Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Med Sci ; 19(2): 353-363, 2022.
Article in English | MEDLINE | ID: mdl-35165521

ABSTRACT

Cultured human skeletal-muscle satellite cells have properties of mesenchymal stem cells (skeletal muscle satellite cell-derived mesenchymal stem cells, SkMSCs) and play anti-inflammatory roles by secreting prostaglandin E2 and hepatocyte growth factor (HGF). To evaluate the utility of SkMSCs in treating liver diseases, we determined whether SkMSCs could ameliorate acute liver and gut inflammation induced by binge ethanol administration. Binge drinking of ethanol led to weight loss in the body and spleen, liver inflammation and steatosis, and increased serum ALT and AST levels (markers of liver injury), along with increased IL-1ß, TNF-α, and iNOS expression levels in mice. However, levels of these binge-drinking-induced indicators were reduced by a single intraperitoneal treatment of SkMSCs. Furthermore, levels of bacteria-derived lipopolysaccharide decreased in the livers and sera of ethanol-exposed mice after SkMSC administration. SkMSCs decreased the extent of tissue inflammation and reduced villus and crypt lengths in the small intestine after alcohol binge drinking. SkMSCs also reduced the leakage of blood albumin, an indicator of leaky gut, in the stool of ethanol-exposed mice. Alcohol-induced damage to human colonic Caco-2/tc7 cells was also alleviated by HGF. Therefore, a single treatment with SkMSCs can attenuate alcoholic liver damage by reducing inflammatory responses in the liver and gut, suggesting that SkMSCs could be used in cell therapy to treat alcoholic liver diseases.


Subject(s)
Binge Drinking/blood , Ethanol/adverse effects , Liver Diseases, Alcoholic/therapy , Mesenchymal Stem Cell Transplantation , Satellite Cells, Skeletal Muscle/transplantation , Animals , Binge Drinking/complications , Caco-2 Cells , Cells, Cultured , Dinoprostone/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Inflammation , Liver/metabolism , Liver Diseases, Alcoholic/etiology , Mesenchymal Stem Cells , Mice
2.
Mol Ther ; 28(5): 1339-1358, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32209436

ABSTRACT

The need to distribute therapy evenly systemically throughout the large muscle volume within the body makes Duchenne muscular dystrophy (DMD) therapy a challenge. Cell and exon-skipping therapies are promising but have limited effects, and thus enhancing their therapeutic potency is of paramount importance to increase the accessibility of these therapies to DMD patients. In this study, we demonstrate that co-administered glycine improves phosphorodiamidate morpholino oligomer (PMO) potency in mdx mice with marked functional improvement and an up to 50-fold increase of dystrophin in abdominal muscles compared to PMO in saline. Glycine boosts satellite cell proliferation and muscle regeneration by increasing activation of mammalian target of rapamycin complex 1 (mTORC1) and replenishing the one-carbon unit pool. The expanded regenerating myofiber population then results in increased PMO uptake. Glycine also augments the transplantation efficiency of exogenous satellite cells and primary myoblasts in mdx mice. Our data provide evidence that glycine enhances satellite cell proliferation, cell transplantation, and oligonucleotide efficacy in mdx mice, and thus it has therapeutic utility for cell therapy and drug delivery in muscle-wasting diseases.


Subject(s)
Cell Proliferation/drug effects , Cell Transplantation/methods , Glycine Agents/administration & dosage , Glycine/administration & dosage , Morpholinos/administration & dosage , Muscular Dystrophy, Duchenne/drug therapy , Myoblasts/transplantation , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Animals , Disease Models, Animal , Drug Synergism , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/physiology , Regeneration/drug effects , Signal Transduction/drug effects , Treatment Outcome
3.
Hum Mol Genet ; 26(19): 3682-3698, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28666318

ABSTRACT

α-Dystroglycanopathies are a group of muscular dystrophies characterized by α-DG hypoglycosylation and reduced extracellular ligand-binding affinity. Among other genes involved in the α-DG glycosylation process, fukutin related protein (FKRP) gene mutations generate a wide range of pathologies from mild limb girdle muscular dystrophy 2I (LGMD2I), severe congenital muscular dystrophy 1C (MDC1C), to Walker-Warburg Syndrome and Muscle-Eye-Brain disease. FKRP gene encodes for a glycosyltransferase that in vivo transfers a ribitol phosphate group from a CDP -ribitol present in muscles to α-DG, while in vitro it can be secreted as monomer of 60kDa. Consistently, new evidences reported glycosyltransferases in the blood, freely circulating or wrapped within vesicles. Although the physiological function of blood stream glycosyltransferases remains unclear, they are likely released from blood borne or distant cells. Thus, we hypothesized that freely or wrapped FKRP might circulate as an extracellular glycosyltransferase, able to exert a "glycan remodelling" process, even at distal compartments. Interestingly, we firstly demonstrated a successful transduction of MDC1C blood-derived CD133+ cells and FKRP L276IKI mouse derived satellite cells by a lentiviral vector expressing the wild-type of human FKRP gene. Moreover, we showed that LV-FKRP cells were driven to release exosomes carrying FKRP. Similarly, we observed the presence of FKRP positive exosomes in the plasma of FKRP L276IKI mice intramuscularly injected with engineered satellite cells. The distribution of FKRP protein boosted by exosomes determined its restoration within muscle tissues, an overall recovery of α-DG glycosylation and improved muscle strength, suggesting a systemic supply of FKRP protein acting as glycosyltransferase.


Subject(s)
Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/therapy , Proteins/metabolism , Animals , Disease Models, Animal , Dystroglycans/metabolism , Exosomes , Glycosylation , Glycosyltransferases/metabolism , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Myoblasts/metabolism , Pentosyltransferases , Proteins/genetics , Satellite Cells, Skeletal Muscle/transplantation , Transferases
4.
Connect Tissue Res ; 60(2): 128-135, 2019 03.
Article in English | MEDLINE | ID: mdl-29651864

ABSTRACT

AIM OF THE STUDY: Blepharoptosis is a drooping of the upper eyelid, usually due to dysfunction of the levator palpebrae superioris (LPS). Recently, skeletal muscle satellite cells (SSCs) have been reported to promote the repair of damaged skeletal muscle. This study aims to investigate the potential contribution of exogenous SSCs to the regeneration of mechanically damaged LPS. MATERIALS AND METHODS: Thirty-two rats were randomly divided into four groups, including control group, SSCs-treated group, SSCs-treated injury group and non-treated injury group. After rats in injury groups were artificially lacerated on both the left and right LPS, HBBS (Hank's Balanced Salt Solution) containing SSCs was injected into upper eyelid tissue. After 7 days, the LPS muscle tissues were excised. In addition, skeletal muscle cells (SMCs) and SSCs were cocultured for use as an in vitro model, and the protective effects of SSCs on cultured SMCs were also investigated. RESULTS: Histological staining revealed that exogenous SSCs repaired the damaged muscle fibers and attenuated the fibrosis of LPS, possibly due to the increased level of IGF-1. In contrast, the level of IL-1ß, IL-6, TGF-ß1 and Smad2/3 (phospho-T8) were significantly reduced in the SSCs-treated group. The in vitro model using coculture of skeletal muscle cells (SMCs) and SSCs also revealed an increased level of IGF-1 and reduced level of inflammatory factors, resulting in a better cell survival rate. CONCLUSIONS: This study found that exogenous SSCs can promote the repair of LPS mechanical damage and provides new insight into the development of novel therapeutic approaches for blepharoptosis.


Subject(s)
Oculomotor Muscles/pathology , Satellite Cells, Skeletal Muscle/transplantation , Stress, Mechanical , Wound Healing , Animals , Coculture Techniques , Inflammation/pathology , Insulin-Like Growth Factor I/metabolism , Lipopolysaccharides/pharmacology , Rats, Sprague-Dawley , Transforming Growth Factor beta1/metabolism
5.
Exp Cell Res ; 352(1): 84-94, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28153781

ABSTRACT

Multipotent muscle satellite cells (MuSCs) have been identified as potential seed cells for bone tissue engineering. However, MuSCs exhibit a rapid loss of stemness after in vitro culturing, thereby compromising their therapeutic efficiency. Muscle segment homeobox gene 1 (msx1) has been found to induce the dedifferentiation of committed progenitor cells, as well as terminally differentiated myotubes. In this study, a Tet-off retroviral gene delivery system was used to modulate msx1 expression. After ten passages, MuSCs that did not express msx-1 (e.g., the non-msx1 group) were compared with MuSCs with induced msx-1 expression (e.g., the msx1 group). The latter group exhibited a more juvenile morphology, it contained a significantly lower percentage of senescent cells characterized by positive ß-galactosidase staining, and it exhibited increased proliferation and a higher proliferation index. Immunocytochemical stainings further detected a more primitive gene expression profile for the msx1 group, while osteogenic differentiation assays and ectopic bone formation assays demonstrated an improved capacity for the msx1 group to undergo osteogenic differentiation. These results suggest that transient expression of msx1 in MuSCs can retain a primitive state, thereby enhancing their capacity for osteogenic differentiation and restoring the potential for MuSCs to serve as seed cells for bone tissue engineering.


Subject(s)
Cell Differentiation , MSX1 Transcription Factor/metabolism , Osteogenesis/physiology , Satellite Cells, Skeletal Muscle/cytology , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Cells, Cultured , Humans , Immunoenzyme Techniques , MSX1 Transcription Factor/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , beta-Galactosidase
6.
Nature ; 490(7420): 355-60, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-23023126

ABSTRACT

The niche is a conserved regulator of stem cell quiescence and function. During ageing, stem cell function declines. To what extent and by what means age-related changes within the niche contribute to this phenomenon are unknown. Here we demonstrate that the aged muscle stem cell niche, the muscle fibre, expresses Fgf2 under homeostatic conditions, driving a subset of satellite cells to break quiescence and lose their self-renewing capacity. We show in mice that relatively dormant aged satellite cells robustly express sprouty 1 (Spry1), an inhibitor of fibroblast growth factor (FGF) signalling. Increasing FGF signalling in aged satellite cells under homeostatic conditions by removing Spry1 results in the loss of quiescence, satellite cell depletion and diminished regenerative capacity. Conversely, reducing niche-derived FGF activity through inhibition of Fgfr1 signalling or overexpression of Spry1 in satellite cells prevents their depletion. These experiments identify an age-dependent change in the stem cell niche that directly influences stem cell quiescence and function.


Subject(s)
Aging/physiology , Cell Cycle , Muscle Cells/cytology , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Niche/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Count , Cell Differentiation , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Flow Cytometry , Homeostasis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Muscle, Skeletal/cytology , PAX7 Transcription Factor/metabolism , Phosphoproteins/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Signal Transduction , Time Factors
7.
Am J Physiol Heart Circ Physiol ; 309(2): H325-34, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25980021

ABSTRACT

Chronic failure in maintenance and regeneration of skeletal muscles leads to lower muscle mass (sarcopenia), muscle weakness, and poor response to injury. Evidence suggests that aberrant p38 MAPK signaling undermines the repair process after injury in aged mice. Previous studies have shown that hyperhomocysteinemia (HHcy) has been associated with muscle weakness and lower than normal body weights. However, whether or not HHcy condition also compromises skeletal muscle regenerative capabilities is not clear. In the current study, we show that CBS-/+ mice, a model for HHcy condition, exhibited compromised regenerative function and cell proliferation upon injury. However, there was no significant difference in Pax7 expression levels in the satellite cells from CBS-/+ mouse skeletal muscles. Interestingly, the satellite cells from CBS-/+ mice not only exhibited diminished in vitro proliferative capabilities, but also there was heightened oxidative stress. In addition, there was enhanced p38 MAPK activation as well as p16 and p21 expression in the CBS-/+ mouse satellite cells. Moreover, the C2C12 myoblasts also exhibited higher p38 MAPK activation and p16 expression upon treatment with homocysteine in addition to enhanced ROS presence. Tissue engraftment potential and regeneration after injury were restored to some extent upon treatment with the p38-MAPK inhibitor, SB203580, in the CBS-/+ mice. These results together suggest that HHcy-induced diminished satellite cell proliferation involves excessive oxidative stress and p38 MAPK signaling. Our study further proposes that HHcy is a potential risk factor for elderly frailty, and need to be considered as a therapeutic target while designing the alleviation interventions/postinjury rehabilitation measures for adults with HHcy.


Subject(s)
Cell Proliferation , Hyperhomocysteinemia/enzymology , MAP Kinase Signaling System , Muscle, Skeletal/enzymology , Regeneration , Satellite Cells, Skeletal Muscle/enzymology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Cystathionine beta-Synthase/deficiency , Cystathionine beta-Synthase/genetics , Disease Models, Animal , Enzyme Activation , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/pathology , Hyperhomocysteinemia/physiopathology , MAP Kinase Signaling System/drug effects , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Oxidative Stress , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Sarcopenia/enzymology , Sarcopenia/pathology , Sarcopenia/physiopathology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/transplantation , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
8.
Patol Fiziol Eksp Ter ; 59(2): 88-98, 2015.
Article in Russian | MEDLINE | ID: mdl-26571813

ABSTRACT

Musculoskeletal functions disorders may develop as a consequence of injuries and various types of congenital / acquired diseases, among which a special place belongs to muscular dystrophy. The technology with use of cells possessing myogenic potential is considered as one of the most promising approaches to solve the problem of effective restoration of skeletal muscles structure and function. In part I of the article the characteristic features, functions and phenotypic characteristics of satellite cells (SC) are reviewed as key factors of skeletal muscle tissue regeneration. Presented analysis of research results (preclinical and clinical) concerning therapeutic possibilities of technology using SC. In the second part of review will be presented data of the therapeutic use of stem cells of muscle and non-muscle origin for the treatment of skeletal muscles diseases.


Subject(s)
Muscle Development , Muscle, Skeletal/metabolism , Musculoskeletal Diseases/metabolism , Musculoskeletal Diseases/therapy , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Animals , Humans , Muscle, Skeletal/pathology , Musculoskeletal Diseases/genetics , Musculoskeletal Diseases/pathology , Musculoskeletal Diseases/physiopathology , Satellite Cells, Skeletal Muscle/pathology
9.
Stem Cells ; 31(8): 1611-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23606600

ABSTRACT

Transplantation of a myogenic cell population into an immunodeficient recipient is an excellent way of assessing the in vivo muscle-generating capacity of that cell population. To facilitate both allogeneic and xenogeneic transplantations of muscle-forming cells in mice, we have developed a novel immunodeficient muscular dystrophy model, the NSG-mdx(4Cv) mouse. The IL2Rg mutation, which is linked to the Dmd gene on the X chromosome, simultaneously depletes NK cells and suppresses thymic lymphomas, issues that limit the utility of the SCID/mdx model. The NSG-mdx(4Cv) mouse presents a muscular dystrophy of similar severity to the conventional mdx mouse. We show that this animal supports robust engraftment of both pig and dog muscle mononuclear cells. The question of whether satellite cells prospectively isolated by flow cytometry can confer a functional benefit upon transplantation has been controversial. Using allogeneic Pax7-ZsGreen donors and NSG-mdx(4Cv) recipients, we demonstrate definitively that as few as 900 FACS-isolated satellite cells can provide functional regeneration in vivo, in the form of an increased mean maximal force-generation capacity in cell-transplanted muscles, compared to a sham-injected control group. These studies highlight the potency of satellite cells to improve muscle function and the utility of the NSG-mdx(4Cv) model for studies on muscle regeneration and Duchenne muscular dystrophy therapy.


Subject(s)
Dystrophin/deficiency , Muscular Dystrophy, Duchenne/surgery , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Animals , Disease Models, Animal , Dogs , Dystrophin/genetics , Dystrophin/metabolism , Female , Genotype , Heterografts , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Satellite Cells, Skeletal Muscle/cytology , Swine , Transplantation, Homologous
10.
Nat Cell Biol ; 9(3): 255-67, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17293855

ABSTRACT

Cells derived from blood vessels of human skeletal muscle can regenerate skeletal muscle, similarly to embryonic mesoangioblasts. However, adult cells do not express endothelial markers, but instead express markers of pericytes, such as NG2 proteoglycan and alkaline phosphatase (ALP), and can be prospectively isolated from freshly dissociated ALP(+) cells. Unlike canonical myogenic precursors (satellite cells), pericyte-derived cells express myogenic markers only in differentiated myotubes, which they form spontaneously with high efficiency. When transplanted into severe combined immune deficient-X-linked, mouse muscular dystrophy (scid-mdx) mice, pericyte-derived cells colonize host muscle and generate numerous fibres expressing human dystrophin. Similar cells isolated from Duchenne patients, and engineered to express human mini-dystrophin, also give rise to many dystrophin-positive fibres in vivo. These data show that myogenic precursors, distinct from satellite cells, are associated with microvascular walls in the human skeletal muscle, may represent a correlate of embryonic 'mesoangioblasts' present after birth and may be a promising candidate for future cell-therapy protocols in patients.


Subject(s)
Adult Stem Cells/cytology , Muscle, Skeletal/cytology , Pericytes/cytology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/cytology , Adolescent , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , Aged , Animals , Antigens, CD/analysis , Cell Culture Techniques/methods , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred mdx , Mice, Nude , Mice, SCID , Middle Aged , Muscle Proteins/analysis , Muscle Proteins/genetics , Muscle, Skeletal/chemistry , Muscle, Skeletal/physiology , Muscular Dystrophy, Duchenne/physiopathology , Muscular Dystrophy, Duchenne/surgery , Pericytes/chemistry , Pericytes/transplantation , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation/methods , Treatment Outcome
11.
J Clin Invest ; 134(12)2024 May 07.
Article in English | MEDLINE | ID: mdl-38713532

ABSTRACT

Satellite cells, the stem cells of skeletal muscle tissue, hold a remarkable regeneration capacity and therapeutic potential in regenerative medicine. However, low satellite cell yield from autologous or donor-derived muscles hinders the adoption of satellite cell transplantation for the treatment of muscle diseases, including Duchenne muscular dystrophy (DMD). To address this limitation, here we investigated whether satellite cells can be derived in allogeneic or xenogeneic animal hosts. First, injection of CRISPR/Cas9-corrected Dmdmdx mouse induced pluripotent stem cells (iPSCs) into mouse blastocysts carrying an ablation system of host satellite cells gave rise to intraspecies chimeras exclusively carrying iPSC-derived satellite cells. Furthermore, injection of genetically corrected DMD iPSCs into rat blastocysts resulted in the formation of interspecies rat-mouse chimeras harboring mouse satellite cells. Notably, iPSC-derived satellite cells or derivative myoblasts produced in intraspecies or interspecies chimeras restored dystrophin expression in DMD mice following intramuscular transplantation and contributed to the satellite cell pool. Collectively, this study demonstrates the feasibility of producing therapeutically competent stem cells across divergent animal species, raising the possibility of generating human muscle stem cells in large animals for regenerative medicine purposes.


Subject(s)
Induced Pluripotent Stem Cells , Muscular Dystrophy, Duchenne , Satellite Cells, Skeletal Muscle , Animals , Mice , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/genetics , Induced Pluripotent Stem Cells/transplantation , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Rats , Satellite Cells, Skeletal Muscle/transplantation , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Transplantation , Humans , Dystrophin/genetics , Dystrophin/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/cytology , Mice, Inbred mdx , Heterografts , Transplantation, Heterologous , Injections, Intramuscular , Transplantation, Homologous
12.
Stem Cells ; 30(9): 1971-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22730231

ABSTRACT

Stem cell transplantation is already in clinical practice for certain genetic diseases and is a promising therapy for dystrophic muscle. We used the mdx mouse model of Duchenne muscular dystrophy to investigate the effect of the host satellite cell niche on the contribution of donor muscle stem cells (satellite cells) to muscle regeneration. We found that incapacitation of the host satellite cells and preservation of the muscle niche promote donor satellite cell contribution to muscle regeneration and functional reconstitution of the satellite cell compartment. But, if the host niche is not promptly refilled, or is filled by competent host satellite cells, it becomes nonfunctional and donor engraftment is negligible. Application of this regimen to aged host muscles also promotes efficient regeneration from aged donor satellite cells. In contrast, if the niche is destroyed, yet host satellite cells remain proliferation-competent, donor-derived engraftment is trivial. Thus preservation of the satellite cell niche, concomitant with functional impairment of the majority of satellite cells within dystrophic human muscles, may improve the efficiency of stem cell therapy.


Subject(s)
Graft Survival/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Satellite Cells, Skeletal Muscle/transplantation , Animals , Cell Communication/physiology , Cell Differentiation/physiology , Cell Survival/physiology , Disease Models, Animal , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Satellite Cells, Skeletal Muscle/cytology , Stem Cell Transplantation/methods
13.
Stem Cells ; 30(10): 2330-41, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22887880

ABSTRACT

The dystrophin-associated glycoprotein complex (DGC) is found at the muscle fiber sarcolemma and forms an essential structural link between the basal lamina and internal cytoskeleton. In a set of muscular dystrophies known as the dystroglycanopathies, hypoglycosylation of the DGC component α-dystroglycan results in reduced binding to basal lamina components, a loss in structural stability, and repeated cycles of muscle fiber degeneration and regeneration. The satellite cells are the key stem cells responsible for muscle repair and reside between the basal lamina and sarcolemma. In this study, we aimed to determine whether pathological changes associated with the dystroglycanopathies affect satellite cell function. In the Large(myd) mouse dystroglycanopathy model, satellite cells are present in significantly greater numbers but display reduced proliferation on their native muscle fibers in vitro, compared with wild type. However, when removed from their fiber, proliferation in culture is restored to that of wild type. Immunohistochemical analysis of Large(myd) muscle reveals alterations to the basal lamina and interstitium, including marked disorganization of laminin, upregulation of fibronectin and collagens. Proliferation and differentiation of wild-type satellite cells is impaired when cultured on substrates such as collagen and fibronectin, compared with laminins. When engrafted into irradiated tibialis anterior muscles of mdx-nude mice, wild-type satellite cells expanded on laminin contribute significantly more to muscle regeneration than those expanded on fibronectin. These results suggest that defects in α-dystroglycan glycosylation are associated with an alteration in the satellite cell niche, and that regenerative potential in the dystroglycanopathies may be perturbed.


Subject(s)
Basement Membrane/metabolism , Dystroglycans/metabolism , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophy, Animal/metabolism , Sarcolemma/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Animals , Basement Membrane/pathology , Cell Differentiation , Cell Proliferation , Collagen/chemistry , Collagen/metabolism , Disease Models, Animal , Fibronectins/chemistry , Fibronectins/metabolism , Glycosylation , Humans , Laminin/chemistry , Laminin/metabolism , Mice , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Protein Binding , Sarcolemma/pathology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/transplantation
14.
J Cell Mol Med ; 16(7): 1353-64, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22129481

ABSTRACT

Muscular dystrophies (MDs) are a heterogeneous group of inherited disorders characterized by progressive muscle wasting and weakness likely associated with exhaustion of muscle regeneration potential. At present, no cures or efficacious treatments are available for these diseases, but cell transplantation could be a potential therapeutic strategy. Transplantation of myoblasts using satellite cells or other myogenic cell populations has been attempted to promote muscle regeneration, based on the hypothesis that the donor cells repopulate the muscle and contribute to its regeneration. Embryonic stem cells (ESCs) and more recently induced pluripotent stem cells (iPSCs) could generate an unlimited source of differentiated cell types, including myogenic cells. Here we review the literature regarding the generation of myogenic cells considering the main techniques employed to date to elicit efficient differentiation of human and murine ESCs or iPSCs into skeletal muscle. We also critically analyse the possibility of using these cellular populations as an alternative source of myogenic cells for cell therapy of MDs.


Subject(s)
Embryonic Stem Cells/transplantation , Induced Pluripotent Stem Cells/transplantation , Muscle Fibers, Skeletal/transplantation , Muscular Dystrophies/therapy , Animals , Cell Culture Techniques , Cell Differentiation , Disease Models, Animal , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation
15.
FASEB J ; 25(7): 2296-304, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21450908

ABSTRACT

The success of skeletal muscle reconstruction depends on finding the most effective, clinically suitable strategy to engineer myogenic cells and biocompatible scaffolds. Satellite cells (SCs), freshly isolated or transplanted within their niche, are presently considered the best source for muscle regeneration. Here, we designed and developed the delivery of either SCs or muscle progenitor cells (MPCs) via an in situ photo-cross-linkable hyaluronan-based hydrogel, hyaluronic acid-photoinitiator (HA-PI) complex. Partially ablated tibialis anterior (TA) of C57BL/6J mice engrafted with freshly isolated satellite cells embedded in hydrogel showed a major improvement in muscle structure and number of new myofibers, compared to muscles receiving hydrogel + MPCs or hydrogel alone. Notably, SCs embedded in HA-PI also promoted functional recovery, as assessed by contractile force measurements. Tissue reconstruction was associated with the formation of both neural and vascular networks and the reconstitution of a functional SC niche. This innovative approach could overcome previous limitations in skeletal muscle tissue engineering.


Subject(s)
Hydrogel, Polyethylene Glycol Dimethacrylate/metabolism , Muscle, Skeletal/cytology , Satellite Cells, Skeletal Muscle/cytology , Tissue Engineering/methods , Animals , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hyaluronic Acid/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/transplantation , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Polymerization/radiation effects , Reproducibility of Results , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation
16.
Cell Biol Int ; 36(12): 1195-205, 2012.
Article in English | MEDLINE | ID: mdl-22988823

ABSTRACT

PRP (platelet-rich plasma)-derived growth factors are a new application of tissue engineering and a developing area for researchers and clinicians. We have assessed the effects of PRP-derived growth factors on the proliferation and osteogenic differentiation of rMSCs (rat muscle satellite cells), and constructed a novel tissue engineering bone composed of PRP-derived growth factors and rMSCs. PRP were created by a freeze-thaw process. rMSCs were isolated from rat masticatory muscle using serial platings technique. Wst-1 assay, SEM (scanning electron microscopy), ALP (alkaline phosphatase) activity, total protein concentration, AR (Alizarin red S) staining, calcium analyses and RT-PCR (reverse transcription-PCR) of osteogenic-related genes were used to assess the effect of PRP-derived growth factors on proliferation and osteogenic differentiation of cultured rMSCs on scaffolds. The different composite scaffolds were implanted to the subcutaneous spaces of nude mice. H&E (haematoxylin and eosin) and Masson's trichrome staining were used to examine the ectopic bone formation. In vitro, we found that PRP-derived growth factors showed excellent cell compatibility and significantly enhanced cell proliferation over serum and control groups at 48 and 72 h. SEM, ALP activity, AR staining, calcium analyses and RT-PCR showed that PRP-derived growth factors significantly increased cells osteogenic differentiation when compared with other groups. In vivo examination showed that more fibrous tissue capsule and bone with lamellar structures appeared in PRP-derived growth factors groups. These results suggest that the PRP-derived growth factors significantly promote rMSCs proliferation, osteogenic differentiation compared with serum and scaffolds alone, and may be suitable for stem cell growth factors delivery and bone tissue engineering.


Subject(s)
Osteogenesis , Platelet-Derived Growth Factor/metabolism , Satellite Cells, Skeletal Muscle/cytology , Alkaline Phosphatase/analysis , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Mice , Mice, Nude , Platelet-Derived Growth Factor/isolation & purification , Platelet-Rich Plasma/chemistry , Rats , Rats, Sprague-Dawley , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Tissue Scaffolds/chemistry
17.
J Cell Biol ; 172(3): 433-40, 2006 Jan 30.
Article in English | MEDLINE | ID: mdl-16449193

ABSTRACT

In contrast to mammals, salamanders can regenerate complex structures after injury, including entire limbs. A central question is whether the generation of progenitor cells during limb regeneration and mammalian tissue repair occur via separate or overlapping mechanisms. Limb regeneration depends on the formation of a blastema, from which the new appendage develops. Dedifferentiation of stump tissues, such as skeletal muscle, precedes blastema formation, but it was not known whether dedifferentiation involves stem cell activation. We describe a multipotent Pax7+ satellite cell population located within the skeletal muscle of the salamander limb. We demonstrate that skeletal muscle dedifferentiation involves satellite cell activation and that these cells can contribute to new limb tissues. Activation of salamander satellite cells occurs in an analogous manner to how the mammalian myofiber mobilizes stem cells during skeletal muscle tissue repair. Thus, limb regeneration and mammalian tissue repair share common cellular and molecular programs. Our findings also identify satellite cells as potential targets in promoting mammalian blastema formation.


Subject(s)
Extremities/physiology , Multipotent Stem Cells/physiology , Regeneration/physiology , Satellite Cells, Skeletal Muscle/physiology , Adipocytes/cytology , Animals , Basement Membrane/cytology , Cadherins/metabolism , Cartilage/cytology , Cell Count , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation , Cell Transplantation , Cells, Cultured , Epidermal Cells , Histones/analysis , Multipotent Stem Cells/cytology , Multipotent Stem Cells/transplantation , Muscle Fibers, Skeletal/cytology , Muscle, Skeletal/chemistry , Muscle, Skeletal/cytology , MyoD Protein/analysis , Myosin Heavy Chains/metabolism , Notophthalmus viridescens , Osteoblasts/cytology , PAX7 Transcription Factor/analysis , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/transplantation
18.
Zhongguo Zhong Xi Yi Jie He Za Zhi ; 31(8): 1093-6, 2011 Aug.
Article in Zh | MEDLINE | ID: mdl-21910342

ABSTRACT

OBJECTIVE: To study the effects of Danggui Buxue Decoction on the hematopoietic reconstruction of mice transplanted by muscle satellite cells ( MSCs). METHODS: MSCs were procured from newly born male mice of the homologous series. The female Kunming receptor mice irradiated with 8Gy137Cs-gamma ray were randomly divided into six groups, i. e., the blank control group, the transplanted MSC group, four groups intervened by different doses of Danggui Buxue Decoction after transplanted MSC (gastrogavage by one, three, five, and ten times of clinically equivalent dose for seven days, as the DGBX 1 group, the DGBX 2 group, the DGBX 3 group, and the DGBX 4 group). After transplantation changes of splenic colony forming unit spleen (CFU-S), white blood cells (WBC), hemoglobin (Hb), and platelet (PLT) in the peripheral blood were observed in pos-transplanted 1-, 2-, and 3-week receptor mice. The 3-week survival rate was calculated. The source of hematopoietic reconstruction was identified using PCR. RESULTS: The desmin protein was positive in the cultured MSCs. WBC of each transplanted MSC group obviously increased at the 2nd week (P<0.05). Hb obviously increased in the DGBX 2 group, the DGBX 3 group, and the DGBX 4 group (P<0.05). WBC and Hb obviously increased in the DGBX 3 group and the DGBX 4 group when compared with the transplanted MSC group at the 3rd week (P<0.05). The recovery of PLT was significant in the DGBX 4 group (P<0.05). Compared with the blank control group at the 2nd week, CFU-S obviously increased in the DGBX 3 group and the DGBX 4 group (P<0.05). PCR results of Y chromosome in the survived transplant mice indicated that the hematopoietic cells of reconstruction female receptor mice were originated from male donors. CONCLUSION: The hematopoiesis of mice transplanted by muscle satellite cells could be constructed after intervention of Danggui Buxue Decoction.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hematopoiesis/drug effects , Satellite Cells, Skeletal Muscle/drug effects , Animals , Female , Humans , Male , Mice , Mice, Inbred Strains , Satellite Cells, Skeletal Muscle/transplantation
19.
Dev Cell ; 56(7): 1014-1029.e7, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33735618

ABSTRACT

Negative elongation factor (NELF) is a critical transcriptional regulator that stabilizes paused RNA polymerase to permit rapid gene expression changes in response to environmental cues. Although NELF is essential for embryonic development, its role in adult stem cells remains unclear. In this study, through a muscle-stem-cell-specific deletion, we showed that NELF is required for efficient muscle regeneration and stem cell pool replenishment. In mechanistic studies using PRO-seq, single-cell trajectory analyses and myofiber cultures revealed that NELF works at a specific stage of regeneration whereby it modulates p53 signaling to permit massive expansion of muscle progenitors. Strikingly, transplantation experiments indicated that these progenitors are also necessary for stem cell pool repopulation, implying that they are able to return to quiescence. Thus, we identified a critical role for NELF in the expansion of muscle progenitors in response to injury and revealed that progenitors returning to quiescence are major contributors to the stem cell pool repopulation.


Subject(s)
Muscle, Skeletal/physiology , Satellite Cells, Skeletal Muscle/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cells, Cultured , Eye Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development , Nerve Growth Factors/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Serpins/metabolism , Signal Transduction , Transcription Factors/genetics , Transcriptome , Tumor Suppressor Protein p53/metabolism
20.
Nat Biomed Eng ; 5(8): 864-879, 2021 08.
Article in English | MEDLINE | ID: mdl-33737730

ABSTRACT

Muscle loss and impairment resulting from traumatic injury can be alleviated by therapies using muscle stem cells. However, collecting sufficient numbers of autologous myogenic stem cells and expanding them efficiently has been challenging. Here we show that myogenic stem cells (predominantly Pax7+ cells)-which were selectively expanded from readily obtainable dermal fibroblasts or skeletal muscle stem cells using a specific cocktail of small molecules and transplanted into muscle injuries in adult, aged or dystrophic mice-led to functional muscle regeneration in the three animal models. We also show that sustained release of the small-molecule cocktail in situ through polymer nanoparticles led to muscle repair by inducing robust activation and expansion of resident satellite cells. Chemically induced stem cell expansion in vitro and in situ may prove to be advantageous for stem cell therapies that aim to regenerate skeletal muscle and other tissues.


Subject(s)
Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/cytology , Animals , Cellular Reprogramming/drug effects , Colforsin/pharmacology , Culture Media/chemistry , Culture Media/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Muscular Diseases/therapy , Nanoparticles/chemistry , PAX7 Transcription Factor/metabolism , Polymers/chemistry , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/transplantation , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/metabolism , Valproic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL