e., transplanting fewer than 1000 cells, revealed no evidence of decline in cell-autonomous self-renewal or regenerative potential of 2-year-old murine satellite cells. CONCLUSION These data demonstrate the value of comparative muscle analysis as opposed to overreliance on locomotory muscles, which are not used physiologically in aging sedentary mice, and suggest that self-renewal impairment with age is precipitously acquired at the geriatric stage, rather than being gradual over time, as previously thought.
Collapse
Affiliation(s)
- Robert W Arpke
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
- Present address: Division of Biological Sciences, University of Missouri, 105 Tucker Hall, Columbia, MO, 65211, USA
| | - Ahmed S Shams
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
- Human Anatomy and Embryology Department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Brittany C Collins
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, MN, 55455, USA
- Present address: Department of Human Genetics, University of Utah, 15 North 2030 East, Salt Lake City, UT, 84112, USA
| | - Alexie A Larson
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Nguyen Lu
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Dawn A Lowe
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, MN, USA
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, Medical School, University of Minnesota, Cancer and Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
13
|
Abizanda G, López-Muneta L, Linares J, Ramos LI, Baraibar-Churio A, Bobadilla M, Iglesias E, Coppiello G, Ripalda-Cemboráin P, Aranguren XL, Prósper F, Pérez-Ruiz A, Carvajal-Vergara X. Local Preirradiation of Infarcted Cardiac Tissue Substantially Enhances Cell Engraftment. Int J Mol Sci 2021; 22:ijms22179126. [PMID: 34502036 PMCID: PMC8430717 DOI: 10.3390/ijms22179126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022] Open
Abstract
The success of cell therapy for the treatment of myocardial infarction depends on finding novel approaches that can substantially implement the engraftment of the transplanted cells. In order to enhance cell engraftment, most studies have focused on the pretreatment of transplantable cells. Here we have considered an alternative approach that involves the preconditioning of infarcted heart tissue to reduce endogenous cell activity and thus provide an advantage to our exogenous cells. This treatment is routinely used in other tissues such as bone marrow and skeletal muscle to improve cell engraftment, but it has never been taken in cardiac tissue. To avoid long-term cardiotoxicity induced by full heart irradiation we developed a rat model of a catheter-based heart irradiation system to locally impact a delimited region of the infarcted cardiac tissue. As proof of concept, we transferred ZsGreen+ iPSCs in the infarcted heart, due to their ease of use and detection. We found a very significant increase in cell engraftment in preirradiated rats. In this study, we demonstrate for the first time that preconditioning the infarcted cardiac tissue with local irradiation can substantially enhance cell engraftment.
Collapse
Affiliation(s)
- Gloria Abizanda
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Leyre López-Muneta
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Javier Linares
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Luis I. Ramos
- Department of Oncology, Clínica Universidad de Navarra, 31008 Pamplona, Spain;
| | - Arantxa Baraibar-Churio
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Miriam Bobadilla
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Elena Iglesias
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Giulia Coppiello
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Purificación Ripalda-Cemboráin
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Xabier L. Aranguren
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
| | - Felipe Prósper
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Department of Hematology and Cell Therapy, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Ana Pérez-Ruiz
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
- Correspondence: (A.P.-R.); (X.C.-V.); Tel.: +34-948-194-700 (A.P.-R. & X.C.-V.)
| | - Xonia Carvajal-Vergara
- Regenerative Medicine Program, Foundation for Applied Medical Research (FIMA), University of Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (L.L.-M.); (J.L.); (A.B.-C.); (M.B.); (E.I.); (G.C.); (P.R.-C.); (X.L.A.); (F.P.)
- Correspondence: (A.P.-R.); (X.C.-V.); Tel.: +34-948-194-700 (A.P.-R. & X.C.-V.)
| |
Collapse
|
14
|
Kwon JB, Ettyreddy AR, Vankara A, Bohning JD, Devlin G, Hauschka SD, Asokan A, Gersbach CA. In Vivo Gene Editing of Muscle Stem Cells with Adeno-Associated Viral Vectors in a Mouse Model of Duchenne Muscular Dystrophy. Mol Ther Methods Clin Dev 2020; 19:320-329. [PMID: 33145368 PMCID: PMC7581966 DOI: 10.1016/j.omtm.2020.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/23/2020] [Indexed: 12/21/2022]
Abstract
Delivery of therapeutic transgenes with adeno-associated viral (AAV) vectors for treatment of myopathies has yielded encouraging results in animal models and early clinical studies. Although certain AAV serotypes efficiently target muscle fibers, transduction of the muscle stem cells, also known as satellite cells, is less studied. Here, we used a Pax7nGFP;Ai9 dual reporter mouse to quantify AAV transduction events in satellite cells. We assessed a panel of AAV serotypes for satellite cell tropism in the mdx mouse model of Duchenne muscular dystrophy and observed the highest satellite cell labeling with AAV9 following local or systemic administration. Subsequently, we used AAV9 to interrogate CRISPR/Cas9-mediated gene editing of satellite cells in the Pax7nGFP;mdx mouse. We quantified the level of gene editing using a Tn5 transposon-based method for unbiased sequencing of editing outcomes at the Dmd locus. We also found that muscle-specific promoters can drive transgene expression and gene editing in satellite cells. Lastly, to demonstrate the functionality of satellite cells edited at the Dmd locus by CRISPR in vivo, we performed a transplantation experiment and observed increased dystrophin-positive fibers in the recipient mouse. Collectively, our results confirm that satellite cells are transduced by AAV and can undergo gene editing to restore the dystrophin reading frame in the mdx mouse.
Collapse
Affiliation(s)
- Jennifer B. Kwon
- University Program in Genetics and Genomics, Duke University Medical Center, Durham, NC 27710, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
| | - Adarsh R. Ettyreddy
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Ashish Vankara
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Joel D. Bohning
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Garth Devlin
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | - Aravind Asokan
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
- Regeneration Next Initiative, Duke University Medical Center, Durham, NC 27710, USA
| | - Charles A. Gersbach
- Center for Advanced Genomic Technologies, Duke University, Durham, NC 27708, USA
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
- Regeneration Next Initiative, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
15
|
Chemoradiation impairs myofiber hypertrophic growth in a pediatric tumor model. Sci Rep 2020; 10:19501. [PMID: 33177579 PMCID: PMC7659015 DOI: 10.1038/s41598-020-75913-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023] Open
Abstract
Pediatric cancer treatment often involves chemotherapy and radiation, where off-target effects can include skeletal muscle decline. The effect of such treatments on juvenile skeletal muscle growth has yet to be investigated. We employed a small animal irradiator to administer fractionated hindlimb irradiation to juvenile mice bearing implanted rhabdomyosarcoma (RMS) tumors. Hindlimb-targeted irradiation (3 × 8.2 Gy) of 4-week-old mice successfully eliminated RMS tumors implanted one week prior. After establishment of this preclinical model, a cohort of tumor-bearing mice were injected with the chemotherapeutic drug, vincristine, alone or in combination with fractionated irradiation (5 × 4.8 Gy). Single myofiber analysis of fast-contracting extensor digitorum longus (EDL) and slow-contracting soleus (SOL) muscles was conducted 3 weeks post-treatment. Although a reduction in myofiber size was apparent, EDL and SOL myonuclear number were differentially affected by juvenile irradiation and/or vincristine treatment. In contrast, a decrease in myonuclear domain (myofiber volume/myonucleus) was observed regardless of muscle or treatment. Thus, inhibition of myofiber hypertrophic growth is a consistent feature of pediatric cancer treatment.
Collapse
|
16
|
Doreste B, Torelli S, Morgan J. Irradiation dependent inflammatory response may enhance satellite cell engraftment. Sci Rep 2020; 10:11119. [PMID: 32632224 PMCID: PMC7338540 DOI: 10.1038/s41598-020-68098-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 05/28/2020] [Indexed: 01/07/2023] Open
Abstract
Skeletal muscle stem (satellite) cells transplanted into host mouse muscles contribute to muscle regeneration. Irradiation of host muscle enhances donor stem cell engraftment by promoting the proliferation of transplanted donor cells. We hypothesised that, similar to other systems, cells damaged by radiation might be effecting this donor cell proliferation. But we found no difference in the percentage of dying (TUNEL+) cells in immunodeficient dystrophic mouse muscles at the times after the irradiation dose that enhances donor cell engraftment. Similarly, irradiation did not significantly increase the number of TUNEL+ cells in non-dystrophic immunodeficient mouse muscles and it only slightly enhanced donor satellite cell engraftment in this mouse strain, suggesting either that the effector cells are present in greater numbers within dystrophic muscle, or that an innate immune response is required for effective donor cell engraftment. Donor cell engraftment within non-irradiated dystrophic host mouse muscles was not enhanced if they were transplanted with either satellite cells, or myofibres, derived from irradiated dystrophic mouse muscle. But a mixture of cells from irradiated muscle transplanted with donor satellite cells promoted donor cell engraftment in a few instances, suggesting that a rare, yet to be identified, cell type within irradiated dystrophic muscle enhances the donor stem cell-mediated regeneration. The mechanism by which cells within irradiated host muscle promote donor cell engraftment remains elusive.
Collapse
Affiliation(s)
- Bruno Doreste
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N1EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Silvia Torelli
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N1EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N1EH, UK.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
17
|
Yanay N, Rabie M, Nevo Y. Impaired Regeneration in Dystrophic Muscle-New Target for Therapy. Front Mol Neurosci 2020; 13:69. [PMID: 32523512 PMCID: PMC7261890 DOI: 10.3389/fnmol.2020.00069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Muscle stem cells (MuSCs), known as satellite cells (SCs) have an incredible ability to regenerate, which enables the maintenance and growth of muscle tissue. In response to damaging stimuli, SCs are activated, proliferate, differentiate, and fuse to repair or generate a new muscle fiber. However, dystrophic muscles are characterized by poor muscle regeneration along with chronic inflammation and fibrosis. Indications for SC involvement in muscular dystrophy pathologies are accumulating, but their contribution to muscle pathophysiology is not precisely understood. In congenital muscular dystrophy type 1A (LAMA2-CMD), mutations in Lama2 gene cause either complete or partial absence in laminin-211 protein. Laminin-211 functions as a link between muscle extracellular matrix (ECM) and two adhesion systems in the sarcolemma; one is the well-known dystrophin-glycoprotein complex (DGC), and the second is the integrin complex. Because of its protein interactions and location, laminin-211 has a crucial role in muscle function and survival by maintaining sarcolemma integrity. In addition, laminin-211 is expressed in SCs and suggested to have a role in SC proliferation and differentiation. Downstream to the primary defect in laminin-211, several secondary genes and pathways accelerate disease mechanism, while at the same time there are unsuccessful attempts to regenerate as compensation for the dystrophic process. Lately, next-generation sequencing platforms have advanced our knowledge about the secondary events occurring in various diseases, elucidate the pathophysiology, and characterize new essential targets for development of new treatment strategies. This review will mainly focus on SC contribution to impaired regeneration in muscular dystrophies and specifically new findings suggesting SC involvement in LAMA2-CMD pathology.
Collapse
Affiliation(s)
- Nurit Yanay
- Felsenstein Medical Research Center (FMRC), Tel-Aviv University, Tel-Aviv, Israel.,Institute of Neurology, Schneider Children's Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Malcolm Rabie
- Felsenstein Medical Research Center (FMRC), Tel-Aviv University, Tel-Aviv, Israel.,Institute of Neurology, Schneider Children's Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| | - Yoram Nevo
- Felsenstein Medical Research Center (FMRC), Tel-Aviv University, Tel-Aviv, Israel.,Institute of Neurology, Schneider Children's Medical Center, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
18
|
Mueller AL, Bloch RJ. Skeletal muscle cell transplantation: models and methods. J Muscle Res Cell Motil 2019; 41:297-311. [PMID: 31392564 DOI: 10.1007/s10974-019-09550-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023]
Abstract
Xenografts of skeletal muscle are used to study muscle repair and regeneration, mechanisms of muscular dystrophies, and potential cell therapies for musculoskeletal disorders. Typically, xenografting involves using an immunodeficient host that is pre-injured to create a niche for human cell engraftment. Cell type and method of delivery to muscle depend on the specific application, but can include myoblasts, satellite cells, induced pluripotent stem cells, mesangioblasts, immortalized muscle precursor cells, and other multipotent cell lines delivered locally or systemically. Some studies follow cell engraftment with interventions to enhance cell proliferation, migration, and differentiation into mature muscle fibers. Recently, several advances in xenografting human-derived muscle cells have been applied to study and treat Duchenne muscular dystrophy and Facioscapulohumeral muscular dystrophy. Here, we review the vast array of techniques available to aid researchers in designing future experiments aimed at creating robust muscle xenografts in rodent hosts.
Collapse
Affiliation(s)
- Amber L Mueller
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA.
| |
Collapse
|
19
|
Han WM, Mohiuddin M, Anderson SE, García AJ, Jang YC. Co-delivery of Wnt7a and muscle stem cells using synthetic bioadhesive hydrogel enhances murine muscle regeneration and cell migration during engraftment. Acta Biomater 2019; 94:243-252. [PMID: 31228633 DOI: 10.1016/j.actbio.2019.06.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/09/2019] [Accepted: 06/17/2019] [Indexed: 01/02/2023]
Abstract
Skeletal muscle possesses efficient ability to regenerate upon minor injuries, but its capacity to regenerate is severely compromised with traumatic injuries and muscle-associated diseases. Recent evidence suggests that skeletal muscle regeneration can be enhanced by transplantation of muscle satellite cells (MuSCs) or treatment with pro-myogenic factors, such as Wingless-type MMTV Integrated 7a (Wnt7a) protein. Although direct intramuscular injection is the simplest method to deliver MuSCs and Wnt7a for regenerative therapy, direct injections are not viable in many clinical cases where structural integrity is severely compromised. To address this challenge, we evaluated the feasibility of co-delivering pro-myogenic factors, such as Wnt7a, and MuSCs using a synthetic poly(ethylene glycol) (PEG)-based hydrogel to the affected skeletal muscles. The Wnt7a release rate can be controlled by modulating the polymer density of the hydrogel, and this release rate can be further accelerated through the proteolytic degradation of the hydrogel. Treating cryo-injured tibialis anterior (TA) muscles with Wnt7a-loaded hydrogels resulted in an improved regenerative response by day 14, measured by increased muscle fiber cross-sectional area, bulk TA mass, and the number of Pax7+ MuSCs at the injury site, compared to the TA muscles treated with Wnt7a-free hydrogels. Co-delivery of Wnt7a and primary MuSCs using the synthetic hydrogel to the cryo-injured TA muscles significantly increased cellular migration during the engraftment process. This work provides a synthetic biomaterial platform for advancing treatment strategies of skeletal muscle conditions where direct intramuscular injection may be challenging. Finally, the current outcomes establish an important foundation for future applications in treating severe muscle trauma and diseases, where the endogenous repair capacity is critically impaired. STATEMENT OF SIGNIFICANCE: Skeletal muscle injuries and diseases cause debilitating health consequences, including disability and diminished quality of life. Treatment using protein and stem cell-based therapeutics may help regenerate the affected muscles, but direct intramuscular injection may not be feasible in severe muscle injuries due to the gravely damaged tissue structure. In chronic muscle diseases, such as Duchenne muscular dystrophy, local treatment of the diaphragm, a muscle critical for respiration, may be necessary but direct injection is difficult due to its thin dimensions. To address this challenge, this work presents a synthetic and bioactive muscle "patch" that enables concurrent administration of proteins and muscle stem cells for accelerated muscle healing.
Collapse
|
20
|
Sah JP, Hao NTT, Kim Y, Eigler T, Tzahor E, Kim SH, Hwang Y, Yoon JK. MBP-FGF2-Immobilized Matrix Maintains Self-Renewal and Myogenic Differentiation Potential of Skeletal Muscle Stem Cells. Int J Stem Cells 2019; 12:360-366. [PMID: 30836735 PMCID: PMC6657940 DOI: 10.15283/ijsc18125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/22/2022] Open
Abstract
The robust capacity of skeletal muscle stem cells (SkMSCs, or satellite cells) to regenerate into new muscles in vivo has offered promising therapeutic options for the treatment of degenerative muscle diseases. However, the practical use of SkMSCs to treat muscle diseases is limited, owing to their inability to expand in vitro under defined cultivation conditions without loss of engraftment efficiency. To develop an optimal cultivation condition for SkMSCs, we investigated the behavior of SkMSCs on synthetic maltose-binding protein (MBP)-fibroblast growth factor 2 (FGF2)-immobilized matrix in vitro. We found that the chemically well-defined, xeno-free MBP-FGF2-immobilized matrix effectively supports SkMSC growth without reducing their differentiation potential in vitro. Our data highlights the possible application of the MBP-FGF2 matrix for SkMSC expansion in vitro.
Collapse
Affiliation(s)
- Jay Prakash Sah
- Soonchunhyang Institute of Medi-bio Science, Soon Chun Hyang University, Cheonan, Korea.,Department of Integrated Biomedical Science, Graduate School, Soon Chun Hyang University, Asan, Korea
| | - Nguyen Thi Thu Hao
- Soonchunhyang Institute of Medi-bio Science, Soon Chun Hyang University, Cheonan, Korea.,Department of Integrated Biomedical Science, Graduate School, Soon Chun Hyang University, Asan, Korea
| | - Yunhye Kim
- Soonchunhyang Institute of Medi-bio Science, Soon Chun Hyang University, Cheonan, Korea.,Department of Integrated Biomedical Science, Graduate School, Soon Chun Hyang University, Asan, Korea
| | - Tamar Eigler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sang-Heon Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Korea
| | - Yongsung Hwang
- Soonchunhyang Institute of Medi-bio Science, Soon Chun Hyang University, Cheonan, Korea.,Department of Integrated Biomedical Science, Graduate School, Soon Chun Hyang University, Asan, Korea
| | - Jeong Kyo Yoon
- Soonchunhyang Institute of Medi-bio Science, Soon Chun Hyang University, Cheonan, Korea.,Department of Integrated Biomedical Science, Graduate School, Soon Chun Hyang University, Asan, Korea
| |
Collapse
|
21
|
Smith LR, Irianto J, Xia Y, Pfeifer CR, Discher DE. Constricted migration modulates stem cell differentiation. Mol Biol Cell 2019; 30:1985-1999. [PMID: 31188712 PMCID: PMC6727770 DOI: 10.1091/mbc.e19-02-0090] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tissue regeneration at an injured site depends on proliferation, migration, and differentiation of resident stem or progenitor cells, but solid tissues are often sufficiently dense and constricting that nuclei are highly stressed by migration. In this study, constricted migration of myoblastic cell types and mesenchymal stem cells (MSCs) increases nuclear rupture, increases DNA damage, and modulates differentiation. Fewer myoblasts fuse into regenerating muscle in vivo after constricted migration in vitro, and myodifferentiation in vitro is likewise suppressed. Myosin II inhibition rescues rupture and DNA damage, implicating nuclear forces, while mitosis and the cell cycle are suppressed by constricted migration, consistent with a checkpoint. Although perturbed proliferation fails to explain defective differentiation, nuclear rupture mislocalizes differentiation-relevant MyoD and KU80 (a DNA repair factor), with nuclear entry of the DNA-binding factor cGAS. Human MSCs exhibit similar damage, but osteogenesis increases-which is relevant to bone and to calcified fibrotic tissues, including diseased muscle. Tissue repair can thus be modulated up or down by the curvature of pores through which stem cells squeeze.
Collapse
Affiliation(s)
- Lucas R. Smith
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616,Department of Physical Medicine and Rehabilitation, University of California, Davis, Sacramento, CA 95817
| | - Jerome Irianto
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104
| | - Yuntao Xia
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104
| | - Charlotte R. Pfeifer
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E. Discher
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104,*Address correspondence to: Dennis E. Discher ()
| |
Collapse
|
22
|
Morgan J, Butler-Browne G, Muntoni F, Patel K. 240th ENMC workshop: The involvement of skeletal muscle stem cells in the pathology of muscular dystrophies 25-27 January 2019, Hoofddorp, The Netherlands. Neuromuscul Disord 2019; 29:704-715. [PMID: 31447279 DOI: 10.1016/j.nmd.2019.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 07/14/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Jennifer Morgan
- University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, UK.
| | - Gillian Butler-Browne
- Center for Research in Myology, Association Institut de Myologie, Inserm, Sorbonne Université, 75013 Paris, France
| | - Francesco Muntoni
- University College London Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, UK
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, RG6 6AS, UK.
| | | |
Collapse
|
23
|
High-Dimensional Single-Cell Cartography Reveals Novel Skeletal Muscle-Resident Cell Populations. Mol Cell 2019; 74:609-621.e6. [PMID: 30922843 DOI: 10.1016/j.molcel.2019.02.026] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 12/13/2018] [Accepted: 02/14/2019] [Indexed: 12/12/2022]
Abstract
Adult tissue repair and regeneration require stem-progenitor cells that can self-renew and generate differentiated progeny. Skeletal muscle regenerative capacity relies on muscle satellite cells (MuSCs) and their interplay with different cell types within the niche. However, our understanding of skeletal muscle tissue cellular composition is limited. Here, using a combined approach of single-cell RNA sequencing and mass cytometry, we precisely mapped 10 different mononuclear cell types in adult mouse muscle. We also characterized gene signatures and determined key discriminating markers of each cell type. We identified two previously understudied cell populations in the interstitial compartment. One expresses the transcription factor scleraxis and generated tenocytes in vitro. The second expresses markers of smooth muscle and mesenchymal cells (SMMCs) and, while distinct from MuSCs, exhibited myogenic potential and promoted MuSC engraftment following transplantation. The blueprint presented here yields crucial insights into muscle-resident cell-type identities and can be exploited to study muscle diseases.
Collapse
|
24
|
Moyle LA, Tedesco FS, Benedetti S. Pericytes in Muscular Dystrophies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:319-344. [PMID: 31147885 DOI: 10.1007/978-3-030-16908-4_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The muscular dystrophies are an heterogeneous group of inherited myopathies characterised by the progressive wasting of skeletal muscle tissue. Pericytes have been shown to make muscle in vitro and to contribute to skeletal muscle regeneration in several animal models, although recent data has shown this to be controversial. In fact, some pericyte subpopulations have been shown to contribute to fibrosis and adipose deposition in muscle. In this chapter, we explore the identity and the multifaceted role of pericytes in dystrophic muscle, potential therapeutic applications and the current need to overcome the hurdles of characterisation (both to identify pericyte subpopulations and track cell fate), to prevent deleterious differentiation towards myogenic-inhibiting subpopulations, and to improve cell proliferation and engraftment efficacy.
Collapse
Affiliation(s)
- Louise Anne Moyle
- Institute of Biomaterials and Biomedical Engineering, Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, UK.
- Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Sara Benedetti
- Great Ormond Street Institute of Child Health, University College London, London, UK.
- NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
25
|
Siemionow M, Cwykiel J, Heydemann A, Garcia-Martinez J, Siemionow K, Szilagyi E. Creation of Dystrophin Expressing Chimeric Cells of Myoblast Origin as a Novel Stem Cell Based Therapy for Duchenne Muscular Dystrophy. Stem Cell Rev Rep 2018; 14:189-199. [PMID: 29305755 PMCID: PMC5887005 DOI: 10.1007/s12015-017-9792-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over the past decade different stem cell (SC) based approaches were tested to treat Duchenne Muscular Dystrophy (DMD), a lethal X-linked disorder caused by mutations in dystrophin gene. Despite research efforts, there is no curative therapy for DMD. Allogeneic SC therapies aim to restore dystrophin in the affected muscles; however, they are challenged by rejection and limited engraftment. Thus, there is a need to develop new more efficacious SC therapies. Chimeric Cells (CC), created via ex vivo fusion of donor and recipient cells, represent a promising therapeutic option for tissue regeneration and Vascularized Composite Allotransplantation (VCA) due to tolerogenic properties that eliminate the need for lifelong immunosuppression. This proof of concept study tested feasibility of myoblast fusion for Dystrophin Expressing. Chimeric Cell (DEC) therapy through in vitro characterization and in vivo assessment of engraftment, survival, and efficacy in the mdx mouse model of DMD. Murine DEC were created via ex vivo fusion of normal (snj) and dystrophin–deficient (mdx) myoblasts using polyethylene glycol. Efficacy of myoblast fusion was confirmed by flow cytometry and dystrophin immunostaining, while proliferative and myogenic differentiation capacity of DEC were assessed in vitro. Therapeutic effect after DEC transplant (0.5 × 106) into the gastrocnemius muscle (GM) of mdx mice was assessed by muscle functional tests. At 30 days post-transplant dystrophin expression in GM of injected mdx mice increased to 37.27 ± 12.1% and correlated with improvement of muscle strength and function. Our study confirmed feasibility and efficacy of DEC therapy and represents a novel SC based approach for treatment of muscular dystrophies.
Collapse
Affiliation(s)
- M Siemionow
- Department of Surgery, Poznan University of Medical Sciences, Poznan, Poland.
- Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA.
| | - J Cwykiel
- Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA
| | - A Heydemann
- Department of Physiology, University of Illinois at Chicago, Chicago, IL, USA
| | - J Garcia-Martinez
- Department of Physiology, University of Illinois at Chicago, Chicago, IL, USA
| | - K Siemionow
- Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA
| | - E Szilagyi
- Department of Orthopedics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
26
|
Lee EJ, Kim M, Kim YD, Chung MJ, Elfadl A, Ulah HMA, Park D, Lee S, Park HS, Kim TH, Hwang D, Jeong KS. Establishment of stably expandable induced myogenic stem cells by four transcription factors. Cell Death Dis 2018; 9:1092. [PMID: 30361642 PMCID: PMC6202407 DOI: 10.1038/s41419-018-1114-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/04/2018] [Accepted: 09/24/2018] [Indexed: 12/14/2022]
Abstract
Life-long regeneration of healthy muscle by cell transplantation is an ideal therapy for patients with degenerative muscle diseases. Yet, obtaining muscle stem cells from patients is very limited due to their exhaustion in disease condition. Thus, development of a method to obtain healthy myogenic stem cells is required. Here, we showed that the four transcription factors, Six1, Eya1, Esrrb, and Pax3, converts fibroblasts into induced myogenic stem cells (iMSCs). The iMSCs showed effective differentiation into multinucleated myotubes and also higher proliferation capacity than muscle derived stem cells both in vitro and in vivo. The iMSCs do not lose their proliferation capacity though the passaging number is increased. We further isolated CD106-negative and α7-integrin-positive iMSCs (sort-iMSCs) showing higher myogenic differentiation capacity than iMSCs. Moreover, genome-wide transcriptomic analysis of iMSCs and sort-iMSCs, followed by network analysis, revealed the genes and signaling pathways associated with enhanced proliferation and differentiation capacity of iMSCs and sort-iMSCs, respectively. The stably expandable iMSCs provide a new source for drug screening and muscle regenerative therapy for muscle wasting disease.
Collapse
Affiliation(s)
- Eun-Joo Lee
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Minhyung Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Yong Deuk Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Myung-Jin Chung
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ahmed Elfadl
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - H M Arif Ulah
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Dongsu Park
- Department of Molecular Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.,Center for Skeletal Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.,Department of Pathology and Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Sunray Lee
- Cell Engineering for Origin Research Center 45-13, Ujeongguk-ro, Jongno-gu, Seoul, 03150, Republic of Korea
| | - Hyun-Sook Park
- Cell Engineering for Origin Research Center 45-13, Ujeongguk-ro, Jongno-gu, Seoul, 03150, Republic of Korea
| | - Tae-Hwan Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea.,Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.,Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea. .,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
27
|
Han WM, Anderson SE, Mohiuddin M, Barros D, Nakhai SA, Shin E, Amaral IF, Pêgo AP, García AJ, Jang YC. Synthetic matrix enhances transplanted satellite cell engraftment in dystrophic and aged skeletal muscle with comorbid trauma. SCIENCE ADVANCES 2018; 4:eaar4008. [PMID: 30116776 PMCID: PMC6093653 DOI: 10.1126/sciadv.aar4008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 07/11/2018] [Indexed: 05/29/2023]
Abstract
Muscle satellite cells (MuSCs) play a central role in muscle regeneration, but their quantity and function decline with comorbidity of trauma, aging, and muscle diseases. Although transplantation of MuSCs in traumatically injured muscle in the comorbid context of aging or pathology is a strategy to boost muscle regeneration, an effective cell delivery strategy in these contexts has not been developed. We engineered a synthetic hydrogel-based matrix with optimal mechanical, cell-adhesive, and protease-degradable properties that promotes MuSC survival, proliferation, and differentiation. Furthermore, we establish a biomaterial-mediated cell delivery strategy for treating muscle trauma, where intramuscular injections may not be applicable. Delivery of MuSCs in the engineered matrix significantly improved in vivo cell survival, proliferation, and engraftment in nonirradiated and immunocompetent muscles of aged and dystrophic mice compared to collagen gels and cell-only controls. This platform may be suitable for treating craniofacial and limb muscle trauma, as well as postoperative wounds of elderly and dystrophic patients.
Collapse
Affiliation(s)
- Woojin M. Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Shannon E. Anderson
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Mahir Mohiuddin
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Daniela Barros
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Shadi A. Nakhai
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Eunjung Shin
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Isabel Freitas Amaral
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Andrés J. García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Young C. Jang
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
28
|
Zhou Y, Lovell D, Bethea M, Yoseph B, Poteracki J, Soker S, Criswell T. * The Impact of Age on Skeletal Muscle Progenitor Cell Survival and Fate After Injury. Tissue Eng Part C Methods 2018; 23:1012-1021. [PMID: 29092672 DOI: 10.1089/ten.tec.2017.0216] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Sarcopenia is defined as the loss of skeletal muscle mass and function due to age, and represents a major cause of disability in the elderly population. The contributing factors to the onset of sarcopenia are not well defined, but appear to involve age-dependent changes in both the tissue microenvironment and muscle progenitor cell (MPC) population. MPC transplantation has the potential to be a novel therapy for treatment of muscle dysfunction due to aging or injury, but has not shown significant clinical efficacy to date. The goal of this research was to use a rat model of skeletal muscle injury to examine the differential effects of age on MPC survival, differentiation, and tissue regeneration after transplantation. Fluorescently labeled MPCs, derived from young (YMPCs) and adult (AMPCs) donor rats, were transplanted in the injured tibialis anterior (TA) muscles of young, adult, and aged rats. Our results demonstrated that integration and maturation of YMPCs into mature myofibers were dependent on the age of the host microenvironment; whereas, the integration and maturation of AMPCs were less dependent on age and more dependent on intrinsic cellular changes. These data suggest that the age of both the host microenvironment and cells for transplantation must be considered when designing cell therapy regimens.
Collapse
Affiliation(s)
- Yu Zhou
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Daniel Lovell
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Maigen Bethea
- 2 Cell Molecular & Developmental Biology, University of Alabama , Birmingham, Alabama
| | - Benyam Yoseph
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - James Poteracki
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Shay Soker
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| | - Tracy Criswell
- 1 Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine , Winston-Salem, North Carolina
| |
Collapse
|
29
|
Forcina L, Miano C, Musarò A. The physiopathologic interplay between stem cells and tissue niche in muscle regeneration and the role of IL-6 on muscle homeostasis and diseases. Cytokine Growth Factor Rev 2018; 41:1-9. [PMID: 29778303 DOI: 10.1016/j.cytogfr.2018.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 12/11/2022]
Abstract
Skeletal muscle is a complex, dynamic tissue characterized by an elevated plasticity. Although the adult muscle is mainly composed of multinucleated fibers with post mitotic nuclei, it retains a remarkable ability to regenerate in response to traumatic events. The regenerative potential of the adult skeletal muscle relies in the activity of satellite cells, mononucleated cells residing within the muscle in intimate association with myofibers. Satellite cells normally remain quiescent in their sublaminar position, sporadically entering the cell cycle to guarantee an efficient cellular turnover, by fusing with pre-existing myofibers, and to maintain the stem cell pool. However, after muscle injury satellite cells undergo an extensive increase of their activity in response to environmental stimuli, thereby participating to the regeneration of a functional muscle tissue. Nevertheless, regeneration is affected in several pathologic conditions and by a wide range of environmental signals that are highly variable, not only through time, but also depending on the physiological or pathological conditions of the musculature. Among these factors, the interleukin-6 (IL-6) plays a critical physiopathologic role on muscle homeostasis and diseases. The basis of muscle regeneration and the impact of IL-6 on the physiopathology of skeletal muscle will be discussed.
Collapse
Affiliation(s)
- Laura Forcina
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Carmen Miano
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy
| | - Antonio Musarò
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Via A. Scarpa, 14, Rome 00161, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy.
| |
Collapse
|
30
|
Smith LR, Barton ER. Regulation of fibrosis in muscular dystrophy. Matrix Biol 2018; 68-69:602-615. [PMID: 29408413 DOI: 10.1016/j.matbio.2018.01.014] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 02/08/2023]
Abstract
The production of force and power are inherent properties of skeletal muscle, and regulated by contractile proteins within muscle fibers. However, skeletal muscle integrity and function also require strong connections between muscle fibers and their extracellular matrix (ECM). A well-organized and pliant ECM is integral to muscle function and the ability for many different cell populations to efficiently migrate through ECM is critical during growth and regeneration. For many neuromuscular diseases, genetic mutations cause disruption of these cytoskeletal-ECM connections, resulting in muscle fragility and chronic injury. Ultimately, these changes shift the balance from myogenic pathways toward fibrogenic pathways, culminating in the loss of muscle fibers and their replacement with fatty-fibrotic matrix. Hence a common pathological hallmark of muscular dystrophy is prominent fibrosis. This review will cover the salient features of muscular dystrophy pathogenesis, highlight the signals and cells that are important for myogenic and fibrogenic actions, and discuss how fibrosis alters the ECM of skeletal muscle, and the consequences of fibrosis in developing therapies.
Collapse
Affiliation(s)
- Lucas R Smith
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Elisabeth R Barton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
31
|
Cid‐Díaz T, Santos‐Zas I, González‐Sánchez J, Gurriarán‐Rodríguez U, Mosteiro CS, Casabiell X, García‐Caballero T, Mouly V, Pazos Y, Camiña JP. Obestatin controls the ubiquitin-proteasome and autophagy-lysosome systems in glucocorticoid-induced muscle cell atrophy. J Cachexia Sarcopenia Muscle 2017; 8:974-990. [PMID: 28675664 PMCID: PMC5700440 DOI: 10.1002/jcsm.12222] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/09/2017] [Accepted: 05/22/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Many pathological states characterized by muscle atrophy are associated with an increase in circulating glucocorticoids and poor patient prognosis, making it an important target for treatment. The development of treatments for glucocorticoid-induced and wasting disorder-related skeletal muscle atrophy should be designed based on how the particular transcriptional program is orchestrated and how the balance of muscle protein synthesis and degradation is deregulated. Here, we investigated whether the obestatin/GPR39 system, an autocrine/paracrine signaling system acting on myogenesis and with anabolic effects on the skeletal muscle, could protect against glucocorticoid-induced muscle cell atrophy. METHODS In the present study, we have utilized mouse C2C12 myotube cultures to examine whether the obestatin/GPR39 signaling pathways can affect the atrophy induced by the synthetic glucocorticoid dexamethasone. We have extended these findings to in vitro effects on human atrophy using human KM155C25 myotubes. RESULTS The activation of the obestatin/GPR39 system protects from glucocorticoid-induced atrophy by regulation of Akt, PKD/PKCμ, CAMKII and AMPK signaling and its downstream targets in the control of protein synthesis, ubiquitin-proteasome system and autophagy-lysosome system in mouse cells. We compared mouse and human myotube cells in their response to glucocorticoid and identified differences in both the triggering of the atrophic program and the response to obestatin stimulation. Notably, we demonstrate that specific patterns of post-translational modifications of FoxO4 and FoxO1 play a key role in directing FoxO activity in response to obestatin in human myotubes. CONCLUSIONS Our findings emphasize the function of the obestatin/GPR39 system in coordinating a variety of pathways involved in the regulation of protein degradation during catabolic conditions.
Collapse
Affiliation(s)
- Tania Cid‐Díaz
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS)Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS)Choupana s/n15706Santiago de CompostelaSpain
| | - Icía Santos‐Zas
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS)Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS)Choupana s/n15706Santiago de CompostelaSpain
| | - Jessica González‐Sánchez
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS)Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS)Choupana s/n15706Santiago de CompostelaSpain
| | - Uxía Gurriarán‐Rodríguez
- Sprott Center for Stem Cell ResearchOttawa Hospital Research Institute501 Smyth RoadOttawaOntarioK1H 8L6Canada
| | - Carlos S. Mosteiro
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS)Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS)Choupana s/n15706Santiago de CompostelaSpain
| | - Xesús Casabiell
- Departamento de FisiologíaFacultad de Veterinaria, Universidad de Santiago de Compostela (USC)Carballo Calero s/n27002LugoSpain
| | - Tomás García‐Caballero
- Departamento de Ciencias MorfológicasFacultad de Medicina, USCSan Francisco s/n15704Santiago de CompostelaSpain
| | - Vincent Mouly
- Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, CNRS FRE3617, Center for Research in Myology47 Boulevard de l'hôpital75013ParisFrance
| | - Yolanda Pazos
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS)Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS)Choupana s/n15706Santiago de CompostelaSpain
| | - Jesús P. Camiña
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS)Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS)Choupana s/n15706Santiago de CompostelaSpain
| |
Collapse
|
32
|
Cezar CA, Arany P, Vermillion SA, Seo BR, Vandenburgh HH, Mooney DJ. Timed Delivery of Therapy Enhances Functional Muscle Regeneration. Adv Healthc Mater 2017; 6:10.1002/adhm.201700202. [PMID: 28703489 PMCID: PMC5641972 DOI: 10.1002/adhm.201700202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/09/2017] [Indexed: 01/14/2023]
Abstract
Cell transplantation is a promising therapeutic strategy for the treatment of traumatic muscle injury in humans. Previous investigations have typically focused on the identification of potent cell and growth factor treatments and optimization of spatial control over delivery. However, the optimal time point for cell transplantation remains unclear. Here, this study reports how myoblast and morphogen delivery timed to coincide with specific phases of the inflammatory response affects donor cell engraftment and the functional repair of severely injured muscle. Delivery of a biomaterial-based therapy timed with the peak of injury-induced inflammation leads to potent early and long-term regenerative benefits. Diminished inflammation and fibrosis, enhanced angiogenesis, and increased cell engraftment are seen during the acute stage following optimally timed treatment. Over the long term, treatment during peak inflammation leads to enhanced functional regeneration, as indicated by reduced chronic inflammation and fibrosis along with increased tissue perfusion and muscle contractile force. Treatments initiated immediately after injury or after inflammation had largely resolved provided more limited benefits. These results demonstrate the importance of appropriately timing the delivery of biologic therapy in the context of muscle regeneration. Biomaterial-based timed delivery can likely be applied to other tissues and is of potential wide utility in regenerative medicine.
Collapse
Affiliation(s)
- Christine A Cezar
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| | - Praveen Arany
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, 3435 Main Street, Buffalo, NY, 14260, USA
| | - Sarah A Vermillion
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| | - Bo Ri Seo
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| | - Herman H Vandenburgh
- Department of Pathology and Lab Medicine, Brown University, Providence, RI, 02912, USA
| | - David J Mooney
- Harvard School of Engineering and Applied Sciences, 29 Oxford Street, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, 60 Oxford Street, Suite 403, Cambridge, MA, 02138, USA
| |
Collapse
|
33
|
Patsalos A, Pap A, Varga T, Trencsenyi G, Contreras GA, Garai I, Papp Z, Dezso B, Pintye E, Nagy L. In situ macrophage phenotypic transition is affected by altered cellular composition prior to acute sterile muscle injury. J Physiol 2017; 595:5815-5842. [PMID: 28714082 DOI: 10.1113/jp274361] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
KEY POINTS The in situ phenotypic switch of macrophages is delayed in acute injury following irradiation. The combination of bone marrow transplantation and local muscle radiation protection allows for the identification of a myeloid cell contribution to tissue repair. PET-MRI allows monitoring of myeloid cell invasion and metabolism. Altered cellular composition prior to acute sterile injury affects the in situ phenotypic transition of invading myeloid cells to repair macrophages. There is reciprocal intercellular communication between local muscle cell compartments, such as PAX7 positive cells, and recruited macrophages during skeletal muscle regeneration. ABSTRACT Skeletal muscle regeneration is a complex interplay between various cell types including invading macrophages. Their recruitment to damaged tissues upon acute sterile injuries is necessary for clearance of necrotic debris and for coordination of tissue regeneration. This highly dynamic process is characterized by an in situ transition of infiltrating monocytes from an inflammatory (Ly6Chigh ) to a repair (Ly6Clow ) macrophage phenotype. The importance of the macrophage phenotypic shift and the cross-talk of the local muscle tissue with the infiltrating macrophages during tissue regeneration upon injury are not fully understood and their study lacks adequate methodology. Here, using an acute sterile skeletal muscle injury model combined with irradiation, bone marrow transplantation and in vivo imaging, we show that preserved muscle integrity and cell composition prior to the injury is necessary for the repair macrophage phenotypic transition and subsequently for proper and complete tissue regeneration. Importantly, by using a model of in vivo ablation of PAX7 positive cells, we show that this radiosensitive skeletal muscle progenitor pool contributes to macrophage phenotypic transition following acute sterile muscle injury. In addition, local muscle tissue radioprotection by lead shielding during irradiation preserves normal macrophage transition dynamics and subsequently muscle tissue regeneration. Taken together, our data suggest the existence of a more extensive and reciprocal cross-talk between muscle tissue compartments, including satellite cells, and infiltrating myeloid cells upon tissue damage. These interactions shape the macrophage in situ phenotypic shift, which is indispensable for normal muscle tissue repair dynamics.
Collapse
Affiliation(s)
- Andreas Patsalos
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Attila Pap
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | - Tamas Varga
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary
| | | | - Gerardo Alvarado Contreras
- Division of Clinical Physiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Zoltan Papp
- Division of Clinical Physiology, Institute of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balazs Dezso
- Department of Pathology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eva Pintye
- Department of Radiotherapy, Institute of Oncology, University of Debrecen, Debrecen, Hungary
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, University of Debrecen, Debrecen, H-4032, Hungary.,MTA-DE 'Lendület' Immunogenomics Research Group, University of Debrecen, Debrecen, Hungary.,Sanford-Burnham-Prebys Medical Discovery Institute at Lake Nona, Orlando, FL, USA
| |
Collapse
|
34
|
The effect of calorie restriction on mouse skeletal muscle is sex, strain and time-dependent. Sci Rep 2017; 7:5160. [PMID: 28698572 PMCID: PMC5505993 DOI: 10.1038/s41598-017-04896-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/22/2017] [Indexed: 01/07/2023] Open
Abstract
Loss of skeletal muscle mass and function occurs with increasing age. Calorie restriction (CR) increases the lifespan of C57Bl/6 mice, but not in the shorter-lived DBA/2 strain. There is some evidence that calorie restriction reduces or delays many of the age-related defects that occur in rodent skeletal muscle. We therefore investigated the effect of short (2.5 month) and longer term (8.5 and 18.5 months) CR on skeletal muscle in male and female C57Bl/6 and DBA/2 mice. We found that short-term CR increased the satellite cell number and collagen VI content of muscle, but resulted in a delayed regenerative response to injury.Consistent with this, the in vitro proliferation of satellite cells derived from these muscles was reduced by CR. The percentage of stromal cells, macrophages, hematopoietic stem cells and fibroadipogenic cells in the mononucleated cell population derived from skeletal muscle was reduced by CR at various stages. But overall, these changes are neither consistent over time, nor between strain and sex. The fact that changes induced by CR do not persist with time and the dissimilarities between the two mouse strains, combined with sex differences, urge caution in applying CR to improve skeletal muscle function across the lifespan in humans.
Collapse
|
35
|
Human myogenic reserve cells are quiescent stem cells that contribute to muscle regeneration after intramuscular transplantation in immunodeficient mice. Sci Rep 2017; 7:3462. [PMID: 28615691 PMCID: PMC5471254 DOI: 10.1038/s41598-017-03703-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/03/2017] [Indexed: 11/08/2022] Open
Abstract
Satellite cells, localized within muscles in vivo, are Pax7+ muscle stem cells supporting skeletal muscle growth and regeneration. Unfortunately, their amplification in vitro, required for their therapeutic use, is associated with reduced regenerative potential. In the present study, we investigated if human myogenic reserve cells (MRC) obtained in vitro, represented a reliable cell source for muscle repair. For this purpose, primary human myoblasts were freshly isolated and expanded. After 2 days of differentiation, 62 ± 2.9% of the nuclei were localized in myotubes and 38 ± 2.9% in the mononucleated non-fusing MRC. Eighty percent of freshly isolated human MRC expressed a phenotype similar to human quiescent satellite cells (CD56+/Pax7+/MyoD−/Ki67− cells). Fourteen days and 21 days after cell transplantation in immunodeficient mice, live human cells were significantly more numerous and the percentage of Pax7+/human lamin A/C+ cells was 2 fold higher in muscles of animals injected with MRC compared to those injected with human myoblasts, despite that percentage of spectrin+ and lamin A/C+ human fibers in both groups MRC were similar. Taken together, these data provide evidence that MRC generated in vitro represent a promising source of cells for improving regeneration of injured skeletal muscles.
Collapse
|
36
|
Obestatin controls skeletal muscle fiber-type determination. Sci Rep 2017; 7:2137. [PMID: 28522824 PMCID: PMC5437042 DOI: 10.1038/s41598-017-02337-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 04/11/2017] [Indexed: 01/27/2023] Open
Abstract
Obestatin/GPR39 signaling stimulates skeletal muscle growth and repair by inducing both G-protein-dependent and -independent mechanisms linking the activated GPR39 receptor with distinct sets of accessory and effector proteins. In this work, we describe a new level of activity where obestatin signaling plays a role in the formation, contractile properties and metabolic profile of skeletal muscle through determination of oxidative fiber type. Our data indicate that obestatin regulates Mef2 activity and PGC-1α expression. Both mechanisms result in a shift in muscle metabolism and function. The increase in Mef2 and PGC-1α signaling activates oxidative capacity, whereas Akt/mTOR signaling positively regulates myofiber growth. Taken together, these data indicate that the obestatin signaling acts on muscle fiber-type program in skeletal muscle.
Collapse
|
37
|
Kufeld M, Escobar H, Marg A, Pasemann D, Budach V, Spuler S. Localized irradiation of mouse legs using an image-guided robotic linear accelerator. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:156. [PMID: 28480192 DOI: 10.21037/atm.2017.03.23] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND To investigate the potential of human satellite cells in muscle regeneration small animal models are useful to evaluate muscle regeneration. To suppress the inherent regeneration ability of the tibialis muscle of mice before transplantation of human muscle fibers, a localized irradiation of the mouse leg should be conducted. We analyzed the feasibility of an image-guided robotic irradiation procedure, a routine treatment method in radiation oncology, for the focal irradiation of mouse legs. METHODS After conducting a planning computed tomography (CT) scan of one mouse in its customized mold a three-dimensional dose plan was calculated using a dedicated planning workstation. 18 Gy have been applied to the right anterior tibial muscle of 4 healthy and 12 mice with immune defect in general anesthesia using an image-guided robotic linear accelerator (LINAC). The mice were fixed in a customized acrylic mold with attached fiducial markers for image guided tracking. RESULTS All 16 mice could be irradiated as prevised without signs of acute radiation toxicity or anesthesiological side effects. The animals survived until scarification after 8, 21 and 49 days as planned. The procedure was straight forward and the irradiation process took 5 minutes to apply the dose of 18 Gy. CONCLUSIONS Localized irradiation of mice legs using a robotic LINAC could be conducted as planned. It is a feasible procedure without recognizable side effects. Image guidance offers precise dose delivery and preserves adjacent body parts and tissues.
Collapse
Affiliation(s)
- Markus Kufeld
- Charité CyberKnife Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Helena Escobar
- Max Delbrück Center for Molecular Medicine, Mobile DNA group, Berlin, Germany
| | - Andreas Marg
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Diana Pasemann
- Charité CyberKnife Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
38
|
Pini V, Morgan JE, Muntoni F, O’Neill HC. Genome Editing and Muscle Stem Cells as a Therapeutic Tool for Muscular Dystrophies. CURRENT STEM CELL REPORTS 2017; 3:137-148. [PMID: 28616376 PMCID: PMC5445179 DOI: 10.1007/s40778-017-0076-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Purpose of Review Muscular dystrophies are a group of severe degenerative disorders characterized by muscle fiber degeneration and death. Therapies designed to restore muscle homeostasis and to replace dying fibers are being experimented, but none of those in clinical trials are suitable to permanently address individual gene mutation. The purpose of this review is to discuss genome editing tools such as CRISPR/Cas (clustered regularly interspaced short palindromic repeats/CRISPR-associated), which enable direct sequence alteration and could potentially be adopted to correct the genetic defect leading to muscle impairment. Recent Findings Recent findings show that advances in gene therapy, when combined with traditional viral vector-based approaches, are bringing the field of regenerative medicine closer to precision-based medicine. Summary The use of such programmable nucleases is proving beneficial for the creation of more accurate in vitro and in vivo disease models. Several gene and cell-therapy studies have been performed on satellite cells, the primary skeletal muscle stem cells involved in muscle regeneration. However, these have mainly been based on artificial replacement or augmentation of the missing protein. Satellite cells are a particularly appealing target to address these innovative technologies for the treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Veronica Pini
- Molecular and Developmental Neurosciences Program, The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Jennifer E. Morgan
- Molecular and Developmental Neurosciences Program, The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Francesco Muntoni
- Molecular and Developmental Neurosciences Program, The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Helen C. O’Neill
- Embryology, IVF and Reproductive Genetics Group, Institute for Women’s Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX UK
| |
Collapse
|
39
|
Garcia SM, Tamaki S, Xu X, Pomerantz JH. Human Satellite Cell Isolation and Xenotransplantation. Methods Mol Biol 2017; 1668:105-123. [PMID: 28842905 DOI: 10.1007/978-1-4939-7283-8_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Satellite cells are mononucleated cells of the skeletal muscle lineage that exist beneath the basal lamina juxtaposed to the sarcolemma of skeletal muscle fibers. It is widely accepted that satellite cells mediate skeletal muscle regeneration. Within the satellite cell pool of adult muscle are skeletal muscle stem cells (MuSCs), also called satellite stem cells, which fulfill criteria of tissue stem cells: They proliferate and their progeny either occupies the adult MuSC niche during self-renewal or differentiates to regenerate mature muscle fibers. Here, we describe robust methods for the isolation of enriched populations of human satellite cells containing MuSCs from fresh human muscle, utilizing mechanical and enzymatic dissociation and purification by fluorescence-activated cell sorting. We also describe a process for xenotransplantation of human satellite cells into mouse muscle by injection into irradiated, immunodeficient, mouse leg muscle with concurrent notexin or bupivacaine muscle injury to increase engraftment efficiency. The engraftment of human MuSCs and the formation of human muscle can then be analyzed by histological and immunofluorescence staining, or subjected to in vivo experimentation.
Collapse
Affiliation(s)
- Steven M Garcia
- Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA.,Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Orofacial Sciences, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Stanley Tamaki
- Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA.,Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Orofacial Sciences, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Xiaoti Xu
- Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA.,Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Orofacial Sciences, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Jason H Pomerantz
- Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA. .,Program in Craniofacial Biology, Division of Plastic and Reconstructive Surgery, Department of Orofacial Sciences, Eli and Edythe Broad Center of Regeneration Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
40
|
Southard S, Kim JR, Low S, Tsika RW, Lepper C. Myofiber-specific TEAD1 overexpression drives satellite cell hyperplasia and counters pathological effects of dystrophin deficiency. eLife 2016; 5. [PMID: 27725085 PMCID: PMC5059137 DOI: 10.7554/elife.15461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 09/17/2016] [Indexed: 12/20/2022] Open
Abstract
When unperturbed, somatic stem cells are poised to affect immediate tissue restoration upon trauma. Yet, little is known regarding the mechanistic basis controlling initial and homeostatic ‘scaling’ of stem cell pool sizes relative to their target tissues for effective regeneration. Here, we show that TEAD1-expressing skeletal muscle of transgenic mice features a dramatic hyperplasia of muscle stem cells (i.e. satellite cells, SCs) but surprisingly without affecting muscle tissue size. Super-numeral SCs attain a ‘normal’ quiescent state, accelerate regeneration, and maintain regenerative capacity over several injury-induced regeneration bouts. In dystrophic muscle, the TEAD1 transgene also ameliorated the pathology. We further demonstrate that hyperplastic SCs accumulate non-cell-autonomously via signal(s) from the TEAD1-expressing myofiber, suggesting that myofiber-specific TEAD1 overexpression activates a physiological signaling pathway(s) that determines initial and homeostatic SC pool size. We propose that TEAD1 and its downstream effectors are medically relevant targets for enhancing muscle regeneration and ameliorating muscle pathology. DOI:http://dx.doi.org/10.7554/eLife.15461.001 Skeletal muscles are primarily composed of cells called muscle fibers, which attach to bones via tendons. These muscle fibers contract to help move the body. Muscle also contains a population of muscle stem cells that repair injured tissue. Normally, in adult skeletal muscle, these stems cells are in a resting state. However, upon injury, the stem cells become activated, divide to increase in number and then develop into new muscle fibers to replace those that were damaged. The balance between the number of stem cells and the size of the muscle must be tightly regulated to ensure that there are enough stem cells to fully regenerate the tissue after injury. However, little is known about how tissues keep their number of stem cells in proportion with their overall size. Previous attempts to make mice with more muscle stem cells invariably also created mice with larger muscles overall. This raised the question: is it possible to increase the numbers of stem cells without changing the size of the muscle? Now, Southard, Kim et al. show it is possible and report that mice engineered to overproduce a protein called Tead1 in their muscle fibers have up to 6-times more stem cells yet normally sized muscles. Tead1 is a transcription factor that controls the activity of a number of genes as part of a major signaling pathway. The stem cells in mice that overproduce Tead1 began to increase in number two weeks after the mice were born because they went through additional rounds of cell division before they entered the resting state. Further experiments then showed that having more stem cells meant that the muscles were repaired more quickly after an injury. Additionally, when mice with extra Tead1 had a mutation that normally leads to muscle wasting, experiments showed that the progression of the disease was stunted. Southard, Kim et al. also show that the muscle fibers that are directly attached to the muscle stem cells are needed for the stem cells to increase in number in the Tead1-overexpressing mice. Together these findings suggest that a signal from the muscle fiber to its stem cells regulates the size of the stem cell population in the tissue. The next challenge is to uncover the molecule (or molecules) that signals from the muscle fiber to the stem cells and to gain deeper insight into how the Tead1 protein can counteract the effects of a muscle wasting disease. DOI:http://dx.doi.org/10.7554/eLife.15461.002
Collapse
Affiliation(s)
- Sheryl Southard
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Ju-Ryoung Kim
- Department of Biochemistry, University of Missouri, Columbia, United States.,School of Medicine, University of Missouri, Columbia, United States.,Department of Biomedical Sciences, University of Missouri, Columbia, United States.,College of Veterinary Medicine, University of Missouri, Columbia, United States
| | - SiewHui Low
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| | - Richard W Tsika
- Department of Biochemistry, University of Missouri, Columbia, United States.,School of Medicine, University of Missouri, Columbia, United States.,Department of Biomedical Sciences, University of Missouri, Columbia, United States.,College of Veterinary Medicine, University of Missouri, Columbia, United States
| | - Christoph Lepper
- Department of Embryology, Carnegie Institution for Science, Baltimore, United States
| |
Collapse
|
41
|
Naldaiz-Gastesi N, Goicoechea M, Alonso-Martín S, Aiastui A, López-Mayorga M, García-Belda P, Lacalle J, San José C, Araúzo-Bravo MJ, Trouilh L, Anton-Leberre V, Herrero D, Matheu A, Bernad A, García-Verdugo JM, Carvajal JJ, Relaix F, Lopez de Munain A, García-Parra P, Izeta A. Identification and Characterization of the Dermal Panniculus Carnosus Muscle Stem Cells. Stem Cell Reports 2016; 7:411-424. [PMID: 27594590 PMCID: PMC5032673 DOI: 10.1016/j.stemcr.2016.08.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 01/05/2023] Open
Abstract
The dermal Panniculus carnosus (PC) muscle is important for wound contraction in lower mammals and represents an interesting model of muscle regeneration due to its high cell turnover. The resident satellite cells (the bona fide muscle stem cells) remain poorly characterized. Here we analyzed PC satellite cells with regard to developmental origin and purported function. Lineage tracing shows that they originate in Myf5+, Pax3/Pax7+ cell populations. Skin and muscle wounding increased PC myofiber turnover, with the satellite cell progeny being involved in muscle regeneration but with no detectable contribution to the wound-bed myofibroblasts. Since hematopoietic stem cells fuse to PC myofibers in the absence of injury, we also studied the contribution of bone marrow-derived cells to the PC satellite cell compartment, demonstrating that cells of donor origin are capable of repopulating the PC muscle stem cell niche after irradiation and bone marrow transplantation but may not fully acquire the relevant myogenic commitment. PC satellite cells originate from Myf5+, Pax3/Pax7+ cell lineages Skin and muscle wounding increase PC myofiber turnover Donor bone marrow cells repopulate the PC satellite niche after BMT Dermis-derived myogenesis originates from the PC satellite cell population
Collapse
Affiliation(s)
- Neia Naldaiz-Gastesi
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - María Goicoechea
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Sonia Alonso-Martín
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Ana Aiastui
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Macarena López-Mayorga
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Paula García-Belda
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaione Lacalle
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; Faculty of Medicine and Nursing, UPV-EHU, San Sebastián 20014, Spain
| | - Carlos San José
- Animal Facility and Experimental Surgery, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Instituto Biodonostia, San Sebastián 20014, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain
| | - Lidwine Trouilh
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Véronique Anton-Leberre
- INSA, UPS, INP, LISBP, Université de Toulouse, 31077 Toulouse, France; INRA, UMR792, Ingénierie des Systèmes Biologiques et des Procédés, 31400 Toulouse, France; CNRS, UMR5504, 31400 Toulouse, France
| | - Diego Herrero
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - Ander Matheu
- IKERBASQUE, Basque Foundation for Science, Bilbao 48013, Spain; Cellular Oncology Group, Oncology Area, Instituto Biodonostia, San Sebastián 20014, Spain
| | - Antonio Bernad
- Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid 28049, Spain
| | - José Manuel García-Verdugo
- CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Laboratorio de Neurobiología Comparada, Instituto Cavanilles, Universidad de Valencia, Valencia 46980, Spain
| | - Jaime J Carvajal
- Molecular Embryology Team, Centro Andaluz de Biología del Desarrollo, Sevilla 41013, Spain
| | - Frédéric Relaix
- INSERM U955-E10, Université Paris Est, Faculté de Médicine, IMRB U955-E10, Creteil 94000, France
| | - Adolfo Lopez de Munain
- Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain; Faculty of Medicine and Nursing, Department of Neurosciences, UPV-EHU, San Sebastián 20014, Spain; Department of Neurology, Hospital Universitario Donostia, San Sebastián 20014, Spain
| | - Patricia García-Parra
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Neuroscience Area, Instituto Biodonostia, San Sebastián 20014, Spain; CIBERNED, Instituto de Salud Carlos III, Madrid 28029, Spain.
| | - Ander Izeta
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián 20014, Spain; Department of Biomedical Engineering, School of Engineering, Tecnun-University of Navarra, San Sebastián 20009, Spain.
| |
Collapse
|
42
|
Walker RG, Poggioli T, Katsimpardi L, Buchanan SM, Oh J, Wattrus S, Heidecker B, Fong YW, Rubin LL, Ganz P, Thompson TB, Wagers AJ, Lee RT. Biochemistry and Biology of GDF11 and Myostatin: Similarities, Differences, and Questions for Future Investigation. Circ Res 2016; 118:1125-41; discussion 1142. [PMID: 27034275 DOI: 10.1161/circresaha.116.308391] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 03/07/2016] [Indexed: 02/06/2023]
Abstract
Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor β superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.
Collapse
Affiliation(s)
- Ryan G Walker
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Tommaso Poggioli
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Lida Katsimpardi
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Sean M Buchanan
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Juhyun Oh
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Sam Wattrus
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Bettina Heidecker
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Yick W Fong
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Lee L Rubin
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Peter Ganz
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Thomas B Thompson
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.)
| | - Amy J Wagers
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.).
| | - Richard T Lee
- From the Department of Molecular Genetics, College of Medicine, University of Cincinnati, OH (R.G.W., T.B.T.); Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA (T.P., L.K., S.M.B., J.O., S.W., L.L.R., A.J.W., R.T.L.); Department of Neuroscience, Institut Pasteur, Paris, France (L.K.); Cardiovascular Division (T.P.), Department of Medicine, Brigham Regenerative Medicine Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (Y.W.F., R.T.L.); Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA (J.O., S.W., A.J.W.); Division of Cardiology, Universitäres Herzzentrum, Zürich, Switzerland (B.H.); Department of Medicine, University of California, San Francisco (B.H., P.G.); and Division of Cardiology, San Francisco General Hospital, CA (P.G.).
| |
Collapse
|
43
|
Alonso-Martin S, Rochat A, Mademtzoglou D, Morais J, de Reyniès A, Auradé F, Chang THT, Zammit PS, Relaix F. Gene Expression Profiling of Muscle Stem Cells Identifies Novel Regulators of Postnatal Myogenesis. Front Cell Dev Biol 2016; 4:58. [PMID: 27446912 PMCID: PMC4914952 DOI: 10.3389/fcell.2016.00058] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 06/02/2016] [Indexed: 01/02/2023] Open
Abstract
Skeletal muscle growth and regeneration require a population of muscle stem cells, the satellite cells, located in close contact to the myofiber. These cells are specified during fetal and early postnatal development in mice from a Pax3/7 population of embryonic progenitor cells. As little is known about the genetic control of their formation and maintenance, we performed a genome-wide chronological expression profile identifying the dynamic transcriptomic changes involved in establishment of muscle stem cells through life, and acquisition of muscle stem cell properties. We have identified multiple genes and pathways associated with satellite cell formation, including set of genes specifically induced (EphA1, EphA2, EfnA1, EphB1, Zbtb4, Zbtb20) or inhibited (EphA3, EphA4, EphA7, EfnA2, EfnA3, EfnA4, EfnA5, EphB2, EphB3, EphB4, EfnBs, Zfp354c, Zcchc5, Hmga2) in adult stem cells. Ephrin receptors and ephrins ligands have been implicated in cell migration and guidance in many tissues including skeletal muscle. Here we show that Ephrin receptors and ephrins ligands are also involved in regulating the adult myogenic program. Strikingly, impairment of EPHB1 function in satellite cells leads to increased differentiation at the expense of self-renewal in isolated myofiber cultures. In addition, we identified new transcription factors, including several zinc finger proteins. ZFP354C and ZCCHC5 decreased self-renewal capacity when overexpressed, whereas ZBTB4 increased it, and ZBTB20 induced myogenic progression. The architectural and transcriptional regulator HMGA2 was involved in satellite cell activation. Together, our study shows that transcriptome profiling coupled with myofiber culture analysis, provides an efficient system to identify and validate candidate genes implicated in establishment/maintenance of muscle stem cells. Furthermore, tour de force transcriptomic profiling provides a wealth of data to inform for future stem cell-based muscle therapies.
Collapse
Affiliation(s)
- Sonia Alonso-Martin
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10Créteil, France; Université Paris Est, Faculté de MedecineCréteil, France; Ecole Nationale Veterinaire d'AlfortMaison Alfort, France
| | - Anne Rochat
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10 Créteil, France
| | - Despoina Mademtzoglou
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10Créteil, France; Université Paris Est, Faculté de MedecineCréteil, France; Ecole Nationale Veterinaire d'AlfortMaison Alfort, France
| | - Jessica Morais
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10 Créteil, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer Paris, France
| | - Frédéric Auradé
- Sorbonne Universités, UPMC Univ Paris 06, INSERM UMRS974, Center for Research in Myology Paris, France
| | - Ted Hung-Tse Chang
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10 Créteil, France
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London London, UK
| | - Frédéric Relaix
- Institut Mondor de Recherche Biomédicale, INSERM U955-E10Créteil, France; Université Paris Est, Faculté de MedecineCréteil, France; Ecole Nationale Veterinaire d'AlfortMaison Alfort, France; Etablissement Français du SangCréteil, France; APHP, Hopitaux Universitaires Henri Mondor, DHU Pepsy and Centre de Référence des Maladies Neuromusculaires GNMHCréteil, France
| |
Collapse
|
44
|
Muscle Satellite Cells: Exploring the Basic Biology to Rule Them. Stem Cells Int 2016; 2016:1078686. [PMID: 27042182 PMCID: PMC4794588 DOI: 10.1155/2016/1078686] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/24/2016] [Indexed: 12/12/2022] Open
Abstract
Adult skeletal muscle is a postmitotic tissue with an enormous capacity to regenerate upon injury. This is accomplished by resident stem cells, named satellite cells, which were identified more than 50 years ago. Since their discovery, many researchers have been concentrating efforts to answer questions about their origin and role in muscle development, the way they contribute to muscle regeneration, and their potential to cell-based therapies. Satellite cells are maintained in a quiescent state and upon requirement are activated, proliferating, and fusing with other cells to form or repair myofibers. In addition, they are able to self-renew and replenish the stem pool. Every phase of satellite cell activity is highly regulated and orchestrated by many molecules and signaling pathways; the elucidation of players and mechanisms involved in satellite cell biology is of extreme importance, being the first step to expose the crucial points that could be modulated to extract the optimal response from these cells in therapeutic strategies. Here, we review the basic aspects about satellite cells biology and briefly discuss recent findings about therapeutic attempts, trying to raise questions about how basic biology could provide a solid scaffold to more successful use of these cells in clinics.
Collapse
|
45
|
Neuromuscular electrical stimulation promotes development in mice of mature human muscle from immortalized human myoblasts. Skelet Muscle 2016; 6:4. [PMID: 26925213 PMCID: PMC4769538 DOI: 10.1186/s13395-016-0078-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 01/06/2016] [Indexed: 12/25/2022] Open
Abstract
Background Studies of the pathogenic mechanisms underlying human myopathies and muscular dystrophies often require animal models, but models of some human diseases are not yet available. Methods to promote the engraftment and development of myogenic cells from individuals with such diseases in mice would accelerate such studies and also provide a useful tool for testing therapeutics. Here, we investigate the ability of immortalized human myogenic precursor cells (hMPCs) to form mature human myofibers following implantation into the hindlimbs of non-obese diabetic-Rag1nullIL2rγnull (NOD-Rag)-immunodeficient mice. Results We report that hindlimbs of NOD-Rag mice that are X-irradiated, treated with cardiotoxin, and then injected with immortalized control hMPCs or hMPCs from an individual with facioscapulohumeral muscular dystrophy (FSHD) develop mature human myofibers. Furthermore, intermittent neuromuscular electrical stimulation (iNMES) of the peroneal nerve of the engrafted limb enhances the development of mature fibers in the grafts formed by both immortal cell lines. With control cells, iNMES increases the number and size of the human myofibers that form and promotes closer fiber-to-fiber packing. The human myofibers in the graft are innervated, fully differentiated, and minimally contaminated with murine myonuclei. Conclusions Our results indicate that control and FSHD human myofibers can form in mice engrafted with hMPCs and that iNMES enhances engraftment and subsequent development of mature human muscle.
Collapse
|
46
|
Santos-Zas I, Gurriarán-Rodríguez U, Cid-Díaz T, Figueroa G, González-Sánchez J, Bouzo-Lorenzo M, Mosteiro CS, Señarís J, Casanueva FF, Casabiell X, Gallego R, Pazos Y, Mouly V, Camiña JP. β-Arrestin scaffolds and signaling elements essential for the obestatin/GPR39 system that determine the myogenic program in human myoblast cells. Cell Mol Life Sci 2016; 73:617-35. [PMID: 26211463 PMCID: PMC11108386 DOI: 10.1007/s00018-015-1994-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/08/2015] [Accepted: 07/16/2015] [Indexed: 12/27/2022]
Abstract
Obestatin/GPR39 signaling stimulates skeletal muscle repair by inducing the expansion of satellite stem cells as well as myofiber hypertrophy. Here, we describe that the obestatin/GPR39 system acts as autocrine/paracrine factor on human myogenesis. Obestatin regulated multiple steps of myogenesis: myoblast proliferation, cell cycle exit, differentiation and recruitment to fuse and form multinucleated hypertrophic myotubes. Obestatin-induced mitogenic action was mediated by ERK1/2 and JunD activity, being orchestrated by a G-dependent mechanism. At a later stage of myogenesis, scaffolding proteins β-arrestin 1 and 2 were essential for the activation of cell cycle exit and differentiation through the transactivation of the epidermal growth factor receptor (EGFR). Upon obestatin stimulus, β-arrestins are recruited to the membrane, where they functionally interact with GPR39 leading to Src activation and signalplex formation to EGFR transactivation by matrix metalloproteinases. This signalplex regulated the mitotic arrest by p21 and p57 expression and the mid- to late stages of differentiation through JNK/c-Jun, CAMKII, Akt and p38 pathways. This finding not only provides the first functional activity for β-arrestins in myogenesis but also identify potential targets for therapeutic approaches by triggering specific signaling arms of the GPR39 signaling involved in myogenesis.
Collapse
Affiliation(s)
- Icía Santos-Zas
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Uxía Gurriarán-Rodríguez
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- Sprott Centre for Stem Cell Research, Ottawa Health Research Institute, Ottawa, Canada
| | - Tania Cid-Díaz
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Gabriela Figueroa
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
| | - Jessica González-Sánchez
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Mónica Bouzo-Lorenzo
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Carlos S Mosteiro
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - José Señarís
- Servicio de Cirugía Ortopédica y Traumatología, CHUS, SERGAS, Santiago de Compostela, Spain
| | - Felipe F Casanueva
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
- Departamento de Medicina, USC, Santiago de Compostela, Spain
| | - Xesús Casabiell
- Departamento de Fisiología, USC, Santiago de Compostela, Spain
| | - Rosalía Gallego
- Departamento de Ciencias Morfológicas, USC, Santiago de Compostela, Spain
| | - Yolanda Pazos
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain
| | - Vincent Mouly
- Institut de Myologie, INSERM, and Sorbonne Universités, Université Pierre et Marie Curie, Paris, France
| | - Jesús P Camiña
- Área de Endocrinología Molecular y Celular, Instituto de Investigación Sanitaria de Santiago (IDIS), Complejo Hospitalario Universitario de Santiago (CHUS), Servicio Gallego de Salud (SERGAS), Santiago de Compostela, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Santiago de Compostela, Spain.
| |
Collapse
|
47
|
Escobar H, Schöwel V, Spuler S, Marg A, Izsvák Z. Full-length Dysferlin Transfer by the Hyperactive Sleeping Beauty Transposase Restores Dysferlin-deficient Muscle. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e277. [PMID: 26784637 PMCID: PMC5012550 DOI: 10.1038/mtna.2015.52] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 11/13/2015] [Indexed: 12/18/2022]
Abstract
Dysferlin-deficient muscular dystrophy is a progressive disease characterized by muscle weakness and wasting for which there is no treatment. It is caused by mutations in DYSF, a large, multiexonic gene that forms a coding sequence of 6.2 kb. Sleeping Beauty (SB) transposon is a nonviral gene transfer vector, already used in clinical trials. The hyperactive SB system consists of a transposon DNA sequence and a transposase protein, SB100X, that can integrate DNA over 10 kb into the target genome. We constructed an SB transposon-based vector to deliver full-length human DYSF cDNA into dysferlin-deficient H2K A/J myoblasts. We demonstrate proper dysferlin expression as well as highly efficient engraftment (>1,100 donor-derived fibers) of the engineered myoblasts in the skeletal muscle of dysferlin- and immunodeficient B6.Cg-Dysf(prmd) Prkdc(scid)/J (Scid/BLA/J) mice. Nonviral gene delivery of full-length human dysferlin into muscle cells, along with a successful and efficient transplantation into skeletal muscle are important advances towards successful gene therapy of dysferlin-deficient muscular dystrophy.
Collapse
Affiliation(s)
- Helena Escobar
- Mobile DNA, Max Delbrück Center for Molecular Medicine of the Helmholtz Society, Berlin, Germany
| | - Verena Schöwel
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité, Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité, Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Andreas Marg
- Muscle Research Unit, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité, Universitätsmedizin Berlin and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Zsuzsanna Izsvák
- Mobile DNA, Max Delbrück Center for Molecular Medicine of the Helmholtz Society, Berlin, Germany
| |
Collapse
|
48
|
Meng J, Bencze M, Asfahani R, Muntoni F, Morgan JE. The effect of the muscle environment on the regenerative capacity of human skeletal muscle stem cells. Skelet Muscle 2015; 5:11. [PMID: 25949786 PMCID: PMC4422426 DOI: 10.1186/s13395-015-0036-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 03/12/2015] [Indexed: 12/31/2022] Open
Abstract
Background Muscle stem cell transplantation is a possible treatment for muscular dystrophy. In addition to the intrinsic properties of the stem cells, the local and systemic environment plays an important role in determining the fate of the grafted cells. We therefore investigated the effect of modulating the host muscle environment in different ways (irradiation or cryoinjury or a combination of irradiation and cryoinjury) in two immunodeficient mouse strains (mdx nude and recombinase-activating gene (Rag)2-/γ chain-/C5-) on the regenerative capacity of two types of human skeletal muscle-derived stem cell (pericytes and CD133+ cells). Methods Human skeletal muscle-derived pericytes or CD133+ cells were transplanted into muscles of either mdx nude or recombinase-activating gene (Rag)2-/γ chain-/C5- host mice. Host muscles were modulated prior to donor cell transplantation by either irradiation, or cryoinjury, or a combination of irradiation and cryoinjury. Muscles were analysed four weeks after transplantation, by staining transverse cryostat sections of grafted muscles with antibodies to human lamin A/C, human spectrin, laminin and Pax 7. The number of nuclei and muscle fibres of donor origin and the number of satellite cells of both host and donor origin were quantified. Results Within both host strains transplanted intra-muscularly with both donor cell types, there were significantly more nuclei and muscle fibres of donor origin in host muscles that had been modulated by cryoinjury, or irradiation+cryoinjury, than by irradiation alone. Irradiation has no additive effects in further enhancing the transplantation efficiency than cryodamage. Donor pericytes did not give rise to satellite cells. However, using CD133+ cells as donor cells, there were significantly more nuclei, muscle fibres, as well as satellite cells of donor origin in Rag2-/γ chain-/C5- mice than mdx nude mice, when the muscles were injured by either cryodamage or irradiation+cryodamage. Conclusions Rag2-/γ chain-/C5- mice are a better recipient mouse strain than mdx nude mice for human muscle stem cell transplantation. Cryodamage of host muscle is the most effective method to enhance the transplantation efficiency of human skeletal muscle stem cells. This study highlights the importance of modulating the muscle environment in preclinical studies to optimise the efficacy of transplanted stem cells. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0036-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jinhong Meng
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Maximilien Bencze
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Rowan Asfahani
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| | - Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Institute of Child Health, 30 Guilford Street, London, WC1N 1EH UK
| |
Collapse
|
49
|
Nik-Ahd F, Bertoni C. Ex vivo gene editing of the dystrophin gene in muscle stem cells mediated by peptide nucleic acid single stranded oligodeoxynucleotides induces stable expression of dystrophin in a mouse model for Duchenne muscular dystrophy. Stem Cells 2015; 32:1817-30. [PMID: 24753122 DOI: 10.1002/stem.1668] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/07/2014] [Accepted: 01/26/2014] [Indexed: 12/27/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disease caused by mutations in the dystrophin gene, which result in the complete absence of dystrophin protein throughout the body. Gene correction strategies hold promise to treating DMD. Our laboratory has previously demonstrated the ability of peptide nucleic acid single-stranded oligodeoxynucleotides (PNA-ssODNs) to permanently correct single-point mutations at the genomic level. In this study, we show that PNA-ssODNs can target and correct muscle satellite cells (SCs), a population of stem cells capable of self-renewing and differentiating into muscle fibers. When transplanted into skeletal muscles, SCs transfected with correcting PNA-ssODNs were able to engraft and to restore dystrophin expression. The number of dystrophin-positive fibers was shown to significantly increase over time. Expression was confirmed to be the result of the activation of a subpopulation of SCs that had undergone repair as demonstrated by immunofluorescence analyses of engrafted muscles using antibodies specific to full-length dystrophin transcripts and by genomic DNA analysis of dystrophin-positive fibers. Furthermore, the increase in dystrophin expression detected over time resulted in a significant improvement in muscle morphology. The ability of transplanted cells to return into quiescence and to activate upon demand was confirmed in all engrafted muscles following injury. These results demonstrate the feasibility of using gene editing strategies to target and correct SCs and further establish the therapeutic potential of this approach to permanently restore dystrophin expression into muscle of DMD patients.
Collapse
Affiliation(s)
- Farnoosh Nik-Ahd
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | | |
Collapse
|
50
|
Partridge TA, Morgan JE. Multiple insights from myogenic cell transplants. Hum Gene Ther 2014; 25:404-5. [PMID: 24848316 DOI: 10.1089/hum.2014.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|