Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Circ Res ; 131(8): 673-686, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36102198

RESUMEN

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a potentially lethal cardiac arrhythmia syndrome triggered by catecholamines released during exercise, stress, or sudden emotion. Variants in the calsequestrin-2 gene (CASQ2), encoding the major calcium (Ca) binding protein in the sarcoplasmic reticulum (SR), are the second most common cause of CPVT. Recently, several CASQ2 gene variants, such as CASQ2-K180R, have been linked to an autosomal dominant form of Casq2-linked CPVT (CPVT2), but the underlying mechanism is not known. METHODS: A K180R mouse model was generated using CRIPSR/Cas9. Heterozygous and homozygous K180R mice were studied using telemetry ECG recordings in vivo. Ventricular cardiomyocytes were isolated and studied using fluorescent Ca indicators and patch clamp. Expression levels and localization of SR Ca-handling proteins were evaluated using Western blotting and immunostaining. Intra-SR Ca kinetics were quantified using low-affinity Ca indicators. RESULTS: K180R mice exhibit an autosomal dominant CPVT phenotype following exercise or catecholamine stress. Upon catecholamine stress, K180R ventricular cardiomyocytes exhibit increased spontaneous SR Ca release events, triggering delayed afterdepolarizations and spontaneous beats. K180R had no effect on levels of Casq2, Casq2 polymers, or other SR Ca-handling proteins. Intra-SR Ca measurements revealed that K180R impaired dynamic intra-SR Ca buffering, resulting in a more rapid rise of free Ca in the SR during diastole. Steady-state SR Ca buffering and total SR Ca content were not changed. Consistent with the reduced dynamic intra-SR buffering, K180R causes reduced SR Ca release refractoriness. CONCLUSIONS: CASQ2-K180R causes CPVT2 via a heretofore unknown mechanism that differs from CASQ2 variants associated with autosomal recessive CPVT2. Unlike autosomal recessive CASQ2 variants, K180R impairs the dynamic buffering of Ca within the SR without affecting total SR Ca content or Casq2 protein levels. Our data provide insight into the molecular mechanism underlying autosomal dominant CPVT2.


Asunto(s)
Retículo Sarcoplasmático , Taquicardia Ventricular , Animales , Ratones , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Calsecuestrina/genética , Calsecuestrina/metabolismo , Catecolaminas/metabolismo , Miocitos Cardíacos/metabolismo , Polímeros , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo
2.
Hum Mol Genet ; 29(24): 3882-3891, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33355670

RESUMEN

Striated preferentially expressed gene (SPEG), a member of the myosin light chain kinase family, is localized at the level of triad surrounding myofibrils in skeletal muscles. In humans, SPEG mutations are associated with centronuclear myopathy and cardiomyopathy. Using a striated muscle-specific Speg-knockout (KO) mouse model, we have previously shown that SPEG is critical for triad maintenance and calcium handling. Here, we further examined the molecular function of SPEG and characterized the effects of SPEG deficiency on triad and focal adhesion proteins. We used yeast two-hybrid assay, and identified desmin, an intermediate filament protein, to interact with SPEG and confirmed this interaction by co-immunoprecipitation. Using domain-mapping assay, we defined that Ig-like and fibronectin III domains of SPEG interact with rod domain of desmin. In skeletal muscles, SPEG depletion leads to desmin aggregates in vivo and a shift in desmin equilibrium from soluble to insoluble fraction. We also profiled the expression and localization of triadic proteins in Speg-KO mice using western blot and immunofluorescence. The amount of RyR1 and triadin were markedly reduced, whereas DHPRα1, SERCA1 and triadin were abnormally accumulated in discrete areas of Speg-KO myofibers. In addition, Speg-KO muscles exhibited internalized vinculin and ß1 integrin, both of which are critical components of the focal adhesion complex. Further, ß1 integrin was abnormally accumulated in early endosomes of Speg-KO myofibers. These results demonstrate that SPEG-deficient skeletal muscles exhibit several pathological features similar to those seen in MTM1 deficiency. Defects of shared cellular pathways may underlie these structural and functional abnormalities in both types of diseases.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Desmina/metabolismo , Adhesiones Focales/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiología , Músculo Esquelético/patología , Miopatías Estructurales Congénitas/patología , Quinasa de Cadena Ligera de Miosina/fisiología , Animales , Calcio/metabolismo , Moléculas de Adhesión Celular/genética , Desmina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Mutación , Miopatías Estructurales Congénitas/etiología , Miopatías Estructurales Congénitas/metabolismo
3.
J Med Genet ; 58(9): 602-608, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-32994279

RESUMEN

BACKGROUND: Congenital nemaline myopathies are rare pathologies characterised by muscle weakness and rod-shaped inclusions in the muscle fibres. METHODS: Using next-generation sequencing, we identified three patients with pathogenic variants in the Troponin T type 1 (TNNT1) gene, coding for the troponin T (TNT) skeletal muscle isoform. RESULTS: The clinical phenotype was similar in all patients, associating hypotonia, orthopaedic deformities and progressive chronic respiratory failure, leading to early death. The anatomopathological phenotype was characterised by a disproportion in the muscle fibre size, endomysial fibrosis and nemaline rods. Molecular analyses of TNNT1 revealed a homozygous deletion of exons 8 and 9 in patient 1; a heterozygous nonsense mutation in exon 9 and retention of part of intron 4 in muscle transcripts in patient 2; and a homozygous, very early nonsense mutation in patient 3.Western blot analyses confirmed the absence of the TNT protein resulting from these mutations. DISCUSSION: The clinical and anatomopathological presentations of our patients reinforce the homogeneous character of the phenotype associated with recessive TNNT1 mutations. Previous studies revealed an impact of recessive variants on the tropomyosin-binding affinity of TNT. We report in our patients a complete loss of TNT protein due to open reading frame disruption or to post-translational degradation of TNT.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación , Miopatías Nemalínicas/diagnóstico , Miopatías Nemalínicas/genética , Fenotipo , Troponina T/genética , Biopsia , Preescolar , Biología Computacional/métodos , Femenino , Estudios de Asociación Genética/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Inmunohistoquímica , Lactante , Análisis de Secuencia de ADN , Eliminación de Secuencia , Troponina T/metabolismo
4.
Mol Ther ; 28(1): 171-179, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31607542

RESUMEN

Recessive forms of catecholaminergic polymorphic ventricular tachycardia (CPVT) are induced by mutations in genes encoding triadin or calsequestrin, two proteins that belong to the Ca2+ release complex, responsible for intracellular Ca2+ release triggering cardiac contractions. To better understand the mechanisms of triadin-induced CPVT and to assay multiple therapeutic interventions, we used a triadin knockout mouse model presenting a CPVT-like phenotype associated with a decrease in calsequestrin protein level. We assessed different approaches to rescue protein expression and to correct intracellular Ca2+ release and cardiac function: pharmacological treatment with kifunensine or a viral gene transfer-based approach, using adeno-associated virus serotype 2/9 (AAV2/9) encoding the triadin or calsequestrin. We observed that the levels of triadin and calsequestrin are intimately linked, and that reduction of both proteins contributes to the CPVT phenotype. Different combinations of triadin and calsequestrin expression level were obtained using these therapeutic approaches. A full expression of each is not necessary to correct the phenotype; a fine-tuning of the relative re-expression of both triadin and calsequestrin is required to correct the CPVT phenotype and rescue the cardiac function. AAV-mediated gene delivery of calsequestrin or triadin and treatment with kifunensine are potential treatments for recessive forms of CPVT due to triadin mutations.


Asunto(s)
Calsecuestrina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Musculares/metabolismo , Taquicardia Ventricular/metabolismo , Alcaloides/uso terapéutico , Animales , Arritmias Cardíacas/tratamiento farmacológico , Calcio/metabolismo , Señalización del Calcio/genética , Calsecuestrina/genética , Dependovirus , Modelos Animales de Enfermedad , Terapia Genética/métodos , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Contracción Miocárdica/efectos de los fármacos , Contracción Miocárdica/genética , Miocitos Cardíacos/metabolismo , Parvovirinae/genética , Fenotipo , Ratas , Taquicardia Ventricular/tratamiento farmacológico , Taquicardia Ventricular/patología , Transducción Genética , Transfección
5.
J Cell Sci ; 129(20): 3744-3755, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27562070

RESUMEN

In skeletal muscle, the triad is a structure comprising a transverse (T)-tubule and sarcoplasmic reticulum (SR) cisternae. Triads constitute the basis of excitation-contraction coupling as the cradle of the Ca2+ release complex. We have shown previously that triadin, a member of this complex, has shaping properties on reticulum membrane and is indirectly involved in a link between triads and microtubules. We have identified here that CLIMP-63 (also known as CKAP4), as the partner of triadin, is responsible for this association of triads and microtubules. Triadin and CLIMP-63 interact through their respective luminal domains and the shaping properties of triadin depend on the capacity of CLIMP-63 to bind microtubules with its cytosolic portion. In skeletal muscle, CLIMP-63 is localized in the SR, including triads, and is associated with the Ca2+ release complex through its interaction with triadin. Knockout of triadin in muscles results in the delocalization of CLIMP-63 from triads, its dissociation from the Ca2+ release complex and a disorganization of the microtubule network. Our results suggest that the association of triadin and CLIMP-63 could be involved in the shaping of SR terminal cisternae and in the guidance of microtubules close to the triads.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Microtúbulos/metabolismo , Células Musculares/metabolismo , Proteínas Musculares/metabolismo , Animales , Células COS , Proteínas Portadoras/química , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas de la Membrana/química , Ratones Noqueados , Proteínas Musculares/química , Fenotipo , Unión Proteica , Dominios Proteicos , Isoformas de Proteínas/metabolismo , Ratas , Transfección
6.
Hum Mutat ; 38(2): 152-159, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27790796

RESUMEN

Dent-2 disease and Lowe syndrome are two pathologies caused by mutations in inositol polyphosphate 5-phosphatase OCRL gene. Both conditions share proximal tubulopathy evolving to chronic kidney failure. Lowe syndrome is in addition defined by a bilateral congenital cataract, intellectual disability, and hypotonia. The pathology evolves in two decades to a severe condition with renal complications and a fatal issue. We describe here a proof of principle for a targeted gene therapy on a mutation of the OCRL gene that is associated with Lowe syndrome. The affected patient bears a deep intronic mutation inducing a pseudo-exon inclusion in the mRNA, leading to a OCRL-1 protein loss. An exon-skipping strategy was designed to correct the effect of the mutation in cultured cells. We show that a recombinant U7-modified small RNA efficiently triggered the restoration of normal OCRL expression at mRNA and protein levels in patient's fibroblasts. Moreover, the PI(4,5)P2 accumulation and cellular alterations that are hallmark of OCRL-1 dysfunction were also rescued. Altogether, we provide evidence that the restoration of OCRL-1 protein, even at a reduced level, through RNA-based therapy represents a potential therapeutic approach for patients with OCRL splice mutations.


Asunto(s)
Intrones , Mutación , Síndrome Oculocerebrorrenal/genética , Síndrome Oculocerebrorrenal/metabolismo , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Alelos , Empalme Alternativo , Sustitución de Aminoácidos , Preescolar , Activación Enzimática , Exones , Fibroblastos , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Imagen Molecular , Síndrome Oculocerebrorrenal/diagnóstico , Fenotipo
7.
J Physiol ; 593(15): 3261-6, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26228554

RESUMEN

The calcium release complex is the major player in excitation-contraction coupling, both in cardiac and skeletal muscle. The core of the complex is the ryanodine receptor, and triadin is a regulating protein. Nevertheless, the precise function of triadin is only partially understood. Besides its function in the anchoring of calsequestrin at the triad/dyad, our recent results allow us to propose hypotheses on new triadin scaffolding functions, based on the studies performed using different models, from triadin knockout mice to human patients, and expression in non-muscle cells, taking into account the presence of multiple triadin isoforms.


Asunto(s)
Señalización del Calcio , Proteínas Portadoras/metabolismo , Proteínas Musculares/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Congresos como Asunto , Humanos
8.
Circulation ; 129(25): 2673-81, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24888331

RESUMEN

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia is an inherited arrhythmogenic disorder characterized by sudden cardiac death in children. Drug therapy is still insufficient to provide full protection against cardiac arrest, and the use of implantable defibrillators in the pediatric population is limited by side effects. There is therefore a need to explore the curative potential of gene therapy for this disease. We investigated the efficacy and durability of viral gene transfer of the calsequestrin 2 (CASQ2) wild-type gene in a catecholaminergic polymorphic ventricular tachycardia knock-in mouse model carrying the CASQ2(R33Q/R33Q) (R33Q) mutation. METHODS AND RESULTS: We engineered an adeno-associated viral vector serotype 9 (AAV9) containing cDNA of CASQ2 wild-type (AAV9-CASQ2) plus the green fluorescent protein (GFP) gene to infect newborn R33Q mice studied by in vivo and in vitro protocols at 6, 9, and 12 months to investigate the ability of the infection to prevent the disease and adult R33Q mice studied after 2 months to assess whether the AAV9-CASQ2 delivery could revert the catecholaminergic polymorphic ventricular tachycardia phenotype. In both protocols, we observed the restoration of physiological expression and interaction of CASQ2, junctin, and triadin; the rescue of electrophysiological and ultrastructural abnormalities in calcium release units present in R33Q mice; and the lack of life-threatening arrhythmias. CONCLUSIONS: Our data demonstrate that viral gene transfer of wild-type CASQ2 into the heart of R33Q mice prevents and reverts severe manifestations of catecholaminergic polymorphic ventricular tachycardia and that this curative effect lasts for 1 year after a single injection of the vector, thus posing the rationale for the design of a clinical trial.


Asunto(s)
Envejecimiento , Calsecuestrina/genética , Dependovirus/genética , Taquicardia Ventricular/terapia , Animales , Proteínas de Unión al Calcio/metabolismo , Calsecuestrina/metabolismo , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Femenino , Terapia Genética , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Oxigenasas de Función Mixta/metabolismo , Proteínas Musculares/metabolismo , Mutación/genética , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología , Resultado del Tratamiento
9.
Hum Mol Genet ; 22(8): 1483-92, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23236030

RESUMEN

Distal arthrogryposis (DA) is a heterogeneous subgroup of arthrogryposis multiplex congenita (AMC), a large family of disorders characterized by multiple congenital joint limitations due to reduced fetal movements. DA is mainly characterized by contractures afflicting especially the distal extremities without overt muscular or neurological signs. Although a limited number of genes mostly implicated in the contractile apparatus have been identified in DA, most patients failed to show mutations in currently known genes. Using a pangenomic approach, we demonstrated linkage of DA to chromosome 2q37 in two consanguineous families and the endothelin-converting enzyme like 1 (ECEL1) gene present in this region was associated with DA. Screening of a panel of 20 families with non-specific DA identified seven homozygous or compound heterozygous mutations of ECEL1 in a total of six families. Mutations resulted mostly in the absence of protein. ECEL1 is a neuronal endopeptidase predominantly expressed in the central nervous system and brain structures during fetal life in mice and human. ECEL1 plays a major role in intramuscular axonal branching of motor neurons in skeletal muscle during embryogenesis. A detailed review of clinical findings of DA patients with ECEL1 mutations revealed a homogeneous and recognizable phenotype characterized by limited knee flexion, flexed third to fifth fingers and severe muscle atrophy predominant on lower limbs and tongue that suggested a common pathogenic mechanism. We described a new and homogenous phenotype of DA associated with ECEL1 that resulted in symptoms involving rather the peripheral than the central nervous system and suggesting a developmental dysfunction.


Asunto(s)
Artrogriposis/genética , Desarrollo Embrionario/genética , Metaloendopeptidasas/genética , Animales , Artrogriposis/embriología , Artrogriposis/patología , Sistema Nervioso Central/patología , Mapeo Cromosómico , Consanguinidad , Genes Recesivos , Ligamiento Genético , Homocigoto , Humanos , Ratones , Neuronas Motoras/patología , Mutación , Linaje , Fenotipo
10.
Hum Mol Genet ; 21(12): 2759-67, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22422768

RESUMEN

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is an inherited arrhythmogenic disease so far related to mutations in the cardiac ryanodine receptor (RYR2) or the cardiac calsequestrin (CASQ2) genes. Because mutations in RYR2 or in CASQ2 are not retrieved in all CPVT cases, we searched for mutations in the physiological protein partners of RyR2 and CSQ2 in a large cohort of CPVT patients with no detected mutation in these two genes. Based on a candidate gene approach, we focused our investigations on triadin and junctin, two proteins that link RyR2 and CSQ2. Mutations in the triadin (TRDN) and in the junctin (ASPH) genes were searched in a cohort of 97 CPVT patients. We identified three mutations in triadin which cosegregated with the disease on a recessive mode of transmission in two families, but no mutation was found in junctin. Two TRDN mutations, a 4 bp deletion and a nonsense mutation, resulted in premature stop codons; the third mutation, a p.T59R missense mutation, was further studied. Expression of the p.T59R mutant in COS-7 cells resulted in intracellular retention and degradation of the mutant protein. This was confirmed after in vivo expression of the mutant triadin in triadin knock-out mice by viral transduction. In this work, we identified TRDN as a new gene responsible for an autosomal recessive form of CPVT. The mutations identified in the two families lead to the absence of the protein, thereby demonstrating the importance of triadin for the normal function of the cardiac calcium release complex in humans.


Asunto(s)
Arritmias Cardíacas/genética , Proteínas Portadoras/genética , Muerte Súbita Cardíaca , Proteínas Musculares/genética , Taquicardia Ventricular/genética , Animales , Arritmias Cardíacas/metabolismo , Western Blotting , Células COS , Calcio/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Salud de la Familia , Femenino , Genes Recesivos , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , Mutación , Miocitos Cardíacos/metabolismo , Linaje , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/patología
11.
EMBO J ; 29(3): 643-54, 2010 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-20033060

RESUMEN

The alpha1S subunit has a dual function in skeletal muscle: it forms the L-type Ca(2+) channel in T-tubules and is the voltage sensor of excitation-contraction coupling at the level of triads. It has been proposed that L-type Ca(2+) channels might also be voltage-gated sensors linked to transcriptional activity controlling differentiation. By using the U7-exon skipping strategy, we have achieved long-lasting downregulation of alpha1S in adult skeletal muscle. Treated muscles underwent massive atrophy while still displaying significant amounts of alpha1S in the tubular system and being not paralysed. This atrophy implicated the autophagy pathway, which was triggered by neuronal nitric oxide synthase redistribution, activation of FoxO3A, upregulation of autophagy-related genes and autophagosome formation. Subcellular investigations showed that this atrophy was correlated with the disappearance of a minor fraction of alpha1S located throughout the sarcolemma. Our results reveal for the first time that this sarcolemmal fraction could have a role in a signalling pathway determining muscle anabolic or catabolic state and might act as a molecular sensor of muscle activity.


Asunto(s)
Canales de Calcio Tipo L/fisiología , Canales de Calcio/fisiología , Morfogénesis/genética , Músculo Esquelético/embriología , Animales , Autofagia/genética , Secuencia de Bases , Canales de Calcio/genética , Canales de Calcio Tipo L/genética , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Fuerza Muscular/genética , Músculo Esquelético/anatomía & histología , Músculo Esquelético/patología , Atrofia Muscular/genética , Atrofia Muscular/patología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Tamaño de los Órganos/genética , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Distribución Tisular/genética
12.
J Cell Sci ; 125(Pt 14): 3443-53, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22505613

RESUMEN

The terminal cisternae represent one of the functional domains of the skeletal muscle sarcoplasmic reticulum (SR). They are closely apposed to plasma membrane invaginations, the T-tubules, with which they form structures called triads. In triads, the physical interaction between the T-tubule-anchored voltage-sensing channel DHPR and the SR calcium channel RyR1 is essential because it allows the depolarization-induced calcium release that triggers muscle contraction. This interaction between DHPR and RyR1 is based on the peculiar membrane structures of both T-tubules and SR terminal cisternae. However, little is known about the molecular mechanisms governing the formation of SR terminal cisternae. We have previously shown that ablation of triadins, a family of SR transmembrane proteins that interact with RyR1, induced skeletal muscle weakness in knockout mice as well as a modification of the shape of triads. Here we explore the intrinsic molecular properties of the longest triadin isoform Trisk 95. We show that when ectopically expressed, Trisk 95 can modulate reticulum membrane morphology. The membrane deformations induced by Trisk 95 are accompanied by modifications of the microtubule network organization. We show that multimerization of Trisk 95 by disulfide bridges, together with interaction with microtubules, are responsible for the ability of Trisk 95 to structure reticulum membrane. When domains responsible for these molecular properties are deleted, anchoring of Trisk 95 to the triads in muscle cells is strongly decreased, suggesting that oligomers of Trisk 95 and microtubules contribute to the organization of the SR terminal cisternae in a triad.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Células COS , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/genética , Chlorocebus aethiops , Cisteína/metabolismo , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Contracción Muscular/fisiología , Proteínas Musculares/biosíntesis , Proteínas Musculares/genética , Ratas , Transfección
13.
Brain ; 135(Pt 4): 1115-27, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22418739

RESUMEN

The skeletal muscle ryanodine receptor is an essential component of the excitation-contraction coupling apparatus. Mutations in RYR1 are associated with several congenital myopathies (termed RYR1-related myopathies) that are the most common non-dystrophic muscle diseases of childhood. Currently, no treatments exist for these disorders. Although the primary pathogenic abnormality involves defective excitation-contraction coupling, other abnormalities likely play a role in disease pathogenesis. In an effort to discover novel pathogenic mechanisms, we analysed two complementary models of RYR1-related myopathies, the relatively relaxed zebrafish and cultured myotubes from patients with RYR1-related myopathies. Expression array analysis in the zebrafish disclosed significant abnormalities in pathways associated with cellular stress. Subsequent studies focused on oxidative stress in relatively relaxed zebrafish and RYR1-related myopathy myotubes and demonstrated increased oxidant activity, the presence of oxidative stress markers, excessive production of oxidants by mitochondria and diminished survival under oxidant conditions. Exposure to the antioxidant N-acetylcysteine reduced oxidative stress and improved survival in the RYR1-related myopathies human myotubes ex vivo and led to significant restoration of aspects of muscle function in the relatively relaxed zebrafish, thereby confirming its efficacy in vivo. We conclude that oxidative stress is an important pathophysiological mechanism in RYR1-related myopathies and that N-acetylcysteine is a successful treatment modality ex vivo and in a vertebrate disease model. We propose that N-acetylcysteine represents the first potential therapeutic strategy for these debilitating muscle diseases.


Asunto(s)
Acetilcisteína/uso terapéutico , Antioxidantes/uso terapéutico , Enfermedades Musculares/tratamiento farmacológico , Enfermedades Musculares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Acetofenonas/farmacología , Animales , Animales Modificados Genéticamente , Conducta Animal , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Humanos , Indometacina/farmacología , Larva , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Contracción Muscular/genética , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Fibras Musculares Esqueléticas/ultraestructura , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Mutación/genética , Estrés Oxidativo/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Pez Cebra
14.
Curr Opin Pharmacol ; 68: 102330, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36529094

RESUMEN

Myopathies related to variations in the RYR1 gene are genetic diseases for which the therapeutic options are sparse, in part because of the very large size of the gene and protein, and of the distribution of variations all along the sequence. Taking advantage of the progress made in the gene therapy field, different approaches can be applied to the different genetic variations, either at the mRNA level or directly at the DNA level, specifically with the new gene editing tools. Some of those have already been tested in cellulo and/or in vivo, and for the development of the most innovative gene editing technology, inspiration can be sought in other genetic diseases.


Asunto(s)
Enfermedades Musculares , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Enfermedades Musculares/genética , Enfermedades Musculares/terapia , Enfermedades Musculares/metabolismo , Músculo Esquelético/metabolismo , Mutación
15.
J Gen Physiol ; 155(1)2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36409218

RESUMEN

The expression of the Huntingtin protein, well known for its involvement in the neurodegenerative Huntington's disease, has been confirmed in skeletal muscle. The impact of HTT deficiency was studied in human skeletal muscle cell lines and in a mouse model with inducible and muscle-specific HTT deletion. Characterization of calcium fluxes in the knock-out cell lines demonstrated a reduction in excitation-contraction (EC) coupling, related to an alteration in the coupling between the dihydropyridine receptor and the ryanodine receptor, and an increase in the amount of calcium stored within the sarcoplasmic reticulum, linked to the hyperactivity of store-operated calcium entry (SOCE). Immunoprecipitation experiments demonstrated an association of HTT with junctophilin 1 (JPH1) and stromal interaction molecule 1 (STIM1), both providing clues on the functional effects of HTT deletion on calcium fluxes. Characterization of muscle strength and muscle anatomy of the muscle-specific HTT-KO mice demonstrated that HTT deletion induced moderate muscle weakness and mild muscle atrophy associated with histological abnormalities, similar to the phenotype observed in tubular aggregate myopathy. Altogether, this study points toward the hypotheses of the involvement of HTT in EC coupling via its interaction with JPH1, and on SOCE via its interaction with JPH1 and/or STIM1.


Asunto(s)
Calcio , Retículo Sarcoplasmático , Ratones , Humanos , Animales , Calcio/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Retículo Sarcoplasmático/metabolismo , Músculo Esquelético/metabolismo , Acoplamiento Excitación-Contracción/fisiología
16.
Stem Cell Reports ; 18(5): 1075-1089, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-37163978

RESUMEN

Triadin knockout syndrome (TKOS) is a malignant arrhythmia disorder caused by recessive null variants in TRDN-encoded cardiac triadin. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were generated from two unrelated TKOS patients and an unrelated control. CRISPR-Cas9 gene editing was used to insert homozygous TRDN-p.D18fs∗13 into a control line to generate a TKOS model (TRDN-/-). Western blot confirmed total knockout of triadin in patient-specific and TRDN-/- iPSC-CMs. iPSC-CMs from both patients revealed a prolonged action potential duration (APD) at 90% repolarization, and this was normalized by protein replacement of triadin. APD prolongation was confirmed in TRDN-/- iPSC-CMs. TRDN-/- iPSC-CMs revealed that loss of triadin underlies decreased expression and co-localization of key calcium handling proteins, slow and decreased calcium release from the sarcoplasmic reticulum, and slow inactivation of the L-type calcium channel leading to frequent cellular arrhythmias, including early and delayed afterdepolarizations and APD alternans.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Calcio/metabolismo , Arritmias Cardíacas/patología , Síndrome , Potenciales de Acción
17.
Elife ; 122023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37083699

RESUMEN

Excitation-contraction coupling requires a highly specialized membrane structure, the triad, composed of a plasma membrane invagination, the T-tubule, surrounded by two sarcoplasmic reticulum terminal cisternae. Although the precise mechanisms governing T-tubule biogenesis and triad formation remain largely unknown, studies have shown that caveolae participate in T-tubule formation and mutations of several of their constituents induce muscle weakness and myopathies. Here, we demonstrate that, at the plasma membrane, Bin1 and caveolae composed of caveolin-3 assemble into ring-like structures from which emerge tubes enriched in the dihydropyridine receptor. Bin1 expression lead to the formation of both rings and tubes and we show that Bin1 forms scaffolds on which caveolae accumulate to form the initial T-tubule. Cav3 deficiency caused by either gene silencing or pathogenic mutations results in defective ring formation and perturbed Bin1-mediated tubulation that may explain defective T-tubule organization in mature muscles. Our results uncover new pathophysiological mechanisms that may prove relevant to myopathies caused by Cav3 or Bin1 dysfunction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Caveolas , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Canales de Calcio Tipo L/metabolismo , Caveolas/metabolismo , Membrana Celular/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Ratones
18.
Curr Pharm Des ; 28(1): 15-25, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34514983

RESUMEN

RyR1-related myopathies are a family of genetic neuromuscular diseases due to mutations in the RYR1 gene. No treatment exists for any of these myopathies today, which could change in the coming years with the growing number of studies dedicated to the pre-clinical assessment of various approaches, from pharmacological to gene therapy strategies, using the numerous models developed up to now. In addition, the first clinical trials for these rare diseases have just been completed or are being launched. We review the most recent results obtained for the treatment of RyR1-related myopathies, and, in view of the progress in therapeutic development for other myopathies, we discuss the possible future therapeutic perspectives for RyR1-related myopathies.


Asunto(s)
Enfermedades Musculares , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Músculo Esquelético , Enfermedades Musculares/genética , Enfermedades Musculares/terapia , Mutación , Canal Liberador de Calcio Receptor de Rianodina/genética
19.
PLoS One ; 17(2): e0264146, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35213584

RESUMEN

Triadin, a protein of the sarcoplasmic reticulum (SR) of striated muscles, anchors the calcium-storing protein calsequestrin to calcium release RyR channels at the junction with t-tubules, and modulates these channels by conformational effects. Triadin ablation induces structural SR changes and alters the expression of other proteins. Here we quantify alterations of calcium signaling in single skeletal myofibers of constitutive triadin-null mice. We find higher resting cytosolic and lower SR-luminal [Ca2+], 40% lower calsequestrin expression, and more CaV1.1, RyR1 and SERCA1. Despite the increased CaV1.1, the mobile intramembrane charge was reduced by ~20% in Triadin-null fibers. The initial peak of calcium release flux by pulse depolarization was minimally altered in the null fibers (revealing an increase in peak calcium permeability). The "hump" phase that followed, attributable to calcium detaching from calsequestrin, was 25% lower, a smaller change than expected from the reduced calsequestrin content and calcium saturation. The exponential decay rate of calcium transients was 25% higher, consistent with the higher SERCA1 content. Recovery of calcium flux after a depleting depolarization was faster in triadin-null myofibers, consistent with the increased uptake rate and lower SR calsequestrin content. In sum, the triadin knockout determines an increased RyR1 channel openness, which depletes the SR, a substantial loss of calsequestrin and gains in other couplon proteins. Powerful functional compensations ensue: activation of SOCE that increases [Ca2+]cyto; increased SERCA1 activity, which limits the decrease in [Ca2+]SR and a restoration of SR calcium storage of unknown substrate. Together, they effectively limit the functional loss in skeletal muscles.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Proteínas Musculares/deficiencia , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Retículo Sarcoplasmático/metabolismo , Animales , Canales de Calcio Tipo L/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Mutantes , Proteínas Musculares/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/genética , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética
20.
J Vis Exp ; (184)2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35781470

RESUMEN

One important application of clustered regulatory interspaced short palindromic repeats (CRISPR)/Cas 9 is the development of knock-out cell lines, specifically to study the function of new genes/proteins associated with a disease, identified during the genetic diagnosis. For the development of such cell lines, two major issues have to be untangled: insertion of the CRISPR tools (the Cas9 and the guide RNA) with high efficiency into the chosen cells, and restriction of the Cas9 activity to the specific deletion of the chosen gene. The protocol described here is dedicated to the insertion of the CRISPR tools in difficult to transfect cells, such as muscle cells. This protocol is based on the use of lentiviruses, produced with plasmids publicly available, for which all the cloning steps are described to target a gene of interest. The control of Cas9 activity has been performed using an adaptation of a previously described system called KamiCas9, in which the transduction of the cells with a lentivirus encoding a guide RNA targeting the Cas9 allows the progressive abolition of Cas9 expression. This protocol has been applied to the development of a RYR1-knock out human muscle cell line, which has been further characterized at the protein and functional level, to confirm the knockout of this important calcium channel involved in muscle intracellular calcium release and in excitation-contraction coupling. The procedure described here can easily be applied to other genes in muscle cells or in other difficult to transfect cells and produce valuable tools to study these genes in human cells.


Asunto(s)
Edición Génica , ARN Guía de Kinetoplastida , Sistemas CRISPR-Cas , Línea Celular , Edición Génica/métodos , Humanos , Lentivirus/genética , Células Musculares , Músculos , ARN Guía de Kinetoplastida/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA