Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
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.
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
4.
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
5.
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
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...