Muscle Stem/Progenitor Cells and Mesenchymal Stem Cells of Bone Marrow Origin for Skeletal Muscle Regeneration in Muscular Dystrophies

[1]  Yoshiya Tanaka,et al.  Induction of Regulatory T Cells and Its Regulation with Insulin-like Growth Factor/Insulin-like Growth Factor Binding Protein-4 by Human Mesenchymal Stem Cells , 2017, The Journal of Immunology.

[2]  E. Pintye,et al.  In situ macrophage phenotypic transition is affected by altered cellular composition prior to acute sterile muscle injury , 2017, The Journal of physiology.

[3]  F. Lewis,et al.  Skeletal muscle-derived interstitial progenitor cells (PICs) display stem cell properties, being clonogenic, self-renewing, and multi-potent in vitro and in vivo , 2017, Stem Cell Research & Therapy.

[4]  P. Hoffmeyer,et al.  Human myogenic reserve cells are quiescent stem cells that contribute to muscle regeneration after intramuscular transplantation in immunodeficient mice , 2017, Scientific Reports.

[5]  Y. Maeda,et al.  CXCL12 and osteopontin from bone marrow-derived mesenchymal stromal cells improve muscle regeneration , 2017, Scientific Reports.

[6]  F. Muntoni,et al.  Genome Editing and Muscle Stem Cells as a Therapeutic Tool for Muscular Dystrophies , 2017, Current Stem Cell Reports.

[7]  A. Boccaccini,et al.  Mesenchymal stem cells and myoblast differentiation under HGF and IGF-1 stimulation for 3D skeletal muscle tissue engineering , 2017, BMC Cell Biology.

[8]  C. Vogelmeier,et al.  Myocardial homing of mesenchymal stem cells following intrapericardial application and amplification by inflammation - an experimental pilot study. , 2017, Canadian journal of physiology and pharmacology.

[9]  Colin Crist Emerging new tools to study and treat muscle pathologies: genetics and molecular mechanisms underlying skeletal muscle development, regeneration, and disease , 2017, The Journal of pathology.

[10]  M. Sormani,et al.  Intra‐arterial transplantation of HLA‐matched donor mesoangioblasts in Duchenne muscular dystrophy , 2016, EMBO molecular medicine.

[11]  Attila Horvath,et al.  Macrophage PPARγ, a Lipid Activated Transcription Factor Controls the Growth Factor GDF3 and Skeletal Muscle Regeneration. , 2016, Immunity.

[12]  F. Rossi,et al.  Loss of niche-satellite cell interactions in syndecan-3 null mice alters muscle progenitor cell homeostasis improving muscle regeneration. , 2016, Skeletal muscle.

[13]  Nicola Elvassore,et al.  Skeletal Muscle Differentiation on a Chip Shows Human Donor Mesoangioblasts’ Efficiency in Restoring Dystrophin in a Duchenne Muscular Dystrophy Model , 2016, Stem cells translational medicine.

[14]  F. Muntoni,et al.  Autologous skeletal muscle derived cells expressing a novel functional dystrophin provide a potential therapy for Duchenne Muscular Dystrophy , 2016, Scientific Reports.

[15]  A. Klimczak,et al.  Mesenchymal Stromal Cells and Tissue-Specific Progenitor Cells: Their Role in Tissue Homeostasis , 2015, Stem cells international.

[16]  Yu Xin Wang,et al.  Dystrophin expression in muscle stem cells regulates their polarity and asymmetric division , 2015, Nature Medicine.

[17]  Radbod Darabi,et al.  Skeletal muscle perfusion and stem cell delivery in muscle disorders using intra-femoral artery canulation in mice. , 2015, Experimental cell research.

[18]  Michael D. Schneider,et al.  Monocyte/Macrophage-derived IGF-1 Orchestrates Murine Skeletal Muscle Regeneration and Modulates Autocrine Polarization. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[19]  Albert E. Almada,et al.  FOXP3+ T Cells Recruited to Sites of Sterile Skeletal Muscle Injury Regulate the Fate of Satellite Cells and Guide Effective Tissue Regeneration , 2015, PloS one.

[20]  T. Terachi,et al.  Therapeutic isolation and expansion of human skeletal muscle-derived stem cells for the use of muscle-nerve-blood vessel reconstitution , 2015, Front. Physiol..

[21]  J. Galipeau,et al.  Bone Marrow Mesenchymal Stromal Cells from Patients with Acute and Chronic Graft-versus-Host Disease Deploy Normal Phenotype, Differentiation Plasticity, and Immune-Suppressive Activity. , 2015, Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation.

[22]  J. Morgan,et al.  Satellite cells from dystrophic muscle retain regenerative capacity , 2015, Stem cell research.

[23]  B. Wang,et al.  Rapid depletion of muscle progenitor cells in dystrophic mdx/utrophin-/- mice. , 2014, Human molecular genetics.

[24]  M. Messi,et al.  Pericytes: multitasking cells in the regeneration of injured, diseased, and aged skeletal muscle , 2014, Front. Aging Neurosci..

[25]  K. English,et al.  Stem Cell Transplantation for Muscular Dystrophy: The Challenge of Immune Response , 2014, BioMed research international.

[26]  M. Bouché,et al.  From Innate to Adaptive Immune Response in Muscular Dystrophies and Skeletal Muscle Regeneration: The Role of Lymphocytes , 2014, BioMed research international.

[27]  P. Rovere-Querini,et al.  Macrophage Plasticity in Skeletal Muscle Repair , 2014, BioMed research international.

[28]  F. Dilworth,et al.  Dystrophic muscle environment induces changes in cell plasticity , 2014, Genes & development.

[29]  J. Rossant,et al.  HDAC-regulated myomiRs control BAF60 variant exchange and direct the functional phenotype of fibro-adipogenic progenitors in dystrophic muscles , 2014, Genes & development.

[30]  L. McManus,et al.  Altered macrophage phenotype transition impairs skeletal muscle regeneration. , 2014, The American journal of pathology.

[31]  J. Sluijter,et al.  Mesenchymal Stem Cell Therapy for Cardiac Inflammation: Immunomodulatory Properties and the Influence of Toll-Like Receptors , 2013, Mediators of inflammation.

[32]  J. Zwaginga,et al.  The angiogenic properties of mesenchymal stem/stromal cells and their therapeutic potential. , 2013, British medical bulletin.

[33]  Heather D. Huntsman,et al.  Defining a role for non-satellite stem cells in the regulation of muscle repair following exercise , 2013, Front. Physiol..

[34]  A. Caplan,et al.  Mesenchymal stem cells: environmentally responsive therapeutics for regenerative medicine , 2013, Experimental & Molecular Medicine.

[35]  F. Rossi,et al.  Tissue‐resident mesenchymal stem/progenitor cells in skeletal muscle: collaborators or saboteurs? , 2013, The FEBS journal.

[36]  G. Enikolopov,et al.  Role of pericytes in skeletal muscle regeneration and fat accumulation. , 2013, Stem cells and development.

[37]  O. Cappellari,et al.  Pericytes in Development and Pathology of Skeletal Muscle , 2013, Circulation research.

[38]  D. Sassoon,et al.  Defining skeletal muscle resident progenitors and their cell fate potentials , 2013, Development.

[39]  Rahul C. Deo,et al.  Type 2 Innate Signals Stimulate Fibro/Adipogenic Progenitors to Facilitate Muscle Regeneration , 2013, Cell.

[40]  T. Lue,et al.  Defining vascular stem cells. , 2013, Stem cells and development.

[41]  G. Borsellino,et al.  Fibroadipogenic progenitors mediate the ability of HDAC inhibitors to promote regeneration in dystrophic muscles of young, but not old Mdx mice , 2013, EMBO molecular medicine.

[42]  H. Northoff,et al.  Mesenchymal stromal/stem cells markers in the human bone marrow. , 2013, Cytotherapy.

[43]  M. Kjaer,et al.  Differentially Activated Macrophages Orchestrate Myogenic Precursor Cell Fate During Human Skeletal Muscle Regeneration , 2013, Stem cells.

[44]  R. Bellamkonda,et al.  Effect of modulating macrophage phenotype on peripheral nerve repair. , 2012, Biomaterials.

[45]  J. Tidball,et al.  IL-10 Triggers Changes in Macrophage Phenotype That Promote Muscle Growth and Regeneration , 2012, The Journal of Immunology.

[46]  B. Kalionis,et al.  Immunosuppressive Properties of Mesenchymal Stem Cells , 2012, Stem Cell Reviews and Reports.

[47]  Yi Lin,et al.  Clinical Application of Mesenchymal Stem Cells in the Treatment and Prevention of Graft-versus-Host Disease , 2011, Advances in hematology.

[48]  K. Tsuchida,et al.  Fibrosis and adipogenesis originate from a common mesenchymal progenitor in skeletal muscle , 2011, Journal of Cell Science.

[49]  K. Davies,et al.  Progress in therapy for Duchenne muscular dystrophy , 2011, Experimental physiology.

[50]  S. Antonini,et al.  Pericytes resident in postnatal skeletal muscle differentiate into muscle fibres and generate satellite cells. , 2011, Nature communications.

[51]  Arnold I Caplan,et al.  The MSC: an injury drugstore. , 2011, Cell stem cell.

[52]  A. Boos,et al.  Myogenic differentiation of mesenchymal stem cells co‐cultured with primary myoblasts , 2011, Cell biology international.

[53]  F. Muntoni,et al.  Stem cells to treat muscular dystrophies – Where are we? , 2011, Neuromuscular Disorders.

[54]  C. Pholpramool,et al.  Satellite cell activity in muscle regeneration after contusion in rats , 2010, Clinical and experimental pharmacology & physiology.

[55]  J. Tidball,et al.  Regulatory interactions between muscle and the immune system during muscle regeneration. , 2010, American journal of physiology. Regulatory, integrative and comparative physiology.

[56]  D. Sassoon,et al.  Identification and characterization of a non-satellite cell muscle resident progenitor during postnatal development , 2010, Nature Cell Biology.

[57]  Michael A. Rudnicki,et al.  Muscle injury activates resident fibro/adipogenic progenitors that facilitate myogenesis , 2010, Nature Cell Biology.

[58]  S. Erratico,et al.  Cell based therapy for duchenne muscular dystrophy , 2009, Journal of cellular physiology.

[59]  P. D. del Nido,et al.  CXCR4 enhances engraftment of muscle progenitor cells , 2009, Muscle & nerve.

[60]  M. Kyba,et al.  Engraftment of mesenchymal stem cells into dystrophin-deficient mice is not accompanied by functional recovery. , 2009, Experimental cell research.

[61]  F. Dazzi,et al.  The Immunosuppressive Properties of Mesenchymal Stem Cells , 2009, Transplantation.

[62]  Philippe Soriano,et al.  Increased PDGFRalpha activation disrupts connective tissue development and drives systemic fibrosis. , 2009, Developmental cell.

[63]  Hulun Li,et al.  Administration of bone marrow stromal cells ameliorates experimental autoimmune myasthenia gravis by altering the balance of Th1/Th2/Th17/Treg cell subsets through the secretion of TGF-β , 2009, Journal of Neuroimmunology.

[64]  A. Caplan,et al.  MSC frequency correlates with blood vessel density in equine adipose tissue. , 2009, Tissue engineering. Part A.

[65]  J. Ryan,et al.  Cell contact, prostaglandin E2 and transforming growth factor beta 1 play non‐redundant roles in human mesenchymal stem cell induction of CD4+CD25Highforkhead box P3+ regulatory T cells , 2008, Clinical and experimental immunology.

[66]  M. Kyba,et al.  Prospective Isolation of Skeletal Muscle Stem Cells with a Pax7 Reporter , 2008, Stem cells.

[67]  M. Post,et al.  The muscle stem cell niche: regulation of satellite cells during regeneration. , 2008, Tissue engineering. Part B, Reviews.

[68]  T. Gotoh,et al.  Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy. , 2008, Human molecular genetics.

[69]  S. Badylak,et al.  A perivascular origin for mesenchymal stem cells in multiple human organs. , 2008, Cell stem cell.

[70]  Arnold I Caplan,et al.  All MSCs are pericytes? , 2008, Cell stem cell.

[71]  James J. Lee,et al.  Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy. , 2008, Human molecular genetics.

[72]  Liwen Chen,et al.  Paracrine Factors of Mesenchymal Stem Cells Recruit Macrophages and Endothelial Lineage Cells and Enhance Wound Healing , 2008, PloS one.

[73]  C. Bönnemann,et al.  Muscle Interstitial Fibroblasts Are the Main Source of Collagen VI Synthesis in Skeletal Muscle: Implications for Congenital Muscular Dystrophy Types Ullrich and Bethlem , 2008, Journal of neuropathology and experimental neurology.

[74]  W. Arap,et al.  A Population of Multipotent CD34-Positive Adipose Stromal Cells Share Pericyte and Mesenchymal Surface Markers, Reside in a Periendothelial Location, and Stabilize Endothelial Networks , 2008, Circulation research.

[75]  M. Sampaolesi,et al.  New therapies for Duchenne muscular dystrophy: challenges, prospects and clinical trials. , 2007, Trends in molecular medicine.

[76]  A. Caplan Adult mesenchymal stem cells for tissue engineering versus regenerative medicine , 2007, Journal of cellular physiology.

[77]  T. Partridge,et al.  Implication of the satellite cell in dystrophic muscle fibrosis: a self-perpetuating mechanism of collagen overproduction. , 2007, American journal of physiology. Cell physiology.

[78]  N. Van Rooijen,et al.  Inflammatory monocytes recruited after skeletal muscle injury switch into antiinflammatory macrophages to support myogenesis , 2007, The Journal of experimental medicine.

[79]  M. Rudnicki,et al.  Stem and progenitor cells in skeletal muscle development, maintenance, and therapy. , 2007, Molecular therapy : the journal of the American Society of Gene Therapy.

[80]  M. Rudnicki,et al.  Stem cell based therapies to treat muscular dystrophy. , 2007, Biochimica et biophysica acta.

[81]  J. Bouchard,et al.  First test of a “high-density injection” protocol for myogenic cell transplantation throughout large volumes of muscles in a Duchenne muscular dystrophy patient: eighteen months follow-up , 2007, Neuromuscular Disorders.

[82]  E. Lam,et al.  Mesenchymal Stem Cells Inhibit Dendritic Cell Differentiation and Function by Preventing Entry Into the Cell Cycle , 2007, Transplantation.

[83]  P. Moreau,et al.  Lung Resection for Invasive Pulmonary Aspergillosis in Neutropenic Patients with Hematologic Malignancies: Long Term Results in Thirty Two Cases. , 2006 .

[84]  N. Bresolin,et al.  VCAM-1 expression on dystrophic muscle vessels has a critical role in the recruitment of human blood-derived CD133+ stem cells after intra-arterial transplantation. , 2006, Blood.

[85]  R. Willemze,et al.  Mesenchymal Stem Cells Inhibit Generation and Function of Both CD34+-Derived and Monocyte-Derived Dendritic Cells1 , 2006, The Journal of Immunology.

[86]  J. Bouchard,et al.  Dystrophin Expression in Muscles of Duchenne Muscular Dystrophy Patients After High-Density Injections of Normal Myogenic Cells , 2006, Journal of neuropathology and experimental neurology.

[87]  A. Bigot,et al.  Myoblast transfer therapy: is there any light at the end of the tunnel? , 2005, Acta myologica : myopathies and cardiomyopathies : official journal of the Mediterranean Society of Myology.

[88]  Simon C Watkins,et al.  Amelioration of laminin-alpha2-deficient congenital muscular dystrophy by somatic gene transfer of miniagrin. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[89]  G. Goldspink Mechanical signals, IGF-I gene splicing, and muscle adaptation. , 2005, Physiology.

[90]  I. Weissman,et al.  Isolation of Adult Mouse Myogenic Progenitors Functional Heterogeneity of Cells within and Engrafting Skeletal Muscle , 2004, Cell.

[91]  S. Bhagavati,et al.  Isolation and enrichment of skeletal muscle progenitor cells from mouse bone marrow. , 2004, Biochemical and biophysical research communications.

[92]  Moustapha Hassan,et al.  Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells , 2004, The Lancet.

[93]  J. Bouchard,et al.  Dystrophin expression in myofibers of Duchenne muscular dystrophy patients following intramuscular injections of normal myogenic cells. , 2004, Molecular therapy : the journal of the American Society of Gene Therapy.

[94]  G. Bassez,et al.  Satellite cells attract monocytes and use macrophages as a support to escape apoptosis and enhance muscle growth , 2003, The Journal of cell biology.

[95]  O. Ringdén,et al.  HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. , 2003, Experimental hematology.

[96]  M. Ratajczak,et al.  Expression of Functional CXCR4 by Muscle Satellite Cells and Secretion of SDF‐1 by Muscle‐Derived Fibroblasts is Associated with the Presence of Both Muscle Progenitors in Bone Marrow and Hematopoietic Stem/Progenitor Cells in Muscles , 2003, Stem cells.

[97]  H. Wiendl,et al.  Antigen processing and presentation in human muscle: cathepsin S is critical for MHC class II expression and upregulated in inflammatory myopathies , 2003, Journal of Neuroimmunology.

[98]  S. Harridge Ageing and local growth factors in muscle , 2003, Scandinavian journal of medicine & science in sports.

[99]  M. Rudnicki,et al.  Myogenic specification of side population cells in skeletal muscle , 2002, The Journal of cell biology.

[100]  J. Tidball,et al.  Helper (CD4(+)) and cytotoxic (CD8(+)) T cells promote the pathology of dystrophin-deficient muscle. , 2001, Clinical immunology.

[101]  L. Tseng-Ong,et al.  Eosinophilia of dystrophin-deficient muscle is promoted by perforin-mediated cytotoxicity by T cell effectors. , 2000, The American journal of pathology.

[102]  D. Skuk,et al.  Progress in myoblast transplantation: a potential treatment of dystrophies , 2000, Microscopy research and technique.

[103]  G. Butler-Browne,et al.  Shorter telomeres in dystrophic muscle consistent with extensive regeneration in young children , 2000, Neuromuscular Disorders.

[104]  S. Gerson,et al.  Phenotypic and functional comparison of cultures of marrow‐derived mesenchymal stem cells (MSCs) and stromal cells , 1998, Journal of cellular physiology.

[105]  H. Blau,et al.  Myoblast implantation in Duchenne muscular dystrophy: The San Francisco study , 1997, Muscle & nerve.

[106]  R. R. Rice,et al.  Myoblast transfer in the treatment of Duchenne's muscular dystrophy. , 1995, The New England journal of medicine.

[107]  K. Campbell,et al.  Deficiency of dystrophin-associated proteins: A common mechanism leading to muscle cell necrosis in severe childhood muscular dystrophies , 1993, Neuromuscular Disorders.

[108]  P. Law,et al.  Cell Transplantation as an Experimental Treatment for Duchenne Muscular Dystrophy , 1993, Cell transplantation.

[109]  George Karpati,et al.  Myoblast transfer in duchenne muscular dystrophy , 1993, Annals of neurology.

[110]  Arnold I. Caplan,et al.  Mesenchymal Stem Cells , 1991, Journal of orthopaedic research : official publication of the Orthopaedic Research Society.

[111]  R. Waterston,et al.  Characterization of dystrophin in muscle-biopsy specimens from patients with Duchenne's or Becker's muscular dystrophy. , 1988, The New England journal of medicine.

[112]  Y. Pouliot,et al.  Expression of immunoreactive major histocompatibility complex products in human skeletal muscles , 1988, Annals of neurology.

[113]  A. Friedenstein,et al.  STROMAL CELLS RESPONSIBLE FOR TRANSFERRING THE MICROENVIRONMENT OF THE HEMOPOIETIC TISSUES: Cloning In Vitro and Retransplantation In Vivo , 1974, Transplantation.

[114]  A. Friedenstein,et al.  Osteogenesis in transplants of bone marrow cells. , 1966, Journal of embryology and experimental morphology.

[115]  Feodor Price,et al.  Satellite cells and the muscle stem cell niche. , 2013, Physiological reviews.

[116]  Yong Li,et al.  Matrix metalloproteinase inhibition negatively affects muscle stem cell behavior. , 2013, International journal of clinical and experimental pathology.

[117]  C. Mann,et al.  Cellular and molecular mechanisms regulating fibrosis in skeletal muscle repair and disease. , 2011, Current topics in developmental biology.

[118]  Fabian Lara,et al.  Mesenchymal stem cells as anti-inflammatories: implications for treatment of Duchenne muscular dystrophy. , 2010, Cellular immunology.

[119]  N. Nardi,et al.  Methodology, biology and clinical applications of mesenchymal stem cells. , 2009, Frontiers in bioscience.

[120]  M. Buckingham,et al.  Regulation of skeletal muscle stem cell behavior by Pax3 and Pax7. , 2008, Cold Spring Harbor symposia on quantitative biology.

[121]  M. Goodell,et al.  Distinct progenitor populations in skeletal muscle are bone marrow derived and exhibit different cell fates during vascular regeneration. , 2003, The Journal of clinical investigation.