Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Circ Res ; 135(5): 554-574, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39011635

RESUMEN

BACKGROUND: Cardiac hypertrophy compensates for increased biomechanical stress of the heart induced by prevalent cardiovascular pathologies but can result in heart failure if left untreated. Here, we hypothesized that the membrane fusion and repair protein dysferlin is critical for the integrity of the transverse-axial tubule (TAT) network inside cardiomyocytes and contributes to the proliferation of TAT endomembranes during pressure overload-induced cardiac hypertrophy. METHODS: Stimulated emission depletion and electron microscopy were used to localize dysferlin in mouse and human cardiomyocytes. Data-independent acquisition mass spectrometry revealed the cardiac dysferlin interactome and proteomic changes of the heart in dysferlin-knockout mice. After transverse aortic constriction, we compared the hypertrophic response of wild-type versus dysferlin-knockout hearts and studied TAT network remodeling mechanisms inside cardiomyocytes by live-cell membrane imaging. RESULTS: We localized dysferlin in a vesicular compartment in nanometric proximity to contact sites of the TAT network with the sarcoplasmic reticulum, a.k.a. junctional complexes for Ca2+-induced Ca2+ release. Interactome analyses demonstrated a novel protein interaction of dysferlin with the membrane-tethering sarcoplasmic reticulum protein juncophilin-2, a putative interactor of L-type Ca2+ channels and ryanodine receptor Ca2+ release channels in junctional complexes. Although the dysferlin-knockout caused a mild progressive phenotype of dilated cardiomyopathy, global proteome analysis revealed changes preceding systolic failure. Following transverse aortic constriction, dysferlin protein expression was significantly increased in hypertrophied wild-type myocardium, while dysferlin-knockout animals presented markedly reduced left-ventricular hypertrophy. Live-cell membrane imaging showed a profound reorganization of the TAT network in wild-type left-ventricular myocytes after transverse aortic constriction with robust proliferation of axial tubules, which critically depended on the increased expression of dysferlin within newly emerging tubule components. CONCLUSIONS: Dysferlin represents a new molecular target in cardiac disease that protects the integrity of tubule-sarcoplasmic reticulum junctional complexes for regulated excitation-contraction coupling and controls TAT network reorganization and tubular membrane proliferation in cardiomyocyte hypertrophy induced by pressure overload.


Asunto(s)
Cardiomegalia , Disferlina , Ratones Noqueados , Miocitos Cardíacos , Retículo Sarcoplasmático , Animales , Disferlina/metabolismo , Disferlina/genética , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Humanos , Ratones , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/patología , Ratones Endogámicos C57BL , Masculino , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proliferación Celular , Células Cultivadas , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Quinasa de Cadena Ligera de Miosina
2.
FASEB J ; 38(20): e70099, 2024 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-39400395

RESUMEN

Alzheimer's disease (AD) is the most common neurodegenerative disease, and a defect in neuronal plasma membrane repair could exacerbate neurotoxicity, neuronal death, and disease progression. In this study, application of AD patient cerebrospinal fluid (CSF) and recombinant human Aß to otherwise healthy neurons induces defective neuronal plasma membrane repair in vitro and ex vivo. We identified Aß as the biochemical component in patient CSF leading to compromised repair capacity and depleting Aß rescued repair capacity. These elevated Aß levels reduced expression of dysferlin, a protein that facilitates membrane repair, by altering autophagy and reducing dysferlin trafficking to sites of membrane injury. Overexpression of dysferlin and autophagy inhibition rescued membrane repair. Overall, these findings indicate an AD pathogenic mechanism where Aß impairs neuronal membrane repair capacity and increases susceptibility to cell death. This suggests that membrane repair could be therapeutically targeted in AD to restore membrane integrity and reduce neurotoxicity and neuronal death.


Asunto(s)
Enfermedad de Alzheimer , Membrana Celular , Disferlina , Neuronas , Humanos , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Disferlina/metabolismo , Disferlina/genética , Neuronas/metabolismo , Membrana Celular/metabolismo , Péptidos beta-Amiloides/metabolismo , Autofagia , Masculino , Femenino , Anciano , Anciano de 80 o más Años , Animales , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética
3.
J Physiol ; 602(9): 1893-1910, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615232

RESUMEN

Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.


Asunto(s)
Calcio , Disferlina , Humanos , Calcio/metabolismo , Disferlina/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/fisiología , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiología , Músculo Estriado/metabolismo , Músculo Estriado/fisiología
4.
Int J Mol Sci ; 25(11)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38891760

RESUMEN

Dysferlin is a large transmembrane protein involved in critical cellular processes including membrane repair and vesicle fusion. Mutations in the dysferlin gene (DYSF) can result in rare forms of muscular dystrophy; Miyoshi myopathy; limb girdle muscular dystrophy type 2B (LGMD2B); and distal myopathy. These conditions are collectively known as dysferlinopathies and are caused by more than 600 mutations that have been identified across the DYSF gene to date. In this review, we discuss the key molecular and clinical features of LGMD2B, the causative gene DYSF, and the associated dysferlin protein structure. We also provide an update on current approaches to LGMD2B diagnosis and advances in drug development, including splice switching antisense oligonucleotides. We give a brief update on clinical trials involving adeno-associated viral gene therapy and the current progress on CRISPR/Cas9 mediated therapy for LGMD2B, and then conclude by discussing the prospects of antisense oligomer-based intervention to treat selected mutations causing dysferlinopathies.


Asunto(s)
Disferlina , Terapia Genética , Distrofia Muscular de Cinturas , Mutación , Humanos , Distrofia Muscular de Cinturas/terapia , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/diagnóstico , Disferlina/genética , Disferlina/metabolismo , Terapia Genética/métodos , Oligonucleótidos Antisentido/uso terapéutico , Animales
5.
Int J Mol Sci ; 24(5)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36902136

RESUMEN

Quantitative surface plasmon resonance (SPR) was utilized to determine binding strength and calcium dependence of direct interactions between dysferlin and proteins likely to mediate skeletal muscle repair, interrupted in limb girdle muscular dystrophy type 2B/R2. Dysferlin canonical C2A (cC2A) and C2F/G domains directly interacted with annexin A1, calpain-3, caveolin-3, affixin, AHNAK1, syntaxin-4, and mitsugumin-53, with cC2A the primary target and C2F lesser involved, overall demonstrating positive calcium dependence. Dysferlin C2 pairings alone showed negative calcium dependence in almost all cases. Like otoferlin, dysferlin directly interacted via its carboxy terminus with FKBP8, an anti-apoptotic outer mitochondrial membrane protein, and via its C2DE domain with apoptosis-linked gene (ALG-2/PDCD6), linking anti-apoptosis with apoptosis. Confocal Z-stack immunofluorescence confirmed co-compartmentalization of PDCD6 and FKBP8 at the sarcolemmal membrane. Our evidence supports the hypothesis that prior to injury, dysferlin C2 domains self-interact and give rise to a folded, compact structure as indicated for otoferlin. With elevation of intracellular Ca2+ in injury, dysferlin would unfold and expose the cC2A domain for interaction with annexin A1, calpain-3, mitsugumin 53, affixin, and caveolin-3, and dysferlin would realign from its interactions with PDCD6 at basal calcium levels to interact strongly with FKBP8, an intramolecular rearrangement facilitating membrane repair.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Proteínas de Unión al Calcio , Calcio , Disferlina , Músculo Esquelético , Regeneración , Proteínas de Unión a Tacrolimus , Anexina A1/metabolismo , Calcio/metabolismo , Calpaína/metabolismo , Caveolina 3/metabolismo , Disferlina/metabolismo , Músculo Esquelético/lesiones , Músculo Esquelético/fisiología , Sarcolema/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Resonancia por Plasmón de Superficie , Proteínas de Unión a Tacrolimus/metabolismo , Apoptosis , Transducción de Señal , Animales , Ratas
6.
Bull Exp Biol Med ; 174(6): 768-773, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37160600

RESUMEN

We studied the effects of a dual-vector DYSF gene delivery system based on adeno-associated virus serotype 9 capsids on pathological manifestations of dysferlinopathy in skeletal muscles of Bla/J mice lacking DYSF expression. The mice received intravenous injection of 3×1013 genomic copies of the virus containing the dual-vector system. M. gastrocnemius, m. psoas major, m. vastus lateralis, and m. gluteus superficialis were isolated for histological examination in 3, 6, and 12 weeks after treatment. Healthy wild-type (C57BL/6) mice served as positive control and were sacrificed 3 weeks after injection of 150 µl of 0.9% NaCl into the caudal vein. To detect dysferlin in muscle cryosections, immunohistochemical analysis with diagnostic antibodies was performed; paraffin sections were stained with hematoxylin and eosin for morphometric analysis. After administration of gene-therapeutic constructs, muscle fibers with membrane or cytoplasmic dysferlin location were detected in all examined muscles. The proportion of necrotic muscle fibers decreased, the number of muscle fibers with central location of the nucleus increased, and the mean cross-section area of the muscle fibers decreased.


Asunto(s)
Músculo Esquelético , Distrofia Muscular de Cinturas , Ratones , Animales , Disferlina/genética , Disferlina/metabolismo , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/terapia , Distrofia Muscular de Cinturas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Técnicas de Transferencia de Gen
7.
Muscle Nerve ; 66(4): 513-522, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35859452

RESUMEN

INTRODUCTION/AIMS: Most mouse models of muscular dystrophy (MD) show mild phenotypes, which limits the translatability of experimental therapies to patients. A growing body of evidence suggests that MD is accompanied by metabolic abnormalities that could potentially exacerbate the primary muscle wasting process. Since thermoneutral (TN) housing of mice (~30°C) has been shown to affect many metabolic parameters, particularly when combined with a Western diet (WD), our aim was to determine whether the combination of TN and WD exacerbates muscle wasting in dysferlin-deficient BLAJ mice, a common model of limb-girdle MD type 2b (LGMD2b). METHODS: The 2-mo-old wild-type (WT) and BLAJ mice were housed at TN or room temperature (RT) and fed a WD or regular chow for 9 mo. Ambulatory function, muscle histology, and protein immunoblots of skeletal muscle were assessed. RESULTS: BLAJ mice at RT and fed a chow diet showed normal ambulation function similar to WT mice, whereas 90% of BLAJ mice under WD and TN combination showed ambulatory dysfunction (p < 0.001), and an up to 4.1-fold increase in quadriceps and gastrocnemius fat infiltration. Western blotting revealed decreased autophagy marker microtubules-associated protein 1 light chain 3-B (LC3BII/LC3BI) ratio and up-regulation of protein kinase B/AKT and ribosomal protein S6 phosphorylation, suggesting inefficient cellular debris and protein clearance in TN BLAJ mice fed a WD. Male and female BLAJ mice under TN and WD combination showed heterogenous fibro-fatty infiltrate composition. DISCUSSION: TN and WD combination exacerbates rodent LGMD2b without affecting WT mice. This improves rodent modeling of human MD and helps elucidate how metabolic abnormalities may play a causal role in muscle wasting.


Asunto(s)
Distrofia Muscular de Cinturas , Distrofias Musculares , Animales , Dieta Occidental/efectos adversos , Disferlina/genética , Disferlina/metabolismo , Femenino , Vivienda , Humanos , Masculino , Ratones , Músculo Esquelético , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , Distrofias Musculares/patología , Distrofia Muscular de Cinturas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína S6 Ribosómica/metabolismo
8.
Biochem J ; 478(1): 197-215, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33449082

RESUMEN

The membrane protein dysferlin (DYSF) is important for calcium-activated plasma membrane repair, especially in muscle fibre cells. Nearly 600 mutations in the DYSF gene have been identified that are causative for rare genetic forms of muscular dystrophy. The dysferlin protein consists of seven C2 domains (C2A-C2G, 13%-33% identity) used to recruit calcium ions and traffic accessory proteins and vesicles to injured membrane sites needed to reseal a wound. Amongst these, the C2A is the most prominent facilitating the calcium-sensitive interaction with membrane surfaces. In this work, we determined the calcium-free and calcium-bound structures of the dysferlin C2A domain using NMR spectroscopy and X-ray crystallography. We show that binding two calcium ions to this domain reduces the flexibility of the Ca2+-binding loops in the structure. Furthermore, calcium titration and mutagenesis experiments reveal the tight coupling of these calcium-binding sites whereby the elimination of one site abolishes calcium binding to its partner site. We propose that the electrostatic potential distributed by the flexible, negatively charged calcium-binding loops in the dysferlin C2A domain control first contact with calcium that promotes subsequent binding. Based on these results, we hypothesize that dysferlin uses a 'calcium-catching' mechanism to respond to calcium influx during membrane repair.


Asunto(s)
Proteínas de Unión al Calcio/química , Calcio/química , Disferlina/química , Proteínas Musculares/química , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cristalografía por Rayos X , Disferlina/genética , Disferlina/metabolismo , Expresión Génica , Modelos Moleculares , Proteínas Musculares/metabolismo , Mutagénesis , Mutación , Resonancia Magnética Nuclear Biomolecular , Unión Proteica , Dominios Proteicos , Estructura Terciaria de Proteína , Electricidad Estática
9.
Ultrastruct Pathol ; 46(4): 359-367, 2022 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-35880824

RESUMEN

A number of sarcolemma proteins are responsible for muscle fiber repair. Dysferlin encoded by the DYSF gene is one of these proteins. Dysferlin promotes membrane repair in striated muscle fibers (MFs). Mutations in DYSF lead to loss of or decreased dysferlin expression, impaired membrane repair in MF, and its destruction, clinically manifesting as dysferlinopathy. Preclinical studies of cell and gene therapies aimed at restoring impaired muscle regeneration require well-characterized small animal models. Our investigation aimed to distinguish the histopathological features of a mouse strain lacking dysferlin expression (Bla/J strain). Ultrastructural changes in the sarcolemma, mitochondria and contractile apparatus were observed. It was shown that postnatal histogenesis of skeletal muscles in genetically determined dysferlin deficiency is characterized by a higher proportion of necrotic muscle fibers, compensatory hypertrophy of muscle fibers with their subsequent atrophy, and decreases in proliferative activity and the level of myogenic differentiation of myogenic progenitor cells compared to wild-type mice (C57Bl/6).


Asunto(s)
Disferlina , Músculo Esquelético , Distrofia Muscular de Cinturas , Animales , Disferlina/genética , Disferlina/metabolismo , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/patología
10.
Muscle Nerve ; 63(2): 239-249, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33125736

RESUMEN

INTRODUCTION: We conducted an open-label study to examine the effects of the flavonoid (-)-epicatechin in seven ambulatory adult patients with Becker muscular dystrophy (BMD). METHODS: Seven participants received (-)-epicatechin 50 mg twice per day for 8 weeks. Pre- and postprocedures included biceps brachii biopsy to assess muscle structure and growth-relevant endpoints by western blotting, mitochondria volume measurement, and cristae abundance by electron microscopy, graded exercise testing, and muscle strength and function tests. RESULTS: Western blotting showed significantly increased levels of enzymes modulating cellular bioenergetics (liver kinase B1 and 5'-adenosine monophosphate-activated protein kinase). Peroxisome proliferator-activated receptor gamma coactivator-1alpha, a transcriptional coactivator of genes involved in mitochondrial biogenesis and cristae-associated mitofilin levels, increased as did cristae abundance. Muscle and plasma follistatin increased significantly while myostatin decreased. Markers of skeletal muscle regeneration myogenin, myogenic regulatory factor-5, myoblast determination protein 1, myocyte enhancer factor-2, and structure-associated proteins, including dysferlin, utrophin, and intracellular creatine kinase, also increased. Exercise testing demonstrated decreased heart rate, maximal oxygen consumption per kilogram, and plasma lactate levels at defined workloads. Tissue saturation index improved in resting and postexercise states. DISCUSSION: (-)-Epicatechin, an exercise mimetic, appears to have short-term positive effects on tissue biomarkers indicative of mitochondrial biogenesis and muscle regeneration, and produced improvements in graded exercise testing parameters in patients with BMD.


Asunto(s)
Catequina/uso terapéutico , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamiento farmacológico , Adulto , Biopsia , Western Blotting , Creatina Quinasa/metabolismo , Disferlina/metabolismo , Prueba de Esfuerzo , Folistatina/metabolismo , Frecuencia Cardíaca , Humanos , Ácido Láctico/sangre , Factores de Transcripción MEF2/metabolismo , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Tamaño Mitocondrial , Proteínas Musculares/metabolismo , Fuerza Muscular , Músculo Esquelético/fisiopatología , Músculo Esquelético/ultraestructura , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Miogenina/metabolismo , Miostatina/metabolismo , Biogénesis de Organelos , Consumo de Oxígeno , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Regeneración , Utrofina/metabolismo
11.
Mol Ther ; 28(4): 1133-1153, 2020 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32087766

RESUMEN

Mutations in dysferlin are responsible for a group of progressive, recessively inherited muscular dystrophies known as dysferlinopathies. Using recombinant proteins and affinity purification methods combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS), we found that AMP-activated protein kinase (AMPK)γ1 was bound to a region of dysferlin located between the third and fourth C2 domains. Using ex vivo laser injury experiments, we demonstrated that the AMPK complex was vital for the sarcolemmal damage repair of skeletal muscle fibers. Injury-induced AMPK complex accumulation was dependent on the presence of Ca2+, and the rate of accumulation was regulated by dysferlin. Furthermore, it was found that the phosphorylation of AMPKα was essential for plasma membrane repair, and treatment with an AMPK activator rescued the membrane-repair impairment observed in immortalized human myotubes with reduced expression of dysferlin and dysferlin-null mouse fibers. Finally, it was determined that treatment with the AMPK activator metformin improved the muscle phenotype in zebrafish and mouse models of dysferlin deficiency. These findings indicate that the AMPK complex is essential for plasma membrane repair and is a potential therapeutic target for dysferlinopathy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Disferlina/química , Disferlina/metabolismo , Metformina/administración & dosificación , Músculo Esquelético/lesiones , Distrofia Muscular de Cinturas/tratamiento farmacológico , Animales , Línea Celular , Modelos Animales de Enfermedad , Disferlina/genética , Humanos , Rayos Láser/efectos adversos , Metformina/farmacología , Ratones , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Mutación , Fosforilación , Dominios Proteicos , Sarcolema/metabolismo , Pez Cebra
12.
Biochem Biophys Res Commun ; 529(3): 720-725, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32736698

RESUMEN

TMEM16E deficiency has been shown to be responsible for human limb-girdle muscular dystrophy LGMD2L. We found that endogenous TMEM16E co-localized with caveolin-3 at cytoplasmic vesicular compartments in a myotube from C2C12 cells (C2C12 myotube) without forming a molecular complex. In contrast, a myotube from murine myoblastic dysferlin-deficient GREG cells (GREG myotube) showed not only co-localization but also constitutive association of caveolin-3 and TMEM16E. GREG myotubes also displayed constitutive association of TMEM16E with DHPRα, which reside in different membrane compartments, indicating increased contact of the different vesicular membrane compartments. Τhese results suggest that a dynamic tethering of different membrane compartments might represent a distorted membrane damage repairing process in the absence of dysferlin.


Asunto(s)
Anoctaminas/metabolismo , Canales de Calcio Tipo L/metabolismo , Disferlina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Animales , Anoctaminas/análisis , Canales de Calcio Tipo L/análisis , Caveolina 3/análisis , Caveolina 3/metabolismo , Disferlina/análisis , Disferlina/genética , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/citología
13.
J Lipid Res ; 60(8): 1350-1364, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31203232

RESUMEN

Defects in the gene coding for dysferlin, a membrane-associated protein, affect many tissues, including skeletal muscles, with a resultant myopathy called dysferlinopathy. Dysferlinopathy manifests postgrowth with a progressive loss of skeletal muscle function, early intramyocellular lipid accumulation, and a striking later replacement of selective muscles by adipocytes. To better understand the changes underpinning this disease, we assessed whole-body energy homeostasis, skeletal muscle fatty acid metabolism, lipolysis in adipose tissue, and the skeletal muscle lipidome using young adult dysferlin-deficient male BLAJ mice and age-matched C57Bl/6J WT mice. BLAJ mice had increased lean mass and reduced fat mass associated with increased physical activity and increased adipose tissue lipolysis. Skeletal muscle fatty acid metabolism was remodeled in BLAJ mice, characterized by a partitioning of fatty acids toward storage rather than oxidation. Lipidomic analysis identified marked changes in almost all lipid classes examined in the skeletal muscle of BLAJ mice, including sphingolipids, phospholipids, cholesterol, and most glycerolipids but, surprisingly, not triacylglycerol. These observations indicate that an early manifestation of dysferlin deficiency is the reprogramming of skeletal muscle and adipose tissue lipid metabolism, which is likely to contribute to the progressive adverse histopathology in dysferlinopathies.


Asunto(s)
Tejido Adiposo/metabolismo , Disferlina/deficiencia , Lipólisis , Músculo Esquelético/metabolismo , Animales , Disferlina/metabolismo , Lipidómica , Ratones , Ratones Mutantes
14.
J Cell Mol Med ; 23(2): 811-818, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30450679

RESUMEN

Muscular dystrophy-dystroglycanopathy (limb-girdle), type C, 9 (MDDGC9) is the rarest type of autosomal recessive muscular dystrophies. MDDGC9 is manifested with an early onset in childhood. Patients with MDDGC9 usually identified with defective glycosylation of DAG1, hence it is known as "dystroglycanopathies". Here, we report a Chinese pedigree presented with mild MDDGC9. The proband is a 64 years old Chinese man. In this family, both the proband and proband's younger brother have been suffering from mild and late onset MDDGC9. Muscle biopsy showed that the left deltoid muscle with an advanced stage of dystrophic change. Immunohistochemistry staining of dystrophin, α-sarcoglycan, ß-sarcoglycan and dysferlin are normal. Molecular genetic analysis of the proband has been done with whole exome sequencing. A homozygous novel missense mutation (c.2326C>T; p.R776C) in the exon 3 of the DAG1 gene has been identified in the proband. Sanger sequencing revealed that this missense mutation is co-segregated well among the affected and unaffected (carrier) family members. This mutation is not detected in 200 normal healthy control individuals. This novel homozygous missense mutation (c.2326C>T) causes substitution of arginine by cystine at the position of 776 (p.R776C) which is evolutionarily highly conserved. Immunoblotting studies revealed that a significant reduction of α-dystroglycan expression in the muscle tissue. The novelty of our study is that it is a first report of DAG1 associated muscular dystrophy-dystroglycanopathy (limb-girdle), type C, 9 (MDDGC9) with mild and late age of onset. In Chinese population this is the first report of DAG1 associated MDDGC9.


Asunto(s)
Distroglicanos/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Mutación Missense , Adulto , Edad de Inicio , Secuencia de Bases , China , Disferlina/genética , Disferlina/metabolismo , Distroglicanos/deficiencia , Distrofina/genética , Distrofina/metabolismo , Exones , Femenino , Expresión Génica , Glicosilación , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/etnología , Distrofia Muscular de Cinturas/patología , Linaje , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Secuenciación del Exoma
15.
Mol Ther ; 26(9): 2231-2242, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30166241

RESUMEN

Mutations of the DYSF gene leading to reduced dysferlin protein level causes limb girdle muscular dystrophy type 2B (LGMD2B). Dysferlin facilitates sarcolemmal membrane repair in healthy myofibers, thus its deficit compromises myofiber repair and leads to chronic muscle inflammation. An experimental therapeutic approach for LGMD2B is to protect damage or improve repair of myofiber sarcolemma. Here, we compared the effects of prednisolone and vamorolone (a dissociative steroid; VBP15) on dysferlin-deficient myofiber repair. Vamorolone, but not prednisolone, stabilized dysferlin-deficient muscle cell membrane and improved repair of dysferlin-deficient mouse (B6A/J) myofibers injured by focal sarcolemmal damage, eccentric contraction-induced injury or injury due to spontaneous in vivo activity. Vamorolone decreased sarcolemmal lipid mobility, increased muscle strength, and decreased late-stage myofiber loss due to adipogenic infiltration. In contrast, the conventional glucocorticoid prednisolone failed to stabilize dysferlin deficient muscle cell membrane or improve repair of dysferlinopathic patient myoblasts and mouse myofibers. Instead, prednisolone treatment increased muscle weakness and myofiber atrophy in B6A/J mice-findings that correlate with reports of prednisolone worsening symptoms of LGMD2B patients. Our findings showing improved cellular and pre-clinical efficacy of vamorolone compared to prednisolone and better safety profile of vamorolone indicates the suitability of vamorolone for clinical trials in LGMD2B.


Asunto(s)
Disferlina/deficiencia , Distrofias Musculares/tratamiento farmacológico , Esteroides/uso terapéutico , Adolescente , Animales , Células Cultivadas , Disferlina/metabolismo , Humanos , Masculino , Ratones , Mioblastos/efectos de los fármacos , Mioblastos/metabolismo , Prednisolona/uso terapéutico , Pregnadienodioles/uso terapéutico
16.
Int J Mol Sci ; 21(1)2019 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-31861684

RESUMEN

Dysferlin is a transmembrane C-2 domain-containing protein involved in vesicle trafficking and membrane remodeling in skeletal muscle cells. However, the mechanism by which dysferlin regulates these cellular processes remains unclear. Since actin dynamics is critical for vesicle trafficking and membrane remodeling, we studied the role of dysferlin in Ca2+-induced G-actin incorporation into filaments in four different immortalized myoblast cell lines (DYSF2, DYSF3, AB320, and ER) derived from patients harboring mutations in the dysferlin gene. As compared with immortalized myoblasts obtained from a control subject, dysferlin expression and G-actin incorporation were significantly decreased in myoblasts from dysferlinopathy patients. Stable knockdown of dysferlin with specific shRNA in control myoblasts also significantly reduced G-actin incorporation. The impaired G-actin incorporation was restored by the expression of full-length dysferlin as well as dysferlin N-terminal or C-terminal regions, both of which contain three C2 domains. DYSF3 myoblasts also exhibited altered distribution of annexin A2, a dysferlin partner involved in actin remodeling. However, dysferlin N-terminal and C-terminal regions appeared to not fully restore such annexin A2 mislocation. Then, our results suggest that dysferlin regulates actin remodeling by a mechanism that does to not involve annexin A2.


Asunto(s)
Actinas/metabolismo , Disferlina/química , Distrofia Muscular de Cinturas/metabolismo , Mioblastos/citología , Citoesqueleto de Actina/metabolismo , Actinas/genética , Adolescente , Adulto , Línea Celular , Disferlina/genética , Disferlina/metabolismo , Femenino , Humanos , Masculino , Distrofia Muscular de Cinturas/genética , Mioblastos/metabolismo , Dominios Proteicos
17.
Bull Exp Biol Med ; 166(5): 593-597, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30903507

RESUMEN

Dysferlin protein plays a key role in the multimolecular complex responsible for the maintenance of sarcolemma integrity and skeletal muscle cell functioning. We studied the membrane distribution of nicotinic acetylcholine receptors and α2 isoform of Na,K-ATPase in motor endplates of m. soleus in dysferlin-deficient Bla/J mice (a dysferlinopathy model). Endplates of Bla/J mice were characterized by increased area (without changes in fragmentation degree) and reduced density of the membrane distribution of nicotinic acetylcholine receptors in comparison with the corresponding parameters in control С57Bl/6 mice. The density of the membrane distribution of α2 isoform of Na,K-ATPase was also reduced, but the level of the corresponding mRNA remained unchanged. It can be hypothesized that abnormal membrane localization of α2 isoform of Na,K-ATPase results from adaptive skeletal muscle remodeling under conditions of chronic motor dysfunction.


Asunto(s)
Membrana Celular/metabolismo , Músculo Esquelético/metabolismo , Isoformas de Proteínas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Disferlina/genética , Disferlina/metabolismo , Masculino , Ratones , Isoformas de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/genética
18.
J Lipid Res ; 59(2): 261-272, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29175948

RESUMEN

Progressive limb and girdle muscle atrophy leading to loss of ambulation is a hallmark of dysferlinopathies, which include limb-girdle muscular dystrophy type 2B and Miyoshi myopathy. However, animal models fail to fully reproduce the disease severity observed in humans, with dysferlin-null (Dysf-/-) mice exhibiting minor muscle damage and weakness without dramatic ambulatory dysfunction. As we have previously reported significant Dysf expression in blood vessels, we investigated the role of vascular function in development of muscle pathology by generating a Dysf-deficient mouse model with vascular disease. This was achieved by crossing Dysf-/- mice with ApoE-/- mice, which have high levels of nonHDL-associated cholesterol. Double-knockout Dysf-/-ApoE-/- mice exhibited severe ambulatory dysfunction by 11 months of age. In limb-girdle muscles, histology confirmed dramatic muscle wasting, fibrofatty replacement, and myofiber damage in Dysf-/-ApoE-/- mice without affecting the ratio of centrally nucleated myofibers. Although there were no major changes in ex vivo diaphragm and soleus muscle function, histological analyses revealed these muscles to be untouched by damage and remodelling. In all, these data suggest that cholesterol may be deleterious to dysferlinopathic muscle and lead to ambulatory dysfunction. Moreover, differences in plasma lipid handling between mice and humans could be a key factor affecting dysferlinopathy severity.


Asunto(s)
Colesterol/metabolismo , Modelos Animales de Enfermedad , Disferlina/metabolismo , Atrofia Muscular/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Animales , Disferlina/deficiencia , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
19.
J Biol Chem ; 292(45): 18577-18591, 2017 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-28904177

RESUMEN

Dysferlin is a large transmembrane protein that plays a key role in cell membrane repair and underlies a recessive form of inherited muscular dystrophy. Dysferlinopathy is characterized by absence or marked reduction of dysferlin protein with 43% of reported pathogenic variants being missense variants that span the length of the dysferlin protein. The unique structure of dysferlin, with seven tandem C2 domains separated by linkers, suggests dysferlin may dynamically associate with phospholipid membranes in response to Ca2+ signaling. However, the overall conformation of the dysferlin protein is uncharacterized. To dissect the structural architecture of dysferlin, we have applied the method of limited proteolysis, which allows nonspecific digestion of unfolded peptides by trypsin. Using five antibodies spanning the dysferlin protein, we identified a highly reproducible jigsaw map of dysferlin fragments protected from digestion. Our data infer a modular architecture of four tertiary domains: 1) C2A, which is readily removed as a solo domain; 2) midregion C2B-C2C-Fer-DysF, commonly excised as an intact module, with subdigestion to different fragments suggesting several dynamic folding options; 3) C-terminal four-C2 domain module; and 4) calpain-cleaved mini-dysferlinC72, which is particularly resistant to proteolysis. Importantly, we reveal a patient missense variant, L344P, that largely escapes proteasomal surveillance and shows subtle but clear changes in tertiary conformation. Accompanying evidence from immunohistochemistry and flow cytometry using antibodies with conformationally sensitive epitopes supports proteolysis data. Collectively, we provide insight into the structural topology of dysferlin and show how a single missense mutation within dysferlin can exert local changes in tertiary conformation.


Asunto(s)
Disferlina/genética , Músculo Esquelético/metabolismo , Distrofias Musculares/genética , Mutación Missense , Complejo de la Endopetidasa Proteasomal/metabolismo , Sustitución de Aminoácidos , Biopsia , Dominios C2 , Calpaína/genética , Calpaína/metabolismo , Células Cultivadas , Disferlina/química , Disferlina/metabolismo , Células HEK293 , Humanos , Peso Molecular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Conformación Proteica , Pliegue de Proteína , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Proteolisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
20.
Mol Ther ; 25(9): 2150-2162, 2017 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-28629822

RESUMEN

Dysferlinopathy is an autosomal recessive muscular dystrophy characterized by the progressive loss of motility that is caused by mutations throughout the DYSF gene. There are currently no approved therapies that ameliorate or reverse dysferlinopathy. Gene delivery using adeno-associated vectors (AAVs) is a leading therapeutic strategy for genetic diseases; however, the large size of dysferlin cDNA (6.2 kB) precludes packaging into a single AAV capsid. Therefore, using 3D structural modeling and hypothesizing dysferlin C2 domain redundancy, a 30% smaller, dysferlin-like molecule amenable to single AAV vector packaging was engineered (termed Nano-Dysferlin). The intracellular distribution of Nano-Dysferlin was similar to wild-type dysferlin and neither demonstrated toxicity when overexpressed in dysferlin-deficient patient myoblasts. Intramuscular injection of AAV-Nano-Dysferlin in young dysferlin-deficient mice significantly improved muscle integrity and decreased muscle turnover 3 weeks after treatment, as determined by Evans blue dye uptake and central nucleated fibers, respectively. Systemically administered AAV-Nano-Dysferlin to young adult dysferlin-deficient mice restored motor function and improved muscle integrity nearly 8 months after a single injection. These preclinical data are the first report of a smaller dysferlin variant tailored for AAV single particle delivery that restores motor function and, therefore, represents an attractive candidate for the treatment of dysferlinopathy.


Asunto(s)
Diseño de Fármacos , Disferlina/química , Disferlina/genética , Distrofia Muscular de Cinturas/genética , Animales , Dependovirus/genética , Modelos Animales de Enfermedad , Disferlina/metabolismo , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Inyecciones Intramusculares , Ratones , Actividad Motora/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/terapia , Regiones Promotoras Genéticas , Dominios Proteicos , Relación Estructura-Actividad , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA