Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
FASEB Bioadv ; 5(11): 453-469, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37936920

ABSTRACT

Store-operated Ca2+ entry (SOCE) is indispensable for intracellular Ca2+ homeostasis in skeletal muscle, and constitutive activation of SOCE causes tubular aggregate myopathy (TAM). To understand the pathogenesis of TAM, we induced pluripotent stem cells (iPSCs) from a TAM patient with a rare mutation (c.1450_1451insGA; p. Ile484ArgfsX21) in the STIM1 gene. This frameshift mutation produces a truncated STIM1 with a disrupted C-terminal inhibitory domain (CTID) and was reported to diminish SOCE. Myotubes induced from the patient's-iPSCs (TAM myotubes) showed severely impaired SOCE, but antioxidants greatly restored SOCE partly via upregulation of an endoplasmic reticulum (ER) chaperone, BiP (GRP78), in the TAM myotubes. Our observation suggests that antioxidants are promising tools for treatment of TAM caused by reduced SOCE.

2.
Stem Cell Res Ther ; 12(1): 532, 2021 10 09.
Article in English | MEDLINE | ID: mdl-34627382

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is a severe X-linked recessive disease caused by mutations in the dystrophin gene. Transplantation of myogenic stem cells holds great promise for treating muscular dystrophies. However, poor engraftment of myogenic stem cells limits the therapeutic effects of cell therapy. Mesenchymal stem cells (MSCs) have been reported to secrete soluble factors necessary for skeletal muscle growth and regeneration. METHODS: We induced MSC-like cells (iMSCs) from induced pluripotent stem cells (iPSCs) and examined the effects of iMSCs on the proliferation and differentiation of human myogenic cells and on the engraftment of human myogenic cells in the tibialis anterior (TA) muscle of NSG-mdx4Cv mice, an immunodeficient dystrophin-deficient DMD model. We also examined the cytokines secreted by iMSCs and tested their effects on the engraftment of human myogenic cells. RESULTS: iMSCs promoted the proliferation and differentiation of human myogenic cells to the same extent as bone marrow-derived (BM)-MSCs in coculture experiments. In cell transplantation experiments, iMSCs significantly improved the engraftment of human myogenic cells injected into the TA muscle of NSG-mdx4Cv mice. Cytokine array analysis revealed that iMSCs produced insulin-like growth factor-binding protein 2 (IGFBP2), urokinase-type plasminogen activator receptor (uPAR), and brain-derived neurotrophic factor (BDNF) at higher levels than did BM-MSCs. We further found that uPAR stimulates the migration of human myogenic cells in vitro and promotes their engraftment into the TA muscles of immunodeficient NOD/Scid mice. CONCLUSIONS: Our results indicate that iMSCs are a new tool to improve the engraftment of myogenic progenitors in dystrophic muscle.


Subject(s)
Induced Pluripotent Stem Cells , Mesenchymal Stem Cells , Muscular Dystrophy, Duchenne , Animals , Cell Differentiation , Dystrophin/genetics , Humans , Mice , Mice, Inbred NOD , Mice, Inbred mdx , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Receptors, Urokinase Plasminogen Activator/genetics
3.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807264

ABSTRACT

Skeletal muscle regeneration is a well-organized process that requires remodeling of the extracellular matrix (ECM). In this study, we revealed the protective role of periostin, a matricellular protein that binds to several ECM proteins during muscle regeneration. In intact muscle, periostin was localized at the neuromuscular junction, muscle spindle, and myotendinous junction, which are connection sites between muscle fibers and nerves or tendons. During muscle regeneration, periostin exhibited robustly increased expression and localization at the interstitial space. Periostin-null mice showed decreased muscle weight due to the loss of muscle fibers during repeated muscle regeneration. Cultured muscle progenitor cells from periostin-null mice showed no deficiencies in their proliferation, differentiation, and the expression of Pax7, MyoD, and myogenin, suggesting that the loss of muscle fibers in periostin-null mice was not due to the impaired function of muscle stem/progenitor cells. Periostin-null mice displayed a decreased number of CD31-positive blood vessels during muscle regeneration, suggesting that the decreased nutritional supply from blood vessels was the cause of muscle fiber loss in periostin-null mice. These results highlight the novel role of periostin in maintaining muscle mass during muscle regeneration.


Subject(s)
Cell Adhesion Molecules/metabolism , Muscle Fibers, Skeletal/metabolism , Regeneration/physiology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/physiology , Cell Differentiation , Cell-Matrix Junctions/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Tendons/metabolism , Wound Healing/physiology
4.
Hum Mol Genet ; 30(11): 1006-1019, 2021 05 31.
Article in English | MEDLINE | ID: mdl-33822956

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder characterized by progressive muscular weakness because of the loss of dystrophin. Extracellular Ca2+ flows into the cytoplasm through membrane tears in dystrophin-deficient myofibers, which leads to muscle contracture and necrosis. Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) takes up cytosolic Ca2+ into the sarcoplasmic reticulum, but its activity is decreased in dystrophic muscle. Here, we show that an allosteric SERCA activator, CDN1163, ameliorates dystrophic phenotypes in dystrophin-deficient mdx mice. The administration of CDN1163 prevented exercise-induced muscular damage and restored mitochondrial function. In addition, treatment with CDN1163 for 7 weeks enhanced muscular strength and reduced muscular degeneration and fibrosis in mdx mice. Our findings provide preclinical proof-of-concept evidence that pharmacological activation of SERCA could be a promising therapeutic strategy for DMD. Moreover, CDN1163 improved muscular strength surprisingly in wild-type mice, which may pave the new way for the treatment of muscular dysfunction.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Duchenne/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Animals , Calcium/metabolism , Disease Models, Animal , Dystrophin/deficiency , Humans , Mice , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle Weakness/genetics , Muscle Weakness/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Dystrophy, Duchenne/pathology , Phenotype , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology
5.
Cells ; 9(11)2020 11 23.
Article in English | MEDLINE | ID: mdl-33238404

ABSTRACT

Perlecan is an extracellular matrix molecule anchored to the sarcolemma by a dystrophin-glycoprotein complex. Perlecan-deficient mice are tolerant to muscle atrophy, suggesting that perlecan negatively regulates mechanical stress-dependent skeletal muscle mass. Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma to the cytosol triggers protein degradation, thereby initiating skeletal muscle atrophy. We hypothesized that perlecan regulates nNOS delocalization and activates protein degradation during this process. To determine the role of perlecan in nNOS-mediated mechanotransduction, we used sciatic nerve transection as a denervation model of gastrocnemius muscles. Gastrocnemius muscle atrophy was significantly lower in perinatal lethality-rescued perlecan-knockout (Hspg2-/--Tg) mice than controls (WT-Tg) on days 4 and 14 following surgery. Immunofluorescence microscopy showed that cell membrane nNOS expression was reduced by denervation in WT-Tg mice, with marginal effects in Hspg2-/--Tg mice. Moreover, levels of atrophy-related proteins-i.e., FoxO1a, FoxO3a, atrogin-1, and Lys48-polyubiquitinated proteins-increased in the denervated muscles of WT-Tg mice but not in Hspg2-/--Tg mice. These findings suggest that during denervation, perlecan promotes nNOS delocalization from the membrane and stimulates protein degradation and muscle atrophy by activating FoxO signaling and the ubiquitin-proteasome system.


Subject(s)
Heparan Sulfate Proteoglycans/therapeutic use , Muscular Atrophy/chemically induced , Nitric Oxide Synthase Type I/drug effects , Animals , Heparan Sulfate Proteoglycans/pharmacology , Humans , Mice , Mice, Knockout
6.
BMC Musculoskelet Disord ; 21(1): 479, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32693782

ABSTRACT

BACKGROUND: Previous research indicated that nitric oxide synthase (NOS) is the key molecule for S-nitrosylation of ryanodine receptor 1 (RyR1) in DMD model mice (mdx mice) and that both neuronal NOS (nNOS) and inducible NOS (iNOS) might contribute to the reaction because nNOS is mislocalized in the cytoplasm and iNOS expression is higher in mdx mice. We investigated the effect of iNOS on RyR1 S-nitrosylation in mdx mice and whether transgenic expression of truncated dystrophin reduced iNOS expression in mdx mice or not. METHODS: Three- to 4-month-old C57BL/6 J, mdx, and transgenic mdx mice expressing exon 45-55-deleted human dystrophin (Tg/mdx mice) were used. We also generated two double mutant mice, mdx iNOS KO and Tg/mdx iNOS KO to reveal the iNOS contribution to RyR1 S-nitrosylation. nNOS and iNOS expression levels in skeletal muscle of these mice were assessed by immunohistochemistry (IHC), qRT-PCR, and Western blotting. Total NOS activity was measured by a citrulline assay. A biotin-switch method was used for detection of RyR1 S-nitrosylation. Statistical differences were assessed by one-way ANOVA with Tukey-Kramer post-hoc analysis. RESULTS: mdx and mdx iNOS KO mice showed the same level of RyR1 S-nitrosylation. Total NOS activity was not changed in mdx iNOS KO mice compared with mdx mice. iNOS expression was undetectable in Tg/mdx mice expressing exon 45-55-deleted human dystrophin, but the level of RyR1 S-nitrosylation was the same in mdx and Tg/mdx mice. CONCLUSION: Similar levels of RyR1 S-nitrosylation and total NOS activity in mdx and mdx iNOS KO demonstrated that the proportion of iNOS in total NOS activity was low, even in mdx mice. Exon 45-55-deleted dystrophin reduced the expression level of iNOS, but it did not correct the RyR1 S-nitrosylation. These results indicate that iNOS was not involved in RyR1 S-nitrosylation in mdx and Tg/mdx mice muscles.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Nitric Oxide Synthase Type II , Ryanodine Receptor Calcium Release Channel , Animals , Dystrophin/genetics , Dystrophin/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Ryanodine Receptor Calcium Release Channel/genetics
7.
PLoS Genet ; 16(5): e1008826, 2020 05.
Article in English | MEDLINE | ID: mdl-32453729

ABSTRACT

Hearing loss (HL) is one of the most common sensory impairments and etiologically and genetically heterogeneous disorders in humans. Muscular dystrophies (MDs) are neuromuscular disorders characterized by progressive degeneration of skeletal muscle accompanied by non-muscular symptoms. Aberrant glycosylation of α-dystroglycan causes at least eighteen subtypes of MD, now categorized as MD-dystroglycanopathy (MD-DG), with a wide spectrum of non-muscular symptoms. Despite a growing number of MD-DG subtypes and increasing evidence regarding their molecular pathogeneses, no comprehensive study has investigated sensorineural HL (SNHL) in MD-DG. Here, we found that two mouse models of MD-DG, Largemyd/myd and POMGnT1-KO mice, exhibited congenital, non-progressive, and mild-to-moderate SNHL in auditory brainstem response (ABR) accompanied by extended latency of wave I. Profoundly abnormal myelination was found at the peripheral segment of the cochlear nerve, which is rich in the glycosylated α-dystroglycan-laminin complex and demarcated by "the glial dome." In addition, patients with Fukuyama congenital MD, a type of MD-DG, also had latent SNHL with extended latency of wave I in ABR. Collectively, these findings indicate that hearing impairment associated with impaired Schwann cell-mediated myelination at the peripheral segment of the cochlear nerve is a notable symptom of MD-DG.


Subject(s)
Cochlear Nerve/metabolism , Dystroglycans/genetics , Hearing Loss, Sensorineural/metabolism , Myelin Basic Protein/metabolism , N-Acetylglucosaminyltransferases/genetics , Walker-Warburg Syndrome/physiopathology , Adolescent , Animals , Child , Child, Preschool , Disease Models, Animal , Female , Gene Knockout Techniques , Glycosylation , Hearing Loss, Sensorineural/etiology , Hearing Loss, Sensorineural/genetics , Humans , Infant , Male , Mice , Walker-Warburg Syndrome/complications , Walker-Warburg Syndrome/genetics , Young Adult
8.
Commun Biol ; 3(1): 182, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313117

ABSTRACT

Understanding the signaling pathways that regulate proliferation and differentiation of muscle progenitors is essential for successful cell transplantation for treatment of Duchenne muscular dystrophy. Here, we report that a γ-secretase inhibitor, DAPT (N-[N-(3,5-difluorophenacetyl-L-alanyl)]-S-phenylglycine tertial butyl ester), which inhibits the release of NICD (Notch intercellular domain), promotes the fusion of human muscle progenitors in vitro and improves their engraftment in the tibialis anterior muscle of immune-deficient mice. Gene expression analysis revealed that DAPT severely down-regulates PTGER2, which encodes prostaglandin (PG) E2 receptor 2 (EP2), in human muscle progenitors in the differentiation condition. Functional analysis suggested that Notch signaling inhibits differentiation and promotes self-renewal of human muscle progenitors via PGE2/EP2 signaling in a cAMP/PKA-independent manner.


Subject(s)
Cell Differentiation , Cell Proliferation , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Receptor, Notch3/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Differentiation/drug effects , Cell Fusion , Cell Line , Cell Proliferation/drug effects , Cell Self Renewal , Cell Survival , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Dipeptides/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Male , Mice, Inbred NOD , Mice, Inbred mdx , Mice, SCID , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/injuries , Muscle, Skeletal/pathology , Muscle, Skeletal/surgery , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/transplantation , Receptor, Notch3/genetics , Receptors, Prostaglandin E, EP2 Subtype/genetics , Signal Transduction , Stem Cell Transplantation
9.
Cell Death Differ ; 26(6): 1024-1036, 2019 06.
Article in English | MEDLINE | ID: mdl-30154444

ABSTRACT

Skeletal muscle is divided into slow- and fast-type muscles, which possess distinct contractile and metabolic properties. Myogenic progenitors associated with each muscle fiber type are known to intrinsically commit to specific muscle fiber lineage during embryonic development. However, it is still unclear whether the functionality of postnatal adult myogenic cells is attributable to the muscle fiber in which they reside, and whether the characteristics of myogenic cells derived from slow- and fast-type fibers can be distinguished at the genetic level. In this study, we isolated adult satellite cells from slow- and fast-type muscle individually and observed that satellite cells from each type of muscle generated myotubes expressing myosin heavy chain isoforms similar to their original muscle, and showed different metabolic features. Notably, we discovered that slow muscle-derived cells had low potential to differentiate but high potential to self-renew compared with fast muscle-derived cells. Additionally, cell transplantation experiments of slow muscle-derived cells into fast-type muscle revealed that slow muscle-derived cells could better contribute to myofiber formation and satellite cell constitution than fast muscle-derived cells, suggesting that the recipient muscle fiber type may not affect the predetermined abilities of myogenic cells. Gene expression analyses identified T-box transcriptional factor Tbx1 as a highly expressed gene in fast muscle-derived myoblasts. Gain- and loss-of-function experiments revealed that Tbx1 modulated muscle fiber types and oxidative metabolism in myotubes, and that Tbx1 stimulated myoblast differentiation, but did not regulate myogenic cell self-renewal. Our data suggest that metabolic and myogenic properties of myogenic progenitor cells vary depending on the type of muscle from which they originate, and that Tbx1 expression partially explains the functional differences of myogenic cells derived from fast-type and slow-type muscles.


Subject(s)
Muscle, Skeletal/metabolism , Myoblasts/metabolism , Stem Cells/metabolism , T-Box Domain Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Muscle Development , T-Box Domain Proteins/genetics
10.
Sci Rep ; 8(1): 6555, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29700358

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) are a potential source for cell therapy of Duchenne muscular dystrophy. To reliably obtain skeletal muscle progenitors from hiPSCs, we treated hiPS cells with a Wnt activator, CHIR-99021 and a BMP receptor inhibitor, LDN-193189, and then induced skeletal muscle cells using a previously reported sphere-based culture. This protocol greatly improved sphere formation efficiency and stably induced the differentiation of myogenic cells from hiPS cells generated from both healthy donors and a patient with congenital myasthenic syndrome. hiPSC-derived myogenic progenitors were enriched in the CD57(-) CD108(-) CD271(+) ERBB3(+) cell fraction, and their differentiation was greatly promoted by TGF-ß inhibitors. TGF-ß inhibitors down-regulated the NFIX transcription factor, and NFIX short hairpin RNA (shRNA) improved the differentiation of iPS cell-derived myogenic progenitors. These results suggest that NFIX inhibited differentiation of myogenic progenitors. hiPSC-derived myogenic cells differentiated into myofibers in muscles of NSG-mdx 4Cv mice after direct transplantation. Our results indicate that our new muscle induction protocol is useful for cell therapy of muscular dystrophies.


Subject(s)
Cell Differentiation , Myoblasts/cytology , Myoblasts/metabolism , Pluripotent Stem Cells/cytology , Biomarkers , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression , Humans , Immunophenotyping , Induced Pluripotent Stem Cells/cytology , Muscle, Skeletal/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Regeneration/genetics , Stem Cell Transplantation , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
12.
World J Stem Cells ; 9(6): 89-97, 2017 Jun 26.
Article in English | MEDLINE | ID: mdl-28717411

ABSTRACT

Human induced pluripotent stem cells (hiPS cells or hiPSCs) can be derived from cells of patients with severe muscle disease. If skeletal muscle induced from patient-iPSCs shows disease-specific phenotypes, it can be useful for studying the disease pathogenesis and for drug development. On the other hand, human iPSCs from healthy donors or hereditary muscle disease-iPSCs whose genomes are edited to express normal protein are expected to be a cell source for cell therapy. Several protocols for the derivation of skeletal muscle from human iPSCs have been reported to allow the development of efficient treatments for devastating muscle diseases. In 2017, the focus of research is shifting to another stage: (1) the establishment of mature myofibers that are suitable for study of the pathogenesis of muscle disease; (2) setting up a high-throughput drug screening system; and (3) the preparation of highly regenerative, non-oncogenic cells in large quantities for cell transplantation, etc.

13.
Stem Cells Int ; 2017: 7906843, 2017.
Article in English | MEDLINE | ID: mdl-28491099

ABSTRACT

Three to eight percent of female carriers of Duchenne muscular dystrophy (DMD) develop dystrophic symptoms ranging from mild muscle weakness to a rapidly progressive DMD-like muscular dystrophy due to skewed inactivation of X chromosomes during early development. Here, we generated human induced pluripotent stem cells (hiPSCs) from a manifesting female carrier using retroviral or Sendai viral (SeV) vectors and determined their X-inactivation status. Although manifesting carrier-derived iPS cells showed normal expression of human embryonic stem cell markers and formed well-differentiated teratomas in vivo, many hiPS clones showed bi-allelic expression of the androgen receptor (AR) gene and loss of X-inactivation-specific transcript and trimethyl-histone H3 (Lys27) signals on X chromosomes, suggesting that both X chromosomes of the hiPS cells are in an active state. Importantly, normal dystrophin was expressed in multinucleated myotubes differentiated from a manifesting carrier of DMD-hiPS cells with XaXa pattern. AR transcripts were also equally transcribed from both alleles in induced myotubes. Our results indicated that the inactivated X chromosome in the patient's fibroblasts was activated during reprogramming, and XCI occurred randomly during differentiation.

14.
Stem Cell Reports ; 7(2): 263-78, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27509136

ABSTRACT

Skeletal muscle contains two distinct stem/progenitor populations. One is the satellite cell, which acts as a muscle stem cell, and the other is the mesenchymal progenitor, which contributes to muscle pathogeneses such as fat infiltration and fibrosis. Detailed and accurate characterization of these progenitors in humans remains elusive. Here, we performed comprehensive cell-surface protein profiling of the two progenitor populations residing in human skeletal muscle and identified three previously unrecognized markers: CD82 and CD318 for satellite cells and CD201 for mesenchymal progenitors. These markers distinguish myogenic and mesenchymal progenitors, and enable efficient isolation of the two types of progenitors. Functional study revealed that CD82 ensures expansion and preservation of myogenic progenitors by suppressing excessive differentiation, and CD201 signaling favors adipogenesis of mesenchymal progenitors. Thus, cell-surface proteins identified here are not only useful markers but also functionally important molecules, and provide valuable insight into human muscle biology and diseases.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/cytology , Proteomics/methods , Stem Cells/metabolism , Adipogenesis , Antibodies/metabolism , Antigens, CD/metabolism , Biomarkers , Cell Separation , Humans
15.
Cell Rep ; 13(2): 302-14, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26440893

ABSTRACT

Calcitonin receptor (Calcr) is expressed in adult muscle stem cells (muscle satellite cells [MuSCs]). To elucidate the role of Calcr, we conditionally depleted Calcr from adult MuSCs and found that impaired regeneration after muscle injury correlated with the decreased number of MuSCs in Calcr-conditional knockout (cKO) mice. Calcr signaling maintained MuSC dormancy via the cAMP-PKA pathway but had no impact on myogenic differentiation of MuSCs in an undifferentiated state. The abnormal quiescent state in Calcr-cKO mice resulted in a reduction of the MuSC pool by apoptosis. Furthermore, MuSCs were found outside their niche in Calcr-cKO mice, demonstrating cell relocation. This emergence from the sublaminar niche was prevented by the Calcr-cAMP-PKA and Calcr-cAMP-Epac pathways downstream of Calcr. Altogether, the findings demonstrated that Calcr exerts its effect specifically by keeping MuSCs in a quiescent state and in their location, maintaining the MuSC pool.


Subject(s)
Myoblasts/metabolism , Receptors, Calcitonin/metabolism , Second Messenger Systems , Stem Cell Niche , Acetylcysteine/analogs & derivatives , Acetylcysteine/metabolism , Animals , Apoptosis , Cell Differentiation , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Erythromycin/analogs & derivatives , Erythromycin/metabolism , Mice , Myoblasts/cytology , Myoblasts/physiology , Receptors, Calcitonin/genetics
16.
Skelet Muscle ; 5: 30, 2015.
Article in English | MEDLINE | ID: mdl-26347253

ABSTRACT

BACKGROUND: Merosin-deficient congenital muscular dystrophy type-1A (MDC1A) is characterized by progressive muscular dystrophy and dysmyelinating neuropathy caused by mutations of the α2 chain of laminin-211, the predominant laminin isoform of muscles and nerves. MDC1A has no available treatment so far, although preclinical studies showed amelioration of the disease by the overexpression of miniagrin (MAG). MAG reconnects orphan laminin-211 receptors to other laminin isoforms available in the extracellular matrix of MDC1A mice. METHODS: Mesoangioblasts (MABs) are vessel-associated progenitors that can form the skeletal muscle and have been shown to restore defective protein levels and motor skills in animal models of muscular dystrophies. As gene therapy in humans still presents challenging technical issues and limitations, we engineered MABs to overexpress MAG to treat MDC1A mouse models, thus combining cell to gene therapy. RESULTS: MABs synthesize and secrete only negligible amount of laminin-211 either in vitro or in vivo. MABs engineered to deliver MAG and injected in muscles of MDC1A mice showed amelioration of muscle histology, increased expression of laminin receptors in muscle, and attenuated deterioration of motor performances. MABs did not enter the peripheral nerves, thus did not affect the associated peripheral neuropathy. CONCLUSIONS: Our study demonstrates the potential efficacy of combining cell with gene therapy to treat MDC1A.

18.
Nat Commun ; 6: 6745, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25865621

ABSTRACT

Duchenne muscular dystrophy (DMD) is a chronic and life-threatening disease that is initially supported by muscle regeneration but eventually shows satellite cell exhaustion and muscular dysfunction. The life-long maintenance of skeletal muscle homoeostasis requires the satellite stem cell pool to be preserved. Asymmetric cell division plays a pivotal role in the maintenance of the satellite cell pool. Here we show that granulocyte colony-stimulating factor receptor (G-CSFR) is asymmetrically expressed in activated satellite cells. G-CSF positively affects the satellite cell population during multiple stages of differentiation in ex vivo cultured fibres. G-CSF could be important in developing an effective therapy for DMD based on its potential to modulate the supply of multiple stages of regenerated myocytes. This study shows that the G-CSF-G-CSFR axis is fundamentally important for long-term muscle regeneration, functional maintenance and lifespan extension in mouse models of DMD with varying severities.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Regeneration/drug effects , Satellite Cells, Skeletal Muscle/drug effects , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Disease Models, Animal , Gene Expression Regulation , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Longevity/drug effects , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Cells/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , MyoD Protein/genetics , MyoD Protein/metabolism , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , Primary Cell Culture , Receptors, Granulocyte Colony-Stimulating Factor/deficiency , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology
19.
Free Radic Biol Med ; 82: 122-36, 2015 May.
Article in English | MEDLINE | ID: mdl-25660994

ABSTRACT

High intensity training induces muscle damage in dystrophin-deficient mdx mice, an animal model for Duchenne muscular dystrophy. However, low intensity training (LIT) rescues the mdx phenotype and even reduces the level of protein carbonylation, a marker of oxidative damage. Until now, beneficial effects of LIT were mainly assessed at the physiological level. We investigated the effects of LIT at the molecular level on 8-week-old wild-type and mdx muscle using 2D Western blot and protein-protein interaction analysis. We found that the fast isoforms of troponin T and myosin binding protein C as well as glycogen phosphorylase were overcarbonylated and downregulated in mdx muscle. Some of the mitochondrial enzymes of the citric acid cycle were overcarbonylated, whereas some proteins of the respiratory chain were downregulated. Of functional importance, ATP synthase was only partially assembled, as revealed by Blue Native PAGE analysis. LIT decreased the carbonylation level and increased the expression of fast isoforms of troponin T and of myosin binding protein C, and glycogen phosphorylase. In addition, it increased the expression of aconitate hydratase and NADH dehydrogenase, and fully restored the ATP synthase complex. Our study demonstrates that the benefits of LIT are associated with lowered oxidative damage as revealed by carbonylation and higher expression of proteins involved in energy metabolism and muscle contraction. Potentially, these results will help to design therapies for DMD based on exercise mimicking drugs.


Subject(s)
Energy Metabolism/physiology , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/methods , Protein Carbonylation/physiology , Aconitate Hydratase/biosynthesis , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Citric Acid Cycle/physiology , Disease Models, Animal , Down-Regulation , Dystrophin/genetics , Glycogen Phosphorylase/biosynthesis , Glycogen Phosphorylase/genetics , Male , Mice , Mice, Inbred mdx , Mice, Transgenic , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondrial Proton-Translocating ATPases/metabolism , Muscular Dystrophy, Duchenne , NADH Dehydrogenase/biosynthesis , Oxidative Stress , Protein Isoforms/genetics , Troponin T/biosynthesis , Troponin T/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...