Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(17)2022 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-36077200

RESUMEN

Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene and dilated cardiomyopathy (DCM) is a major cause of morbidity and mortality in DMD patients. We tested the hypothesis that DCM is caused by metabolic impairments by employing induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) generated from four DMD patients; an adult male, an adult female, a 7-year-old (7y) male and a 13-year-old (13y) male, all compared to two healthy volunteers. To test the hypothesis, we measured the bioenergetics, metabolomics, electrophysiology, mitochondrial morphology and mitochondrial activity of CMs, using respirometry, LC-MS, patch clamp, electron microscopy (EM) and confocal microscopy methods. We found that: (1) adult DMD CMs exhibited impaired energy metabolism and abnormal mitochondrial structure and function. (2) The 7y CMs demonstrated arrhythmia-free spontaneous firing along with "healthy-like" metabolic status, normal mitochondrial morphology and activity. In contrast, the 13y CMs were mildly arrhythmogenic and showed adult DMD-like bioenergetics deficiencies. (3) In DMD adult CMs, mitochondrial activities were attenuated by 45-48%, whereas the 7y CM activity was similar to that of healthy CMs. (4) In DMD CMs, but not in 7y CMs, there was a 75% decrease in the mitochondrial ATP production rate compared to healthy iPSC-CMs. In summary, DMD iPSC-CMs exhibit bioenergetic and metabolic impairments that are associated with rhythm disturbances corresponding to the patient's phenotype, thereby constituting novel targets for alleviating cardiomyopathy in DMD patients.


Asunto(s)
Cardiomiopatía Dilatada , Células Madre Pluripotentes Inducidas , Distrofia Muscular de Duchenne , Cardiomiopatía Dilatada/metabolismo , Diferenciación Celular , Distrofina/genética , Metabolismo Energético , Femenino , Humanos , Masculino , Distrofia Muscular de Duchenne/genética , Miocitos Cardíacos/metabolismo
2.
J Biol Chem ; 294(41): 14991-15002, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31416832

RESUMEN

Iron is a micronutrient fundamental for life. Iron homeostasis in mammals requires sustained postnatal intestinal iron absorption that maintains intracellular iron concentrations for central and systemic metabolism as well as for erythropoiesis and oxygen transport. More than 1 billion people worldwide suffer from iron deficiency anemia (IDA), a state of systemic iron insufficiency that limits the production of red blood cells and leads to tissue hypoxia and intracellular iron stress. Despite this tremendous public health concern, very few genetic models of IDA are available to study its progression. Here we developed and characterized a novel genetic mouse model of IDA. We found that tamoxifen-inducible deletion of the mammalian iron exporter ferroportin exclusively in intestinal epithelial cells leads to loss of intestinal iron absorption. Ferroportin ablation yielded a robust phenotype of progressive IDA that develops in as little as 3 months following disruption of intestinal iron absorption. We noted that, at end-stage IDA, tissue-specific transcriptional stress responses occur in which the heart shows little to no hypoxic and iron stress compared with other peripheral organs. However, morphometric and echocardiographic analysis revealed massive cardiac hypertrophy and chamber dilation, albeit with increased cardiac output at very low basal heart rates. We propose that our intestine-specific ferroportin knockout mouse model of end-stage IDA could be used in future studies to investigate IDA progression and cell-specific responses to hypoxic and iron stress.


Asunto(s)
Anemia Ferropénica/genética , Anemia Ferropénica/patología , Remodelación Atrial/genética , Estrés Fisiológico/genética , Transcripción Genética , Animales , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Hipoxia de la Célula/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Intestinos/patología , Ratones , Miocardio/patología , Especificidad de Órganos
3.
Am J Physiol Heart Circ Physiol ; 319(3): H582-H603, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32762558

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked disease caused by null mutations in dystrophin and characterized by muscle degeneration. Cardiomyopathy is common and often prevalent at similar frequency in female DMD carriers irrespective of whether they manifest skeletal muscle disease. Impaired muscle nitric oxide (NO) production in DMD disrupts muscle blood flow regulation and exaggerates postexercise fatigue. We show that circulating levels of endogenous methylated arginines including asymmetric dimethylarginine (ADMA), which act as NO synthase inhibitors, are elevated by acute necrotic muscle damage and in chronically necrotic dystrophin-deficient mice. We therefore hypothesized that excessive ADMA impairs muscle NO production and diminishes exercise tolerance in DMD. We used transgenic expression of dimethylarginine dimethylaminohydrolase 1 (DDAH), which degrades methylated arginines, to investigate their contribution to exercise-induced fatigue in DMD. Although infusion of exogenous ADMA was sufficient to impair exercise performance in wild-type mice, transgenic DDAH expression did not rescue exercise-induced fatigue in dystrophin-deficient male mdx mice. Surprisingly, DDAH transgene expression did attenuate exercise-induced fatigue in dystrophin-heterozygous female mdx carrier mice. Improved exercise tolerance was associated with reduced heart weight and improved cardiac ß-adrenergic responsiveness in DDAH-transgenic mdx carriers. We conclude that DDAH overexpression increases exercise tolerance in female DMD carriers, possibly by limiting cardiac pathology and preserving the heart's responses to changes in physiological demand. Methylated arginine metabolism may be a new target to improve exercise tolerance and cardiac function in DMD carriers or act as an adjuvant to promote NO signaling alongside therapies that partially restore dystrophin expression in patients with DMD.NEW & NOTEWORTHY Duchenne muscular dystrophy (DMD) carriers are at risk for cardiomyopathy. The nitric oxide synthase inhibitor asymmetric dimethylarginine (ADMA) is released from damaged muscle in DMD and impairs exercise performance. Transgenic expression of dimethylarginine dimethylaminohydrolase to degrade ADMA prevents cardiac hypertrophy, improves cardiac function, and improves exercise tolerance in DMD carrier mice. These findings highlight the relevance of ADMA to muscular dystrophy and have important implications for therapies targeting nitric oxide in patients with DMD and DMD carriers.


Asunto(s)
Arginina/análogos & derivados , Cardiomiopatías/metabolismo , Circulación Coronaria , Tolerancia al Ejercicio , Heterocigoto , Distrofia Muscular de Duchenne/metabolismo , Miocardio/metabolismo , Músculo Cuádriceps/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Arginina/metabolismo , Cardiomiopatías/genética , Cardiomiopatías/fisiopatología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Miocardio/patología , Necrosis , Músculo Cuádriceps/patología , Músculo Cuádriceps/fisiopatología , Función Ventricular Izquierda
4.
J Cell Mol Med ; 23(3): 2125-2135, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30618214

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked progressive muscle degenerative disease, caused by mutations in the dystrophin gene and resulting in death because of respiratory or cardiac failure. To investigate the cardiac cellular manifestation of DMD, we generated induced pluripotent stem cells (iPSCs) and iPSC-derived cardiomyocytes (iPSC-CMs) from two DMD patients: a male and female manifesting heterozygous carrier. Dystrophin mRNA and protein expression were analysed by qRT-PCR, RNAseq, Western blot and immunofluorescence staining. For comprehensive electrophysiological analysis, current and voltage clamp were used to record transmembrane action potentials and ion currents, respectively. Microelectrode array was used to record extracellular electrograms. X-inactive specific transcript (XIST) and dystrophin expression analyses revealed that female iPSCs underwent X chromosome reactivation (XCR) or erosion of X chromosome inactivation, which was maintained in female iPSC-CMs displaying mixed X chromosome expression of wild type (WT) and mutated alleles. Both DMD female and male iPSC-CMs presented low spontaneous firing rate, arrhythmias and prolonged action potential duration. DMD female iPSC-CMs displayed increased beat rate variability (BRV). DMD male iPSC-CMs manifested decreased If density, and DMD female and male iPSC-CMs showed increased ICa,L density. Our findings demonstrate cellular mechanisms underlying electrophysiological abnormalities and cardiac arrhythmias in DMD.


Asunto(s)
Heterocigoto , Células Madre Pluripotentes Inducidas/fisiología , Distrofia Muscular de Duchenne/fisiopatología , Miocitos Cardíacos/fisiología , Potenciales de Acción/genética , Adulto , Diferenciación Celular/genética , Distrofina/genética , Distrofina/metabolismo , Fenómenos Electrofisiológicos , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/ultraestructura , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura
5.
Proc Natl Acad Sci U S A ; 112(44): 13663-8, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26483453

RESUMEN

Patients deficient in dystrophin, a protein that links the cytoskeleton to the extracellular matrix via the dystrophin-glycoprotein complex (DGC), exhibit muscular dystrophy, cardiomyopathy, and impaired muscle nitric oxide (NO) production. We used live-cell NO imaging and in vitro cyclic stretch of isolated adult mouse cardiomyocytes as a model system to investigate if and how the DGC directly regulates the mechanical activation of muscle NO signaling. Acute activation of NO synthesis by mechanical stretch was impaired in dystrophin-deficient mdx cardiomyocytes, accompanied by loss of stretch-induced neuronal NO synthase (nNOS) S1412 phosphorylation. Intriguingly, stretch induced the acute activation of AMP-activated protein kinase (AMPK) in normal cardiomyocytes but not in mdx cardiomyocytes, and specific inhibition of AMPK was sufficient to attenuate mechanoactivation of NO production. Therefore, we tested whether direct pharmacologic activation of AMPK could bypass defective mechanical signaling to restore nNOS activity in dystrophin-deficient cardiomyocytes. Indeed, activation of AMPK with 5-aminoimidazole-4-carboxamide riboside or salicylate increased nNOS S1412 phosphorylation and was sufficient to enhance NO production in mdx cardiomyocytes. We conclude that the DGC promotes the mechanical activation of cardiac nNOS by acting as a mechanosensor to regulate AMPK activity, and that pharmacologic AMPK activation may be a suitable therapeutic strategy for restoring nNOS activity in dystrophin-deficient hearts and muscle.


Asunto(s)
Adenilato Quinasa/metabolismo , Distrofina/metabolismo , Glicoproteínas/metabolismo , Miocitos Cardíacos/metabolismo , Óxido Nítrico/biosíntesis , Transducción de Señal , Animales , Activación Enzimática , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Fosforilación
6.
Hum Mol Genet ; 23(7): 1677-86, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24203699

RESUMEN

Mutations in the dysferlin gene resulting in dysferlin-deficiency lead to limb-girdle muscular dystrophy 2B and Myoshi myopathy in humans. Dysferlin has been proposed as a critical regulator of vesicle-mediated membrane resealing in muscle fibers, and localizes to muscle fiber wounds following sarcolemma damage. Studies in fibroblasts and urchin eggs suggest that trafficking and fusion of intracellular vesicles with the plasma membrane during resealing requires the intracellular cytoskeleton. However, the contribution of dysferlin-containing vesicles to resealing in muscle and the role of the cytoskeleton in regulating dysferlin-containing vesicle biology is unclear. Here, we use live-cell imaging to examine the behavior of dysferlin-containing vesicles following cellular wounding in muscle cells and examine the role of microtubules and kinesin in dysferlin-containing vesicle behavior following wounding. Our data indicate that dysferlin-containing vesicles move along microtubules via the kinesin motor KIF5B in muscle cells. Membrane wounding induces dysferlin-containing vesicle-vesicle fusion and the formation of extremely large cytoplasmic vesicles, and this response depends on both microtubules and functional KIF5B. In non-muscle cell types, lysosomes are critical mediators of membrane resealing, and our data indicate that dysferlin-containing vesicles are capable of fusing with lysosomes following wounding which may contribute to formation of large wound sealing vesicles in muscle cells. Overall, our data provide mechanistic evidence that microtubule-based transport of dysferlin-containing vesicles may be critical for resealing, and highlight a critical role for dysferlin-containing vesicle-vesicle and vesicle-organelle fusion in response to wounding in muscle cells.


Asunto(s)
Membrana Celular/patología , Vesículas Citoplasmáticas/patología , Cinesinas/metabolismo , Fusión de Membrana/fisiología , Proteínas de la Membrana/genética , Microtúbulos/metabolismo , Proteínas Musculares/genética , Animales , Línea Celular , Proteínas Fluorescentes Verdes , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Cinesinas/genética , Lisosomas/metabolismo , Células Musculares/citología , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Cinturas/genética , Miosina Tipo II/antagonistas & inhibidores , Nocodazol/farmacología , Ratas , Moduladores de Tubulina/farmacología
7.
Am J Physiol Heart Circ Physiol ; 310(9): H1140-50, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26968544

RESUMEN

Delta-sarcoglycan is a component of the sarcoglycan subcomplex within the dystrophin-glycoprotein complex located at the plasma membrane of muscle cells. While recessive mutations in δ-sarcoglycan cause limb girdle muscular dystrophy 2F, dominant mutations in δ-sarcoglycan have been linked to inherited dilated cardiomyopathy (DCM). The purpose of this study was to investigate functional cellular defects present in adult cardiac myocytes expressing mutant δ-sarcoglycans harboring the dominant inherited DCM mutations R71T or R97Q. This study demonstrates that DCM mutant δ-sarcoglycans can be stably expressed in adult rat cardiac myocytes and traffic similarly to wild-type δ-sarcoglycan to the plasma membrane, without perturbing assembly of the dystrophin-glycoprotein complex. However, expression of DCM mutant δ-sarcoglycan in adult rat cardiac myocytes is sufficient to alter cardiac myocyte plasma membrane stability in the presence of mechanical strain. Upon cyclical cell stretching, cardiac myocytes expressing mutant δ-sarcoglycan R97Q or R71T have increased cell-impermeant dye uptake and undergo contractures at greater frequencies than myocytes expressing normal δ-sarcoglycan. Additionally, the R71T mutation creates an ectopic N-linked glycosylation site that results in aberrant glycosylation of the extracellular domain of δ-sarcoglycan. Therefore, appropriate glycosylation of δ-sarcoglycan may also be necessary for proper δ-sarcoglycan function and overall dystrophin-glycoprotein complex function. These studies demonstrate that DCM mutations in δ-sarcoglycan can exert a dominant negative effect on dystrophin-glycoprotein complex function leading to myocardial mechanical instability that may underlie the pathogenesis of δ-sarcoglycan-associated DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Genes Dominantes , Mecanotransducción Celular , Mutación , Miocitos Cardíacos/metabolismo , Sarcoglicanos/genética , Animales , Cardiomiopatía Dilatada/metabolismo , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular , Células Cultivadas , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Contracción Miocárdica , Fenotipo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Ratas Sprague-Dawley , Sarcoglicanos/metabolismo , Estrés Mecánico , Transfección
8.
J Virol ; 89(1): 468-79, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25320326

RESUMEN

UNLABELLED: Adenoviruses are frequent causes of pediatric myocarditis. Little is known about the pathogenesis of adenovirus myocarditis, and the species specificity of human adenoviruses has limited the development of animal models, which is a significant barrier to strategies for prevention or treatment. We have developed a mouse model of myocarditis following mouse adenovirus 1 (MAV-1) infection to study the pathogenic mechanisms of this important cause of pediatric myocarditis. Following intranasal infection of neonatal C57BL/6 mice, we detected viral replication and induction of interferon gamma (IFN-γ) in the hearts of infected mice. MAV-1 caused myocyte necrosis and induced substantial cellular inflammation that was composed predominantly of CD3(+) T lymphocytes. Depletion of IFN-γ during acute infection reduced cardiac inflammation in MAV-1-infected mice without affecting viral replication. We observed decreased contractility during acute infection of neonatal mice, and persistent viral infection in the heart was associated with cardiac remodeling and hypertrophy in adulthood. IFN-γ is a proinflammatory mediator during adenovirus-induced myocarditis, and persistent adenovirus infection may contribute to ongoing cardiac dysfunction. IMPORTANCE: Studying the pathogenesis of myocarditis caused by different viruses is essential in order to characterize both virus-specific and generalized factors that contribute to disease. Very little is known about the pathogenesis of adenovirus myocarditis, which is a significant impediment to the development of treatment or prevention strategies. We used MAV-1 to establish a mouse model of human adenovirus myocarditis, providing the means to study host and pathogen factors contributing to adenovirus-induced cardiac disease during acute and persistent infection. The MAV-1 model will enable fundamental studies of viral myocarditis, including IFN-γ modulation as a therapeutic strategy.


Asunto(s)
Interferón gamma/metabolismo , Mastadenovirus/inmunología , Miocarditis/inmunología , Miocarditis/patología , Miocardio/patología , Animales , Complejo CD3/análisis , Femenino , Masculino , Ratones Endogámicos C57BL , Monocitos/inmunología , Linfocitos T/química , Linfocitos T/inmunología , Replicación Viral
9.
Hum Mol Genet ; 22(4): 757-68, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23222475

RESUMEN

Mutations in several glycosyltransferases underlie a group of muscular dystrophies known as glycosylation-deficient muscular dystrophy. A common feature of these diseases is loss of glycosylation and consequent dystroglycan function that is correlated with severe pathology in muscle, brain and other tissues. Although glycosylation of dystroglycan is essential for function in skeletal muscle, whether glycosylation-dependent function of dystroglycan is sufficient to explain all complex pathological features associated with these diseases is less clear. Dystroglycan glycosylation is defective in LARGE(myd) (myd) mice as a result of a mutation in like-acetylglucosaminyltransferase (LARGE), a glycosyltransferase known to cause muscle disease in humans. We generated animals with restored dystroglycan function exclusively in skeletal muscle by crossing myd animals to a recently created transgenic line that expresses LARGE selectively in differentiated muscle. Transgenic myd mice were indistinguishable from wild-type littermates and demonstrated an amelioration of muscle disease as evidenced by an absence of muscle pathology, restored contractile function and a reduction in serum creatine kinase activity. Moreover, although deficits in nerve conduction and neuromuscular transmission were observed in myd animals, these deficits were fully rescued by muscle-specific expression of LARGE, which resulted in restored structure of the neuromuscular junction (NMJ). These data demonstrate that, in addition to muscle degeneration and dystrophy, impaired neuromuscular transmission contributes to muscle weakness in dystrophic myd mice and that the noted defects are primarily due to the effects of LARGE and glycosylated dystroglycan in stabilizing the endplate of the NMJ.


Asunto(s)
Músculo Esquelético/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , Unión Neuromuscular/fisiopatología , Animales , Distroglicanos/metabolismo , Glicosilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Destreza Motora , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/fisiopatología , Miocardio/metabolismo , Unión Neuromuscular/metabolismo , Unión Neuromuscular/patología , Especificidad de Órganos , Procesamiento Proteico-Postraduccional , Transmisión Sináptica
10.
Am J Pathol ; 184(6): 1819-30, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24726497

RESUMEN

Abnormalities in phosphoinositide metabolism are an emerging theme in human neurodegenerative disease. Myotubular myopathy is a prototypical disorder of phosphoinositide dysregulation that is characterized by profound muscle pathology and weakness and that is caused by mutations in MTM1, which encodes a phosphatase that targets 3-position phosphoinositides, including phosphatidylinositol 3-phosphate. Although the association between MTM1 and muscle disease has become increasingly clarified, the normal role(s) of phosphatidylinositol 3-phosphate metabolism in muscle development and homeostasis remain poorly understood. To begin to address the function of phosphatidylinositol 3-phosphate in skeletal muscle, we focused on the primary kinase responsible for its production, and created a muscle-specific conditional knockout of the class III phosphatidylinositol 3-kinase, Pik3c3. Muscle-specific deletion of Pik3c3 did not disturb embryogenesis or early postnatal development, but resulted in progressive disease characterized by reduced activity and death by 2 months of age. Histopathological analysis demonstrated changes consistent with a murine muscular dystrophy. Examination for cellular mechanism(s) responsible for the dystrophic phenotype revealed significant alterations in the autophagolysosomal pathway with mislocation of known dystrophy proteins to the lysosomal compartment. In all, we present the first analysis of Pik3c3 in skeletal muscle, and report a novel association between deletion of Pik3c3 and muscular dystrophy.


Asunto(s)
Autofagia , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/enzimología , Distrofia Muscular Animal/enzimología , Fosfatos de Fosfatidilinositol/metabolismo , Animales , Fosfatidilinositol 3-Quinasas Clase III/genética , Lisosomas/metabolismo , Lisosomas/patología , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patología , Fosfatos de Fosfatidilinositol/genética
11.
FASEB J ; 28(8): 3660-70, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24784578

RESUMEN

Deficits in membrane repair may contribute to disease progression in dysferlin-deficient muscular dystrophy. Dysferlin, a type-II transmembrane phospholipid-binding protein, is hypothesized to regulate fusion of repair vesicles with the sarcolemma to facilitate membrane repair, but the dysferlin-containing compartments involved in membrane repair and the mechanism by which these compartments contribute to resealing are unclear. A dysferlin-pHluorin [dysf-pH-sensitive green fluorescent protein (pHGFP)] muscle-specific transgenic mouse was developed to examine the dynamic behavior and subcellular localization of dysferlin during membrane repair in adult skeletal muscle fibers. Live-cell confocal microscopy of uninjured adult dysf-pHGFP muscle fibers revealed that dysferlin is highly enriched in the sarcolemma and transverse tubules. Laser-wounding induced rapid recruitment of ∼30 µm of local dysferlin-containing sarcolemma, leading to formation of stable dysferlin accumulations surrounding lesions, endocytosis of dysferlin, and formation of large cytoplasmic vesicles from distal regions of the fiber. Disruption of the actin cytoskeleton decreased recruitment of sarcolemma-derived dysferlin to lesions in dysf-pHGFP fibers without affecting endocytosis and impaired membrane resealing in wild-type fibers, similar to findings in dysferlin deficiency (a 2-fold increase in FM1-43 uptake). Our data support a new mechanism whereby recruitment of sarcolemma-derived dysferlin creates an active zone of high lipid-binding activity at wounds to interact with repair vesicles and facilitate membrane resealing in skeletal muscle.


Asunto(s)
Citoesqueleto/fisiología , Proteínas de la Membrana/fisiología , Fibras Musculares Esqueléticas/metabolismo , Miocitos Cardíacos/metabolismo , Sarcolema/metabolismo , Cicatrización de Heridas/fisiología , Actinas/fisiología , Animales , Citocalasina D/farmacología , Citoesqueleto/efectos de los fármacos , Disferlina , Endocitosis , Genes Reporteros , Genes Sintéticos , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fibras Musculares Esqueléticas/ultraestructura , Músculo Esquelético/lesiones , Miocitos Cardíacos/ultraestructura , Proteínas Recombinantes de Fusión/metabolismo , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestructura , Cicatrización de Heridas/efectos de los fármacos
12.
Brain ; 136(Pt 1): 269-81, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23288328

RESUMEN

Dystroglycanopathies are a clinically and genetically diverse group of recessively inherited conditions ranging from the most severe of the congenital muscular dystrophies, Walker-Warburg syndrome, to mild forms of adult-onset limb-girdle muscular dystrophy. Their hallmark is a reduction in the functional glycosylation of α-dystroglycan, which can be detected in muscle biopsies. An important part of this glycosylation is a unique O-mannosylation, essential for the interaction of α-dystroglycan with extracellular matrix proteins such as laminin-α2. Mutations in eight genes coding for proteins in the glycosylation pathway are responsible for ∼50% of dystroglycanopathy cases. Despite multiple efforts using traditional positional cloning, the causative genes for unsolved dystroglycanopathy cases have escaped discovery for several years. In a recent collaborative study, we discovered that loss-of-function recessive mutations in a novel gene, called isoprenoid synthase domain containing (ISPD), are a relatively common cause of Walker-Warburg syndrome. In this article, we report the involvement of the ISPD gene in milder dystroglycanopathy phenotypes ranging from congenital muscular dystrophy to limb-girdle muscular dystrophy and identified allelic ISPD variants in nine cases belonging to seven families. In two ambulant cases, there was evidence of structural brain involvement, whereas in seven, the clinical manifestation was restricted to a dystrophic skeletal muscle phenotype. Although the function of ISPD in mammals is not yet known, mutations in this gene clearly lead to a reduction in the functional glycosylation of α-dystroglycan, which not only causes the severe Walker-Warburg syndrome but is also a common cause of the milder forms of dystroglycanopathy.


Asunto(s)
Distrofias Musculares/congénito , Distrofias Musculares/genética , Mutación , Nucleotidiltransferasas/genética , Adolescente , Niño , Preescolar , Distroglicanos/genética , Distroglicanos/metabolismo , Femenino , Glicosilación , Humanos , Imagen por Resonancia Magnética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/patología , Adulto Joven
13.
PLoS Genet ; 7(5): e1002062, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21625620

RESUMEN

Muscle-eye-brain disease (MEB) and Walker Warburg Syndrome (WWS) belong to a spectrum of autosomal recessive diseases characterized by ocular dysgenesis, neuronal migration defects, and congenital muscular dystrophy. Until now, the pathophysiology of MEB/WWS has been attributed to alteration in dystroglycan post-translational modification. Here, we provide evidence that mutations in a gene coding for a major basement membrane protein, collagen IV alpha 1 (COL4A1), are a novel cause of MEB/WWS. Using a combination of histological, molecular, and biochemical approaches, we show that heterozygous Col4a1 mutant mice have ocular dysgenesis, neuronal localization defects, and myopathy characteristic of MEB/WWS. Importantly, we identified putative heterozygous mutations in COL4A1 in two MEB/WWS patients. Both mutations occur within conserved amino acids of the triple-helix-forming domain of the protein, and at least one mutation interferes with secretion of the mutant proteins, resulting instead in intracellular accumulation. Expression and posttranslational modification of dystroglycan is unaltered in Col4a1 mutant mice indicating that COL4A1 mutations represent a distinct pathogenic mechanism underlying MEB/WWS. These findings implicate a novel gene and a novel mechanism in the etiology of MEB/WWS and expand the clinical spectrum of COL4A1-associated disorders.


Asunto(s)
Colágeno Tipo IV/genética , Ojo/patología , Enfermedades Musculares/genética , Mutación , Neuronas/patología , Síndrome de Walker-Warburg/genética , Animales , Apoptosis , Secuencia de Bases , Colágeno Tipo IV/metabolismo , Humanos , Ratones , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Alineación de Secuencia , Síndrome de Walker-Warburg/metabolismo , Síndrome de Walker-Warburg/patología
14.
bioRxiv ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38853840

RESUMEN

Cardiomyocytes require the HSP70 chaperone BiP to maintain proteostasis in the endoplasmic reticulum (ER) following cardiac stress. The adenylyl transferase (AMPylase) FICD is increasingly recognized to regulate BiP activity through the post-translational addition of an adenosine monophosphate moiety to BiP surface residues. However, the physiological impact of FICD-mediated BiP regulation in the context of cardiovascular health is unknown. Here, we find that FICD deficiency prevents pressure overload-associated heart failure, hypertrophy, and fibrosis, and that FICD knockout mice maintain normal cardiac function after cardiac pressure overload. At a cellular level, we observe that FICD-mediated BiP AMPylation blunts the induction of the unfolded protein response (UPR ER ) and impairs BiP interaction with FAM134B, an ER-phagy receptor, thus limiting ER-phagy induction under stress. In contrast, FICD loss significantly increases BiP-dependent UPR ER induction and ER-phagy in stressed cardiomyocytes. We also uncover cell type-specific consequences of FICD activity in response to ER stress, positioning FICD as a critical proteostasis regulator in cardiac tissue. Our results highlight a novel regulatory paradigm controlling stress resilience in cardiomyocytes and offer a rationale to consider FICD as a therapeutic target to treat cardiac hypertrophy.

15.
JCI Insight ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38954467

RESUMEN

Pulmonary disorders impact 40% to 80% of individuals with obesity. Respiratory muscle dysfunction is linked to these conditions; however, its pathophysiology remains largely undefined. Mice subjected to diet-induced obesity (DIO) develop diaphragmatic weakness. Increased intra-diaphragmatic adiposity and extracellular matrix (ECM) content correlate with reductions in contractile force. Thrombospondin-1 (THBS1) is an obesity-associated matricellular protein linked with muscular damage in genetic myopathies. THBS1 induces proliferation of fibro-adipogenic progenitors (FAPs) - mesenchymal cells that differentiate into adipocytes and fibroblasts. We hypothesized that THBS1 drives FAP-mediated diaphragm remodeling and contractile dysfunction in DIO. We tested this by comparing the effects of dietary challenge on diaphragms of wild-type (WT) and Thbs1 knockout (Thbs1-/-) mice. Bulk and single-cell transcriptomics demonstrated DIO-induced stromal expansion in WT diaphragms. Diaphragm FAPs displayed upregulation of ECM and TGF ß-related expression signatures and augmentation of a Thy1-expressing sub-population previously linked to type 2 diabetes. Despite similar weight gain, Thbs1-/- mice were protected from these transcriptomic changes and from obesity-induced increases in diaphragm adiposity and ECM deposition. Unlike WT controls, Thbs1-/- diaphragms maintained normal contractile force and motion after DIO challenge. These findings establish THBS1 as a necessary mediator of diaphragm stromal remodeling and contractile dysfunction in overnutrition and a potential therapeutic target in obesity-associated respiratory dysfunction.

16.
Hum Mol Genet ; 20(17): 3346-55, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21628317

RESUMEN

The abnormal glycosylation and loss of extracellular matrix receptor function of the protein dystroglycan (DG) lead to the development of muscular dystrophy and cardiomyopathy. Dystroglycan is an important receptor for extracellular matrix proteins, such as laminin, in the basement membrane surrounding muscle. Large(myd) mice have a null mutation in a gene encoding the glycosyltransferase LARGE that results in abnormal glycosylation of α-DG and phenotypes similar to those in human α-DG glycosylation-deficient muscular dystrophy. Here, we show that Large(myd) hearts with the loss of DG extracellular matrix receptor function display a cardiomyopathy characterized by myocyte damage in patches of cells positive for membrane impermeant dyes. To examine the cellular mechanisms, we show that isolated adult cardiac myocytes from Large(myd) mice retain normal laminin-dependent cell adhesion, cell surface laminin deposition and basement membrane assembly. However, although isolated adult cardiac myocytes with the loss of α-DG glycosylation adhere normally to laminin substrates both passively and in the presence of mechanical activity, Large(myd) myocytes rapidly take up membrane impermeant dye following cyclical cell stretching. Therefore, while other cell surface laminin receptors are likely responsible for myocardial cell adhesion to the basement membrane, DG has a unique function of stabilizing the cardiac myocyte plasma membrane during repetitive mechanical activity by tightly binding the transmembrane dystrophin-glycoprotein complex to the extracellular matrix. This function of DG to stabilize the myocyte membrane during normal physiologic cell length changes is likely critical for the prevention of the myocardial damage and subsequent remodeling observed in α-DG glycosylation-deficient muscular dystrophies.


Asunto(s)
Distroglicanos/metabolismo , Miocitos Cardíacos/metabolismo , Receptores de Superficie Celular/metabolismo , Sarcolema/metabolismo , Animales , Western Blotting , Adhesión Celular/fisiología , Células Cultivadas , Ratones , Miocitos Cardíacos/citología
17.
bioRxiv ; 2023 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-37645822

RESUMEN

Pulmonary disorders impact 40-80% of individuals with obesity. Respiratory muscle dysfunction is linked to these conditions; however, its pathophysiology remains largely undefined. Mice subjected to diet-induced obesity (DIO) develop diaphragmatic weakness. Increased intra-diaphragmatic adiposity and extracellular matrix (ECM) content correlate with reductions in contractile force. Thrombospondin-1 (THBS1) is an obesity-associated matricellular protein linked with muscular damage in genetic myopathies. THBS1 induces proliferation of fibro-adipogenic progenitors (FAPs)-mesenchymal cells that differentiate into adipocytes and fibroblasts. We hypothesized that THBS1 drives FAP-mediated diaphragm remodeling and contractile dysfunction in DIO. We tested this by comparing effects of dietary challenge on diaphragms of wild-type (WT) and Thbs1 knockout ( Thbs1 -/- ) mice. Bulk and single-cell transcriptomics demonstrated DIO-induced stromal expansion in WT diaphragms. Diaphragm FAPs displayed upregulation of ECM and TGFß-related expression signatures, and augmentation of a Thy1 -expressing sub-population previously linked to type 2 diabetes. Despite similar weight gain, Thbs1 -/- mice were protected from these transcriptomic changes, and from obesity-induced increases in diaphragm adiposity and ECM deposition. Unlike WT controls, Thbs1 -/- diaphragms maintained normal contractile force and motion after DIO challenge. These findings establish THBS1 as a necessary mediator of diaphragm stromal remodeling and contractile dysfunction in overnutrition, and potential therapeutic target in obesity-associated respiratory dysfunction.

18.
Proc Natl Acad Sci U S A ; 106(31): 12573-9, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19633189

RESUMEN

Skeletal muscle basal lamina is linked to the sarcolemma through transmembrane receptors, including integrins and dystroglycan. The function of dystroglycan relies critically on posttranslational glycosylation, a common target shared by a genetically heterogeneous group of muscular dystrophies characterized by alpha-dystroglycan hypoglycosylation. Here we show that both dystroglycan and integrin alpha7 contribute to force-production of muscles, but that only disruption of dystroglycan causes detachment of the basal lamina from the sarcolemma and renders muscle prone to contraction-induced injury. These phenotypes of dystroglycan-null muscles are recapitulated by Large(myd) muscles, which have an intact dystrophin-glycoprotein complex and lack only the laminin globular domain-binding motif on alpha-dystroglycan. Compromised sarcolemmal integrity is directly shown in Large(myd) muscles and similarly in normal muscles when arenaviruses compete with matrix proteins for binding alpha-dystroglycan. These data provide direct mechanistic insight into how the dystroglycan-linked basal lamina contributes to the maintenance of sarcolemmal integrity and protects muscles from damage.


Asunto(s)
Membrana Basal/fisiología , Distroglicanos/fisiología , Laminina/fisiología , Sarcolema/fisiología , Animales , Sitios de Unión , Distroglicanos/química , Glicosilación , Integrinas/fisiología , Laminina/química , Virus de la Coriomeningitis Linfocítica , Ratones , Distrofia Muscular Animal/etiología
19.
FEBS J ; 289(21): 6460-6462, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35929418

RESUMEN

Skeletal muscle is a structurally and functionally remarkable tissue composed of multinucleated post-mitotic muscle fibres. These fibres are filled with an exquisite, near crystalline array of assembled contractile proteins, capable of coupling ATP utilization to mechanical muscle contraction. Fully differentiated muscle has an incredible ability to protect and repair itself from significant muscle injuries. In fact, through activation of a resident population of stem cells known as satellite cells, muscle fibres can be completely regenerated, and normal function can be restored in a matter of a few weeks after a major myocellular necrotic injury. The loss of key mechanisms to protect muscle from injuries or loss of the capacity to repair muscle after injury is thought to underlie several forms of muscular dystrophy and also the age-related decline of muscle function. In this Subject Collection, The FEBS Journal highlights articles that review or investigate key mechanisms of muscle repair and regeneration in response to injuries, and the contributions of these pathways to health and disease of skeletal muscle.


Asunto(s)
Enfermedades Musculares , Células Satélite del Músculo Esquelético , Humanos , Células Satélite del Músculo Esquelético/metabolismo , Músculo Esquelético/metabolismo , Regeneración/fisiología , Fibras Musculares Esqueléticas/metabolismo , Enfermedades Musculares/metabolismo
20.
Elife ; 112022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35762211

RESUMEN

Background: Patients with cardiomyopathy of Duchenne Muscular Dystrophy (DMD) are at risk of developing life-threatening arrhythmias, but the mechanisms are unknown. We aimed to determine the role of ion channels controlling cardiac excitability in the mechanisms of arrhythmias in DMD patients. Methods: To test whether dystrophin mutations lead to defective cardiac NaV1.5-Kir2.1 channelosomes and arrhythmias, we generated iPSC-CMs from two hemizygous DMD males, a heterozygous female, and two unrelated control males. We conducted studies including confocal microscopy, protein expression analysis, patch-clamping, non-viral piggy-bac gene expression, optical mapping and contractility assays. Results: Two patients had abnormal ECGs with frequent runs of ventricular tachycardia. iPSC-CMs from all DMD patients showed abnormal action potential profiles, slowed conduction velocities, and reduced sodium (INa) and inward rectifier potassium (IK1) currents. Membrane NaV1.5 and Kir2.1 protein levels were reduced in hemizygous DMD iPSC-CMs but not in heterozygous iPSC-CMs. Remarkably, transfecting just one component of the dystrophin protein complex (α1-syntrophin) in hemizygous iPSC-CMs from one patient restored channelosome function, INa and IK1 densities, and action potential profile in single cells. In addition, α1-syntrophin expression restored impulse conduction and contractility and prevented reentrant arrhythmias in hiPSC-CM monolayers. Conclusions: We provide the first demonstration that iPSC-CMs reprogrammed from skin fibroblasts of DMD patients with cardiomyopathy have a dysfunction of the NaV1.5-Kir2.1 channelosome, with consequent reduction of cardiac excitability and conduction. Altogether, iPSC-CMs from patients with DMD cardiomyopathy have a NaV1.5-Kir2.1 channelosome dysfunction, which can be rescued by the scaffolding protein α1-syntrophin to restore excitability and prevent arrhythmias. Funding: Supported by National Institutes of Health R01 HL122352 grant; 'la Caixa' Banking Foundation (HR18-00304); Fundación La Marató TV3: Ayudas a la investigación en enfermedades raras 2020 (LA MARATO-2020); Instituto de Salud Carlos III/FEDER/FSE; Horizon 2020 - Research and Innovation Framework Programme GA-965286 to JJ; the CNIC is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia e Innovación (MCIN) and the Pro CNIC Foundation), and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033). American Heart Association postdoctoral fellowship 19POST34380706s to JVEN. Israel Science Foundation to OB and MA [824/19]. Rappaport grant [01012020RI]; and Niedersachsen Foundation [ZN3452] to OB; US-Israel Binational Science Foundation (BSF) to OB and TH [2019039]; Dr. Bernard Lublin Donation to OB; and The Duchenne Parent Project Netherlands (DPPNL 2029771) to OB. National Institutes of Health R01 AR068428 to DM and US-Israel Binational Science Foundation Grant [2013032] to DM and OB.


Asunto(s)
Proteínas de Unión al Calcio , Cardiomiopatías , Células Madre Pluripotentes Inducidas , Proteínas de la Membrana , Proteínas Musculares , Distrofia Muscular de Duchenne , Canales de Potasio de Rectificación Interna , Potenciales de Acción , Arritmias Cardíacas/metabolismo , Proteínas de Unión al Calcio/genética , Cardiomiopatías/metabolismo , Distrofina/genética , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas Musculares/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio de Rectificación Interna/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA