Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 30(11): 1006-1019, 2021 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-33822956

RESUMEN

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.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Animales , Calcio/metabolismo , Modelos Animales de Enfermedad , Distrofina/deficiencia , Humanos , Ratones , Ratones Endogámicos mdx , Contracción Muscular/genética , Debilidad Muscular/genética , Debilidad Muscular/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Distrofia Muscular de Duchenne/patología , Fenotipo , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/patología
2.
PLoS Genet ; 16(5): e1008826, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32453729

RESUMEN

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.


Asunto(s)
Nervio Coclear/metabolismo , Distroglicanos/genética , Pérdida Auditiva Sensorineural/metabolismo , Proteína Básica de Mielina/metabolismo , N-Acetilglucosaminiltransferasas/genética , Síndrome de Walker-Warburg/fisiopatología , Adolescente , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Técnicas de Inactivación de Genes , Glicosilación , Pérdida Auditiva Sensorineural/etiología , Pérdida Auditiva Sensorineural/genética , Humanos , Lactante , Masculino , Ratones , Síndrome de Walker-Warburg/complicaciones , Síndrome de Walker-Warburg/genética , Adulto Joven
3.
Int J Mol Sci ; 22(7)2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33807264

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Regeneración/fisiología , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Diferenciación Celular , Uniones Célula-Matriz/metabolismo , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Fibras Musculares Esqueléticas/fisiología , Músculo Esquelético/metabolismo , Enfermedades Musculares/metabolismo , Tendones/metabolismo , Cicatrización de Heridas/fisiología
4.
BMC Musculoskelet Disord ; 21(1): 479, 2020 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-32693782

RESUMEN

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.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Óxido Nítrico Sintasa de Tipo II , Canal Liberador de Calcio Receptor de Rianodina , Animales , Distrofina/genética , Distrofina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Canal Liberador de Calcio Receptor de Rianodina/genética
5.
Development ; 142(1): 51-61, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25480916

RESUMEN

Muscle satellite cells are indispensable for muscle regeneration, but the functional diversity of their daughter cells is unknown. Here, we show that many Pax7(+)MyoD(-) cells locate both beneath and outside the basal lamina during myofiber maturation. A large majority of these Pax7(+)MyoD(-) cells are not self-renewed satellite cells, but have different potentials for both proliferation and differentiation from Pax7(+)MyoD(+) myoblasts (classical daughter cells), and are specifically marked by expression of the doublecortin (Dcx) gene. Transplantation and lineage-tracing experiments demonstrated that Dcx-expressing cells originate from quiescent satellite cells and that the microenvironment induces Dcx in myoblasts. Expression of Dcx seems to be necessary for myofiber maturation because Dcx-deficient mice exhibited impaired myofiber maturation resulting from a decrease in the number of myonuclei. Furthermore, in vitro and in vivo studies suggest that one function of Dcx in myogenic cells is acceleration of cell motility. These results indicate that Dcx is a new marker for the Pax7(+)MyoD(-) subpopulation, which contributes to myofiber maturation during muscle regeneration.


Asunto(s)
Diferenciación Celular , Proteínas Asociadas a Microtúbulos/metabolismo , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Neuropéptidos/metabolismo , Regeneración/fisiología , Células Madre/citología , Animales , Cardiotoxinas/administración & dosificación , Movimiento Celular , Microambiente Celular , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/deficiencia , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/metabolismo , Neuropéptidos/deficiencia , Factor de Transcripción PAX7/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Madre/metabolismo
6.
Hum Mol Genet ; 22(15): 3003-15, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23562821

RESUMEN

A group of muscular dystrophies, dystroglycanopathy is caused by abnormalities in post-translational modifications of dystroglycan (DG). To understand better the pathophysiological roles of DG modification and to establish effective clinical treatment for dystroglycanopathy, we here generated two distinct conditional knock-out (cKO) mice for fukutin, the first dystroglycanopathy gene identified for Fukuyama congenital muscular dystrophy. The first dystroglycanopathy model-myofiber-selective fukutin-cKO [muscle creatine kinase (MCK)-fukutin-cKO] mice-showed mild muscular dystrophy. Forced exercise experiments in presymptomatic MCK-fukutin-cKO mice revealed that myofiber membrane fragility triggered disease manifestation. The second dystroglycanopathy model-muscle precursor cell (MPC)-selective cKO (Myf5-fukutin-cKO) mice-exhibited more severe phenotypes of muscular dystrophy. Using an isolated MPC culture system, we demonstrated, for the first time, that defects in the fukutin-dependent modification of DG lead to impairment of MPC proliferation, differentiation and muscle regeneration. These results suggest that impaired MPC viability contributes to the pathology of dystroglycanopathy. Since our data suggested that frequent cycles of myofiber degeneration/regeneration accelerate substantial and/or functional loss of MPC, we expected that protection from disease-triggering myofiber degeneration provides therapeutic effects even in mouse models with MPC defects; therefore, we restored fukutin expression in myofibers. Adeno-associated virus (AAV)-mediated rescue of fukutin expression that was limited in myofibers successfully ameliorated the severe pathology even after disease progression. In addition, compared with other gene therapy studies, considerably low AAV titers were associated with therapeutic effects. Together, our findings indicated that fukutin-deficient dystroglycanopathy is a regeneration-defective disorder, and gene therapy is a feasible treatment for the wide range of dystroglycanopathy even after disease progression.


Asunto(s)
Distroglicanos/metabolismo , Expresión Génica , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Mioblastos/metabolismo , Fenotipo , Animales , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Glicosilación , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Factor 5 Regulador Miogénico/genética , Proteínas/genética , Proteínas/metabolismo , Transferasas
7.
Glycobiology ; 24(3): 314-24, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24352591

RESUMEN

The human natural killer-1 (HNK-1) carbohydrate comprising a sulfated trisaccharide (HSO3-3GlcAß1-3Galß1-4GlcNAc-) is expressed on N-linked and O-mannose-linked glycans in the nervous system and involved in learning and memory functions. Although whole/core glycan structures and carrier glycoproteins for the N-linked HNK-1 epitope have been studied, carrier glycoproteins and the biosynthetic pathway of the O-mannose-linked HNK-1 epitope have not been fully characterized. Here, using mass spectrometric analyses, we identified the major carrier glycoprotein of the O-linked HNK-1 as phosphacan in developing mouse brains and determined the major O-glycan structures having the terminal HNK-1 epitope from partially purified phosphacan. The O-linked HNK-1 epitope on phosphacan almost disappeared due to the knockout of protein O-mannose ß1,2-N-acetylglucosaminyltransferase 1, an N-acetylglucosaminyltransferase essential for O-mannose-linked glycan synthesis, indicating that the reducing terminal of the O-linked HNK-1 is mannose. We also showed that glucuronyltransferase-P (GlcAT-P) was involved in the biosynthesis of O-mannose-linked HNK-1 using the gene-deficient mice of GlcAT-P, one of the glucuronyltransferases for HNK-1 synthesis. Consistent with this result, we revealed that GlcAT-P specifically synthesized O-linked HNK-1 onto phosphacan using cultured cells. Furthermore, we characterized the as-yet-unknown epitope of the 6B4 monoclonal antibody (mAb), which was thought to recognize a unique phosphacan glycoform. The reactivity of the 6B4 mAb almost completely disappeared in GlcAT-P-deficient mice, and exogenously expressed phosphacan was selectively recognized by the 6B4 mAb when co-expressed with GlcAT-P, suggesting that the 6B4 mAb preferentially recognizes O-mannose-linked HNK-1 on phosphacan. This is the first study to show that 6B4 mAb-reactive O-mannose-linked HNK-1 in the brain is mainly carried by phosphacan.


Asunto(s)
Encéfalo/metabolismo , Antígenos CD57/metabolismo , Manosa/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/metabolismo , Animales , Encéfalo/crecimiento & desarrollo , Antígenos CD57/química , Células COS , Conformación de Carbohidratos , Chlorocebus aethiops , Glucuronosiltransferasa/metabolismo , Glicosilación , Células HEK293 , Humanos , Manosa/química , Ratones , Ratones Endogámicos C57BL , N-Acetilglucosaminiltransferasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 5 Similares a Receptores/química
8.
J Cell Sci ; 125(Pt 5): 1309-17, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22349695

RESUMEN

Satellite cells are muscle stem cells that have important roles in postnatal muscle growth and adult muscle regeneration. Although fast- and slow-dividing populations in activated satellite cells have been observed, the functional differences between them remain unclear. Here we elucidated the relationship between proliferation behaviour and satellite cell function. To assess the frequency of cell division, satellite cells isolated from mouse EDL muscle were labelled with the fluorescent dye PKH26, stimulated to proliferate and then sorted by FACS. The vast majority of activated satellite cells were PKH26(low) fast-dividing cells, whereas PKH26(high) slow-dividing cells were observed as a minority population. The fast-dividing cells generated a higher number of differentiated and self-renewed cells compared with the slow-dividing cells. However, cells derived from the slow-dividing population formed secondary myogenic colonies when passaged, whereas those from the fast-dividing population rapidly underwent myogenic differentiation without producing self-renewing cells after a few rounds of cell division. Furthermore, slow-dividing cells transplanted into injured muscle extensively contributed to muscle regeneration in vivo. Id1, a HLH protein, was expressed by all activated satellite cells, but the expression level varied within the slow-dividing cell population. We show that the slow-dividing cells retaining long-term self-renewal ability are restricted to an undifferentiated population that express high levels of Id1 protein (PKH26(high)Id1(high) population). Finally, genome-wide gene expression analysis described the molecular characteristics of the PKH26(high)Id1(high) population. Taken together, our results indicate that undifferentiated slow-dividing satellite cells retain stemness for generating progeny capable of long-term self-renewal, and so might be essential for muscle homeostasis throughout life.


Asunto(s)
División Celular , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/fisiología , Regeneración , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Citometría de Flujo , Colorantes Fluorescentes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Fibras Musculares Esqueléticas/metabolismo
9.
Development ; 138(21): 4609-19, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21989910

RESUMEN

Satellite cells, which are skeletal muscle stem cells, divide to provide new myonuclei to growing muscle fibers during postnatal development, and then are maintained in an undifferentiated quiescent state in adult skeletal muscle. This state is considered to be essential for the maintenance of satellite cells, but their molecular regulation is unknown. We show that Hesr1 (Hey1) and Hesr3 (Heyl) (which are known Notch target genes) are expressed simultaneously in skeletal muscle only in satellite cells. In Hesr1 and Hesr3 single-knockout mice, no obvious abnormalities of satellite cells or muscle regenerative potentials are observed. However, the generation of undifferentiated quiescent satellite cells is impaired during postnatal development in Hesr1/3 double-knockout mice. As a result, myogenic (MyoD and myogenin) and proliferative (Ki67) proteins are expressed in adult satellite cells. Consistent with the in vivo results, Hesr1/3-null myoblasts generate very few Pax7(+) MyoD(-) undifferentiated cells in vitro. Furthermore, the satellite cell number gradually decreases in Hesr1/3 double-knockout mice even after it has stabilized in control mice, and an age-dependent regeneration defect is observed. In vivo results suggest that premature differentiation, but not cell death, is the reason for the reduced number of satellite cells in Hesr1/3 double-knockout mice. These results indicate that Hesr1 and Hesr3 are essential for the generation of adult satellite cells and for the maintenance of skeletal muscle homeostasis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/fisiología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores/metabolismo , Peso Corporal , Recuento de Células , Proteínas de Ciclo Celular/genética , Proliferación Celular , Células Cultivadas , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Tamaño de los Órganos , Fenotipo
10.
Muscle Nerve ; 49(5): 728-35, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24037898

RESUMEN

INTRODUCTION: α1-syntrophin, a member of the dystrophin complex, recruits membrane molecules, including aquaporin-4, at the sarcolemma. The physiological functions of α1-syntrophin are poorly understood. METHODS: We examined the physiological characteristics of α1-syntrophin-deficient muscles under osmotic stress conditions to test the possibility that mutant muscles are less tolerant of osmotic shock. RESULTS: Isolated muscle bundles from mutant mice showed markedly reduced force production after hypo-osmotic shock. In addition, the mutant muscle bundles showed delayed recovery of specific gravity after being exposed to hypo-osmotic conditions. Two consecutive exercise tests on the treadmill revealed their performance in the second test was significantly lower than for wild-type mice. Furthermore, mutant mice had higher serum lactate concentrations after treadmill exercise. CONCLUSIONS: Although the lack of α1-syntrophin from the sarcolemma does not lead to muscle degeneration, our results suggest that it may be partly involved in the pathophysiology of dystrophin-deficient Duchenne muscular dystrophy.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Proteínas de la Membrana/fisiología , Fatiga Muscular/genética , Fibras Musculares Esqueléticas/fisiología , Proteínas Musculares/fisiología , Fuerza Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/fisiopatología , Presión Osmótica/fisiología , Animales , Proteínas de Unión al Calcio/deficiencia , Proteínas de Unión al Calcio/genética , Modelos Animales de Enfermedad , Técnicas In Vitro , Ácido Láctico/sangre , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Fatiga Muscular/fisiología , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Fuerza Muscular/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética
11.
J Cell Sci ; 124(Pt 21): 3654-64, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22045730

RESUMEN

Accumulation of adipocytes and collagen type-I-producing cells (fibrosis) is observed in muscular dystrophies. The origin of these cells had been largely unknown, but recently we identified mesenchymal progenitors positive for platelet-derived growth factor receptor alpha (PDGFRα) as the origin of adipocytes in skeletal muscle. However, the origin of muscle fibrosis remains largely unknown. In this study, clonal analyses show that PDGFRα(+) cells also differentiate into collagen type-I-producing cells. In fact, PDGFRα(+) cells accumulated in fibrotic areas of the diaphragm in the mdx mouse, a model of Duchenne muscular dystrophy. Furthermore, mRNA of fibrosis markers was expressed exclusively in the PDGFRα(+) cell fraction in the mdx diaphragm. Importantly, TGF-ß isoforms, known as potent profibrotic cytokines, induced expression of markers of fibrosis in PDGFRα(+) cells but not in myogenic cells. Transplantation studies revealed that fibrogenic PDGFRα(+) cells mainly derived from pre-existing PDGFRα(+) cells and that the contribution of PDGFRα(-) cells and circulating cells was limited. These results indicate that mesenchymal progenitors are the main origin of not only fat accumulation but also fibrosis in skeletal muscle.


Asunto(s)
Adipogénesis , Fibrosis/fisiopatología , Células Madre Mesenquimatosas/citología , Músculo Esquelético/citología , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Fibrosis/genética , Fibrosis/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
13.
FASEB Bioadv ; 5(11): 453-469, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37936920

RESUMEN

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.

14.
Glia ; 60(10): 1451-67, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22706957

RESUMEN

The laminin family of extracellular matrix proteins are expressed broadly during embryonic brain development, but are enriched at ventricular and pial surfaces where laminins mediate radial glial attachment during corticogenesis. In the adult brain, however, laminin distribution is restricted, yet is found within the vascular basal lamina and associated fractones of the ventricular zone (VZ)-subventricular zone (SVZ) stem cell niche, where laminins regulate adult neural progenitor cell proliferation. It remains unknown, however, if laminins regulate the wave of oligodendrogenesis that occurs in the neonatal/early postnatal VZ-SVZ. Here we report that Lama2, the gene that encodes the laminin α2-subunit, regulates postnatal oligodendrogenesis. At birth, Lama2-/- mice had significantly higher levels of dying oligodendrocyte progenitor cells (OPCs) in the OPC germinal zone of the dorsal SVZ. This translated into fewer OPCs, both in the dorsal SVZ well as in an adjacent developing white matter tract, the corpus callosum. In addition, intermediate progenitor cells that give rise to OPCs in the Lama2-/- VZ-SVZ were mislocalized and proliferated nearer to the ventricle surface. Later, delays in oligodendrocyte maturation (with accompanying OPC accumulation), were observed in the Lama2-/- corpus callosum, leading to dysmyelination by postnatal day 21. Together these data suggest that prosurvival laminin interactions in the developing postnatal VZ-SVZ germinal zone regulate the ability, or timing, of oligodendrocyte production to occur appropriately.


Asunto(s)
Diferenciación Celular/fisiología , Ventrículos Cerebrales/citología , Regulación del Desarrollo de la Expresión Génica/genética , Laminina/metabolismo , Oligodendroglía/fisiología , Células Madre/fisiología , Factores de Edad , Animales , Animales Recién Nacidos , Antígenos/metabolismo , Proliferación Celular , Supervivencia Celular/genética , Proteínas del Ojo/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas de Homeodominio/metabolismo , Etiquetado Corte-Fin in Situ , Laminina/deficiencia , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Fibras Nerviosas Mielínicas/fisiología , Oligodendroglía/ultraestructura , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteoglicanos/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción SOXB1/genética , Células Madre/ultraestructura
15.
Mol Cell Neurosci ; 47(2): 119-30, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21447391

RESUMEN

Protein O-linked mannose beta1, 2-N-acetylglucosaminyltransferase 1 (POMGnT1) is an enzyme that catalyzes the transfer of N-acetylglucosamine to O-mannose of glycoproteins. Alpha-dystroglycan, a substrate of POMGnT1, is concentrated around the blood vessels, in the outer plexiform layer (OPL), and in the inner limiting membrane (ILM) of the retina. Mutations of the POMGnT1 gene in humans cause muscle-eye-brain (MEB) disease. Several ocular abnormalities including retinal dysplasia, ERG abnormalities, and retinal detachments have been reported in patients with MEB. We have analyzed the eyes of POMGnT1-deficient mice, generated by standard gene targeting technique, to study the retinal abnormalities. Clinical examination of adult mutant mice revealed a high incidence (81% by 12-months-of-age) of retinal detachments. Sheathing of the retinal vessels and the presence of ectopic fibrous tissues around the optic nerve head were also found. Histological examinations showed focal retinal detachment associated with GFAP immunopositivity. The ILM of the mutant mice was disrupted with ectopic cells near the disruptions. The expression of Dp71, a shorter isoform of dystrophin, was severely reduced in the ILM and around retinal blood vessels of POMGnT1-deficient mice. The expression of Dp427, Dp260, Dp140 were also reduced in the OPL of the mutant mice. Electroretinographic (ERG) analyses showed reduced a- and b-wave amplitudes. Examinations of flat mounts revealed abnormal vascular network associated with highly irregular astrocytic processes. In addition, ER-TR7-positive fibrous tissue was found closely associated with reactive astrocytes especially around the optic nerve head. Our results suggest that altered glycosylation of alpha-DG may be responsible for the reactive gliosis and reticular fibrosis in the retina, and the subsequent developments of retinal dysplasia, abnormal ERGs, and retinal detachment in the mutant mice.


Asunto(s)
Astrocitos/metabolismo , Gliosis/patología , N-Acetilglucosaminiltransferasas/genética , Retina/citología , Retina/patología , Animales , Astrocitos/patología , Distroglicanos/genética , Distroglicanos/metabolismo , Distrofina/genética , Distrofina/metabolismo , Electrorretinografía , Gliosis/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , N-Acetilglucosaminiltransferasas/metabolismo , Retina/fisiología , Retina/fisiopatología , Desprendimiento de Retina/metabolismo , Desprendimiento de Retina/patología
16.
J Biol Chem ; 285(41): 31208-16, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20682766

RESUMEN

Pikachurin, the most recently identified ligand of dystroglycan, plays a crucial role in the formation of the photoreceptor ribbon synapse. It is known that glycosylation of dystroglycan is necessary for its ligand binding activity, and hypoglycosylation is associated with a group of muscular dystrophies that often involve eye abnormalities. Because little is known about the interaction between pikachurin and dystroglycan and its impact on molecular pathogenesis, here we characterize the interaction using deletion constructs and mouse models of muscular dystrophies with glycosylation defects (Large(myd) and POMGnT1-deficient mice). Pikachurin-dystroglycan binding is calcium-dependent and relatively less sensitive to inhibition by heparin and high NaCl concentration, as compared with other dystroglycan ligand proteins. Using deletion constructs of the laminin globular domains in the pikachurin C terminus, we show that a certain steric structure formed by the second and the third laminin globular domains is necessary for the pikachurin-dystroglycan interaction. Binding assays using dystroglycan deletion constructs and tissue samples from Large-deficient (Large(myd)) mice show that Large-dependent modification of dystroglycan is necessary for pikachurin binding. In addition, the ability of pikachurin to bind to dystroglycan prepared from POMGnT1-deficient mice is severely reduced, suggesting that modification of the GlcNAc-ß1,2-branch on O-mannose is also necessary for the interaction. Immunofluorescence analysis reveals a disruption of pikachurin localization in the photoreceptor ribbon synapse of these model animals. Together, our data demonstrate that post-translational modification on O-mannose, which is mediated by Large and POMGnT1, is essential for pikachurin binding and proper localization, and suggest that their disruption underlies the molecular pathogenesis of eye abnormalities in a group of muscular dystrophies.


Asunto(s)
Proteínas Portadoras/metabolismo , Distroglicanos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Procesamiento Proteico-Postraduccional , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Calcio/metabolismo , Proteínas Portadoras/genética , Línea Celular , Distroglicanos/genética , Glicosilación , Humanos , Ratones , Ratones Mutantes , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Proteínas del Tejido Nervioso/genética , Células Fotorreceptoras de Vertebrados/patología , Unión Proteica , Estructura Terciaria de Proteína , Eliminación de Secuencia , Sinapsis/genética , Sinapsis/patología
17.
Hum Mol Genet ; 18(4): 621-31, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19017726

RESUMEN

Hypoglycosylation and reduced laminin-binding activity of alpha-dystroglycan are common characteristics of dystroglycanopathy, which is a group of congenital and limb-girdle muscular dystrophies. Fukuyama-type congenital muscular dystrophy (FCMD), caused by a mutation in the fukutin gene, is a severe form of dystroglycanopathy. A retrotransposal insertion in fukutin is seen in almost all cases of FCMD. To better understand the molecular pathogenesis of dystroglycanopathies and to explore therapeutic strategies, we generated knock-in mice carrying the retrotransposal insertion in the mouse fukutin ortholog. Knock-in mice exhibited hypoglycosylated alpha-dystroglycan; however, no signs of muscular dystrophy were observed. More sensitive methods detected minor levels of intact alpha-dystroglycan, and solid-phase assays determined laminin binding levels to be approximately 50% of normal. In contrast, intact alpha-dystroglycan is undetectable in the dystrophic Large(myd) mouse, and laminin-binding activity is markedly reduced. These data indicate that a small amount of intact alpha-dystroglycan is sufficient to maintain muscle cell integrity in knock-in mice, suggesting that the treatment of dystroglycanopathies might not require the full recovery of glycosylation. To examine whether glycosylation defects can be restored in vivo, we performed mouse gene transfer experiments. Transfer of fukutin into knock-in mice restored glycosylation of alpha-dystroglycan. In addition, transfer of LARGE produced laminin-binding forms of alpha-dystroglycan in both knock-in mice and the POMGnT1 mutant mouse, which is another model of dystroglycanopathy. Overall, these data suggest that even partial restoration of alpha-dystroglycan glycosylation and laminin-binding activity by replacing or augmenting glycosylation-related genes might effectively deter dystroglycanopathy progression and thus provide therapeutic benefits.


Asunto(s)
Distroglicanos/metabolismo , Laminina/metabolismo , Distrofias Musculares/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Modelos Animales de Enfermedad , Técnicas de Sustitución del Gen , Glicosilación , Humanos , Laminina/genética , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/terapia , Mutagénesis Insercional , N-Acetilglucosaminiltransferasas/genética , Unión Proteica , Proteínas/genética , Proteínas/metabolismo , Transferasas
18.
Am J Pathol ; 176(5): 2414-24, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20304955

RESUMEN

Duchenne muscular dystrophy (DMD) is the most common lethal genetic disorder of children. The mdx (C57BL/10 background, C57BL/10-mdx) mouse is a widely used model of DMD, but the histopathological hallmarks of DMD, such as the smaller number of myofibers, accumulation of fat and fibrosis, and insufficient regeneration of myofibers, are not observed in adult C57BL/10-mdx except for in the diaphragm. In this study, we showed that DBA/2 mice exhibited decreased muscle weight, as well as lower myofiber numbers after repeated degeneration-regeneration cycles. Furthermore, the self-renewal efficiency of satellite cells of DBA/2 is lower than that of C57BL/6. Therefore, we produced a DBA/2-mdx strain by crossing DBA/2 and C57BL/10-mdx. The hind limb muscles of DBA/2-mdx mice exhibited lower muscle weight, fewer myofibers, and increased fat and fibrosis, in comparison with C57BL/10-mdx. Moreover, remarkable muscle weakness was observed in DBA/2-mdx. These results indicate that the DBA/2-mdx mouse is a more suitable model for DMD studies, and the efficient satellite cell self-renewal ability of C57BL/10-mdx might explain the difference in pathologies between humans and mice.


Asunto(s)
Células Satélite del Músculo Esquelético/citología , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos mdx , Modelos Biológicos , Modelos Genéticos , Músculo Esquelético/patología , Fenotipo
19.
Exp Cell Res ; 316(18): 3087-92, 2010 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-20580709

RESUMEN

The molecular mechanisms of Duchenne muscular dystrophy (DMD) have been extensively investigated since the discovery of the dystrophin gene in 1986. Nonetheless, there is currently no effective treatment for DMD. Recent reports, however, indicate that adenoassociated viral (AAV) vector-mediated transfer of a functional dystrophin cDNA into the affected muscle is a promising strategy. In addition, antisense-mediated exon skipping technology has been emerging as another promising approach to restore dystrophin expression in DMD muscle. Ongoing clinical trials show restoration of dystrophin in DMD patients without serious side effects. Here, we summarize the recent progress in gene therapy, with an emphasis on exon skipping for DMD.


Asunto(s)
Distrofina/genética , Terapia Genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Exones/genética , Humanos
20.
Exp Cell Res ; 316(17): 2932-44, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20696153

RESUMEN

Muscle satellite cells are essential for muscle growth and regeneration and their morphology, behavior and gene expression have been extensively studied. However, the mechanisms involved in their proliferation and differentiation remain elusive. Six1 and Six4 proteins were expressed in the nuclei of myofibers of adult mice and the numbers of myoblasts positive for Six1 and Six4 increased during regeneration of skeletal muscles. Six1 and Six4 were expressed in quiescent, activated and differentiated muscle satellite cells isolated from adult skeletal muscle. Overexpression of Six4 and Six5 repressed the proliferation and differentiation of satellite cells. Conversely, knockdown of Six5 resulted in augmented proliferation, and that of Six4 inhibited differentiation. Muscle satellite cells isolated from Six4(+/-)Six5(-/-) mice proliferated to higher cell density though their differentiation was not altered. Meanwhile, overproduction of Six1 repressed proliferation and promoted differentiation of satellite cells. In addition, Six4 and Six5 repressed, while Six1 activated myogenin expression, suggesting that the differential regulation of myogenin expression is responsible for the differential effects of Six genes. The results indicated the involvement of Six genes in the behavior of satellite cells and identified Six genes as potential target for manipulation of proliferation and differentiation of muscle satellite cells for therapeutic applications.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular , Proteínas de Homeodominio/genética , Células Satélite del Músculo Esquelético/citología , Transactivadores/genética , Adulto , Animales , Regulación de la Expresión Génica/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Ratones , Ratones Noqueados , Células Musculares/citología , Miogenina/biosíntesis , Transactivadores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA