Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Biomed Res Int ; 2019: 7638946, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31165076

RESUMEN

The skeletal muscle ryanodine receptor (RyR1), i.e., the Ca2+ channel of the sarco/endoplasmic reticulum (S/ER), and the voltage-dependent calcium channel Cav1.1 are the principal channels involved in excitation-contraction coupling in skeletal muscle. RYR1 gene variants are linked to distinct skeletal muscle disorders, including malignant hyperthermia susceptibility and central core disease (CCD), mainly with autosomal dominant inheritance, and autosomal recessive myopathies with a broad phenotypic and histopathological spectrum. The age at onset of RYR1-related myopathies varies from infancy to adulthood. We report the identification of four RYR1 variants in two Italian families: one with myopathy and variants c.4003C>T (p.R1335C) and c.7035C>A (p.S2345R), and another with CCD and variants c.9293G>T (p.S3098I) and c.14771_14772insTAGACAGGGTGTTGCTCTGTTGCCCTTCTT (p.F4924_V4925insRQGVALLPFF). We demonstrate that, in patient-specific lymphoblastoid cells, the c.4003C>T (p.R1335C) variant is not expressed and the in-frame 30-nucleotide insertion variant is expressed at a low level. Moreover, Ca2+ release in response to the RyR1 agonist 4-chloro-m-cresol and to thapsigargin showed that the c.7035C>A (p.S2345R) variant causes depletion of S/ER Ca2+ stores and that the compound heterozygosity for variant c.9293G>T (p.S3098I) and the 30-nucleotide insertion increases RyR1-dependent Ca2+ release without affecting ER Ca2+ stores. In conclusion, we detected and functionally characterized disease-causing variants of the RyR1 channel in patient-specific lymphoblastoid cells. This paper is dedicated to the memory and contribution of Luigi Del Vecchio.


Asunto(s)
Familia , Regulación de la Expresión Génica , Variación Genética , Hipertermia Maligna , Músculo Esquelético , Miopatía del Núcleo Central , Canal Liberador de Calcio Receptor de Rianodina , Adulto , Preescolar , Femenino , Humanos , Italia , Masculino , Hipertermia Maligna/genética , Hipertermia Maligna/metabolismo , Hipertermia Maligna/patología , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/metabolismo , Miopatía del Núcleo Central/patología , Canal Liberador de Calcio Receptor de Rianodina/biosíntesis , Canal Liberador de Calcio Receptor de Rianodina/genética
2.
Biochem J ; 474(16): 2749-2761, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28687594

RESUMEN

Mutations in the skeletal muscle ryanodine receptor (RyR1) cause malignant hyperthermia (MH) and central core disease (CCD), whereas mutations in the cardiac ryanodine receptor (RyR2) lead to catecholaminergic polymorphic ventricular tachycardia (CPVT). Most disease-associated RyR1 and RyR2 mutations are located in the N-terminal, central, and C-terminal regions of the corresponding ryanodine receptor (RyR) isoform. An increasing body of evidence demonstrates that CPVT-associated RyR2 mutations enhance the propensity for spontaneous Ca2+ release during store Ca2+ overload, a process known as store overload-induced Ca2+ release (SOICR). Considering the similar locations of disease-associated RyR1 and RyR2 mutations in the RyR structure, we hypothesize that like CPVT-associated RyR2 mutations, MH/CCD-associated RyR1 mutations also enhance SOICR. To test this hypothesis, we determined the impact on SOICR of 12 MH/CCD-associated RyR1 mutations E2347-del, R2163H, G2434R, R2435L, R2435H, and R2454H located in the central region, and Y4796C, T4826I, L4838V, A4940T, G4943V, and P4973L located in the C-terminal region of the channel. We found that all these RyR1 mutations reduced the threshold for SOICR. Dantrolene, an acute treatment for MH, suppressed SOICR in HEK293 cells expressing the RyR1 mutants R164C, Y523S, R2136H, R2435H, and Y4796C. Interestingly, carvedilol, a commonly used ß-blocker that suppresses RyR2-mediated SOICR, also inhibits SOICR in these RyR1 mutant HEK293 cells. Therefore, these results indicate that a reduced SOICR threshold is a common defect of MH/CCD-associated RyR1 mutations, and that carvedilol, like dantrolene, can suppress RyR1-mediated SOICR. Clinical studies of the effectiveness of carvedilol as a long-term treatment for MH/CCD or other RyR1-associated disorders may be warranted.


Asunto(s)
Señalización del Calcio , Hipertermia Maligna/genética , Modelos Moleculares , Miopatía del Núcleo Central/genética , Mutación Puntual , Canal Liberador de Calcio Receptor de Rianodina/genética , Antagonistas Adrenérgicos beta/farmacología , Sustitución de Aminoácidos , Animales , Señalización del Calcio/efectos de los fármacos , Carbazoles/farmacología , Carvedilol , Dantroleno/farmacología , Transferencia Resonante de Energía de Fluorescencia , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Hipertermia Maligna/tratamiento farmacológico , Hipertermia Maligna/metabolismo , Microscopía Fluorescente , Relajantes Musculares Centrales/farmacología , Mutagénesis Sitio-Dirigida , Miopatía del Núcleo Central/metabolismo , Propanolaminas/farmacología , Conformación Proteica , Conejos , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Análisis de la Célula Individual
3.
J Neuromuscul Dis ; 4(2): 147-158, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28527222

RESUMEN

BACKGROUND: Central core disease and malignant hyperthermia are human disorders of skeletal muscle resulting from aberrant Ca2+ handling. Most malignant hyperthermia and central core disease cases are associated with amino acid changes in the type 1 ryanodine receptor (RyR1), the skeletal muscle Ca2+-release channel. Malignant hyperthermia exhibits a gain-of-function phenotype, and central core disease results from loss of channel function. For a variant to be classified as pathogenic, functional studies must demonstrate a correlation with the pathophysiology of malignant hyperthermia or central core disease. OBJECTIVE: We assessed the pathogenicity of four C-terminal variants of the ryanodine receptor using functional analysis. The variants were identified in families affected by either malignant hyperthermia or central core disease. METHODS: Four variants were introduced separately into human cDNA encoding the skeletal muscle ryanodine receptor. Following transient expression in HEK-293T cells, functional studies were carried out using calcium release assays in response to an agonist. Two previously characterized variants and wild-type skeletal muscle ryanodine receptor were used as controls. RESULTS: The p.Met4640Ile variant associated with central core disease showed no difference in calcium release compared to wild-type. The p.Val4849Ile variant associated with malignant hyperthermia was more sensitive to agonist than wild-type but did not reach statistical significance and two variants (p.Phe4857Ser and p.Asp4918Asn) associated with central core disease were completely inactive. CONCLUSIONS: The p.Val4849Ile variant should be considered a risk factor for malignant hyperthermia, while the p.Phe4857Ser and p.Asp4918Asn variants should be classified as pathogenic for central core disease.


Asunto(s)
Variación Genética , Hipertermia Maligna/genética , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Adulto , Anciano , Calcio/metabolismo , Familia , Femenino , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Masculino , Hipertermia Maligna/metabolismo , Hipertermia Maligna/terapia , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Miopatía del Núcleo Central/metabolismo , Miopatía del Núcleo Central/terapia , Linaje
4.
Hum Mutat ; 37(11): 1231-1241, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27586648

RESUMEN

Type 1 ryanodine receptor (RYR1) is a Ca2+ release channel in the sarcoplasmic reticulum of skeletal muscle and is mutated in some muscle diseases, including malignant hyperthermia (MH) and central core disease (CCD). Over 200 mutations associated with these diseases have been identified, and most mutations accelerate Ca2+ -induced Ca2+ release (CICR), resulting in abnormal Ca2+ homeostasis in skeletal muscle. However, it remains largely unknown how specific mutations cause different phenotypes. In this study, we investigated the CICR activity of 14 mutations at 10 different positions in the central region of RYR1 (10 MH and four MH/CCD mutations) using a heterologous expression system in HEK293 cells. In live-cell Ca2+ imaging, the mutant channels exhibited an enhanced sensitivity to caffeine, a reduced endoplasmic reticulum Ca2+ content, and an increased resting cytoplasmic Ca2+ level. The three parameters for CICR (Ca2+ sensitivity for activation, Ca2+ sensitivity for inactivation, and attainable maximum activity, i.e., gain) were obtained by [3 H]ryanodine binding and fitting analysis. The mutant channels showed increased gain and Ca2+ sensitivity for activation in a site-specific manner. Genotype-phenotype correlations were explained well by the near-atomic structure of RYR1. Our data suggest that divergent CICR activity may cause various disease phenotypes by specific mutations.


Asunto(s)
Calcio/metabolismo , Hipertermia Maligna/genética , Mutación , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Retículo Endoplásmico/metabolismo , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Hipertermia Maligna/metabolismo , Modelos Moleculares , Miopatía del Núcleo Central/metabolismo , Estructura Secundaria de Proteína , Canal Liberador de Calcio Receptor de Rianodina/química , Retículo Sarcoplasmático/metabolismo
5.
PLoS One ; 10(6): e0130606, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26115329

RESUMEN

The type 1 ryanodine receptor (RyR1) is a Ca2+ release channel in the sarcoplasmic reticulum of skeletal muscle and is mutated in several diseases, including malignant hyperthermia (MH) and central core disease (CCD). Most MH and CCD mutations cause accelerated Ca2+ release, resulting in abnormal Ca2+ homeostasis in skeletal muscle. However, how specific mutations affect the channel to produce different phenotypes is not well understood. In this study, we have investigated 11 mutations at 7 different positions in the amino (N)-terminal region of RyR1 (9 MH and 2 MH/CCD mutations) using a heterologous expression system in HEK293 cells. In live-cell Ca2+ imaging at room temperature (~25 °C), cells expressing mutant channels exhibited alterations in Ca2+ homeostasis, i.e., an enhanced sensitivity to caffeine, a depletion of Ca2+ in the ER and an increase in resting cytoplasmic Ca2+. RyR1 channel activity was quantitatively evaluated by [3H]ryanodine binding and three parameters (sensitivity to activating Ca2+, sensitivity to inactivating Ca2+ and attainable maximum activity, i.e., gain) were obtained by fitting analysis. The mutations increased the gain and the sensitivity to activating Ca2+ in a site-specific manner. The gain was consistently higher in both MH and MH/CCD mutations. Sensitivity to activating Ca2+ was markedly enhanced in MH/CCD mutations. The channel activity estimated from the three parameters provides a reasonable explanation to the pathological phenotype assessed by Ca2+ homeostasis. These properties were also observed at higher temperatures (~37 °C). Our data suggest that divergent activity profiles may cause varied disease phenotypes by specific mutations. This approach should be useful for diagnosis and treatment of diseases with mutations in RyR1.


Asunto(s)
Calcio/metabolismo , Citoplasma/metabolismo , Hipertermia Maligna/metabolismo , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Línea Celular , Humanos , Mutación , Canal Liberador de Calcio Receptor de Rianodina/genética
6.
Neuromuscul Disord ; 25(7): 567-76, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25958340

RESUMEN

Malignant hyperthermia (MH) is a potentially fatal pharmacogenetic myopathy triggered by exposure to volatile anesthetics and/or depolarizing muscle relaxants. Susceptibility to MH is primarily associated with dominant mutations in the ryanodine receptor type 1 gene (RYR1). Recent genetic studies have shown that RYR1 variants are the most common cause of dominant and recessive congenital myopathies - central core and multi-minicore disease, congenital fiber type disproportion, and centronuclear myopathy. However, the MH status of many patients, especially with recessive RYR1-related myopathies, remains uncertain. We report the occurrence of a triplet of RYR1 variants, c.4711A>G (p.Ile1571Val), c.10097G>A (p.Arg3366His), c.11798A>G (p.Tyr3933Cys), found in cis in four unrelated families, one from Belgium, one from The Netherlands and two from Canada. Phenotype-genotype correlation analysis indicates that the presence of the triplet allele alone confers susceptibility to MH, and that the presence of this allele in a compound heterozygous state with the MH-associated RYR1 variant c.14545G>A (p.Val4849Ile) results in the MH susceptibility phenotype and a congenital myopathy with cores and rods. Our study underlines the notion that assigning pathogenicity to individual RYR1 variants or combination of variants, and counseling in RYR1-related myopathies may require integration of clinical, histopathological, in vitro contracture testing, MRI and genetic findings.


Asunto(s)
Predisposición Genética a la Enfermedad , Heterocigoto , Hipertermia Maligna/genética , Miopatía del Núcleo Central/genética , Fenotipo , Canal Liberador de Calcio Receptor de Rianodina/genética , Adulto , Niño , Preescolar , Familia , Femenino , Estudios de Asociación Genética , Variación Genética , Humanos , Pierna/patología , Masculino , Hipertermia Maligna/metabolismo , Hipertermia Maligna/patología , Persona de Mediana Edad , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miopatía del Núcleo Central/metabolismo , Miopatía del Núcleo Central/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Población Blanca/genética
7.
Eur J Pediatr ; 173(12): 1691-4, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24706162

RESUMEN

UNLABELLED: We describe a 5-year-old girl with marked hypotonia, poor feeding and reduced facial expression since birth. Congenital myopathy was suspected; muscle biopsy showed unspecific type 1 fibre predominance. The possibility of a ryanodine receptor 1 gene (RYR1)-associated myopathy was considered, but not further investigated. At the age of 2 years, she presented with exophthalmos. Brain MRI revealed optic pathway glioma. On clinical examination, she had six café-au-lait spots, thus fulfilling the diagnostic criteria for neurofibromatosis type 1 (NF1). The hypotonia was then attributed to NF1. At the age of 3 years, she developed scoliosis and had an unusually severe motor delay for NF1, as she was not able to walk independently. Dual pathology was suspected, and muscle MRI showed the typical pattern for RYR1-related myopathy. This was genetically confirmed with the discovery of two heterozygous mutations. CONCLUSION: NF1 is one of the most frequent genetic diseases in children. RYR1-related myopathy is one of the most frequent causes of congenital myopathy. The combination of these two pathologies has not yet been described. In cases of unusual presentations or clinical course, the possibility of genetic "double trouble" should be considered.


Asunto(s)
Anomalías Múltiples , ADN/genética , Genes de Neurofibromatosis 1 , Mutación Missense , Miopatía del Núcleo Central/genética , Neurofibromatosis 1/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Biopsia , Preescolar , Análisis Mutacional de ADN , Diagnóstico Diferencial , Femenino , Humanos , Imagen por Resonancia Magnética , Miopatía del Núcleo Central/diagnóstico , Miopatía del Núcleo Central/metabolismo , Neurofibromatosis 1/diagnóstico , Neurofibromatosis 1/metabolismo , Fenotipo , Músculo Cuádriceps/metabolismo , Músculo Cuádriceps/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
8.
Curr Drug Targets ; 12(5): 709-23, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21291389

RESUMEN

The ryanodine receptor (RyR) calcium release channel is an essential intracellular ion channel that is central to Ca(2+) signaling and contraction in the heart and skeletal muscle. The rapid release of Ca(2+) from the internal sarcoplasmic reticulum Ca(2+) stores through the RyR during excitation-contraction coupling is facilitated by the unique arrangement of the surface and sarcoplasmic reticulum membrane systems. Debilitating and sometimes fatal skeletal and cardiomyopathies result from changes in RyR activity that disrupt normal Ca(2+) signaling. Such changes can be caused by point mutations in many different regions of the RyR protein or acquired as a result of stress associated with exercise, heart failure, age or drugs. In general, both inherited and acquired changes include an increase in RyR channel activity. Because of its central function, the RyR is a potential therapeutic target for the inherited disorders and many of the acquired disorders. The RyR is currently used as a therapeutic target in malignant hyperthermia where dantrolene is effective and to relieve ventricular arrhythmia, with the use of JTV519 and flecainide. These drugs show that the RyR is a valid therapeutic target, but have side effects that prevent their chronic use. Thus there is an urgent need for the development of skeletal and cardiac specific drugs to treat these diverse muscle disorders. In this review, we discuss the mutations that cause skeletal myopathies and cardiac arrhythmias and how these mutations pinpoint residues within the RyR protein that are functionally significant and might be developed as targets for therapeutic drugs.


Asunto(s)
Calcio/metabolismo , Cardiopatías/metabolismo , Hipertermia Maligna/metabolismo , Terapia Molecular Dirigida , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Señalización del Calcio , Modelos Animales de Enfermedad , Cardiopatías/tratamiento farmacológico , Cardiopatías/genética , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/metabolismo , Humanos , Hipertermia Maligna/tratamiento farmacológico , Hipertermia Maligna/genética , Mutación , Miopatía del Núcleo Central/tratamiento farmacológico , Miopatía del Núcleo Central/genética
9.
Hum Mol Genet ; 20(3): 589-600, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21088110

RESUMEN

Prolonged depolarization of skeletal muscle cells induces entry of extracellular calcium into muscle cells, an event referred to as excitation-coupled calcium entry. Skeletal muscle excitation-coupled calcium entry relies on the interaction between the 1,4-dihydropyridine receptor on the sarcolemma and the ryanodine receptor on the sarcoplasmic reticulum membrane. In this study, we directly measured excitation-coupled calcium entry by total internal reflection fluorescence microscopy in human skeletal muscle myotubes harbouring mutations in the RYR1 gene linked to malignant hyperthermia (MH) and central core disease (CCD). We found that excitation-coupled calcium entry is strongly enhanced in cells from patients with CCD compared with individuals with MH and controls. Furthermore, excitation-coupled calcium entry induces generation of reactive nitrogen species and enhances nuclear localization of NFATc1, which in turn may be responsible for the increased IL-6 released by myotubes from patients with CCD.


Asunto(s)
Calcio/metabolismo , Interleucina-6/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Miopatía del Núcleo Central/metabolismo , Factores de Transcripción NFATC/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Hipertermia Maligna/genética , Microscopía Fluorescente , Músculo Esquelético/metabolismo , Mutación , Miopatía del Núcleo Central/genética , Reacción en Cadena de la Polimerasa , Especies de Nitrógeno Reactivo/biosíntesis , Especies de Nitrógeno Reactivo/metabolismo
10.
Neuropathol Appl Neurobiol ; 37(3): 271-84, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21062345

RESUMEN

AIMS: To report the clinical, pathological and genetic findings in a group of patients with a previously not described phenotype of congenital myopathy due to recessive mutations in the gene encoding the type 1 muscle ryanodine receptor channel (RYR1). METHODS: Seven unrelated patients shared a predominant axial and proximal weakness of varying severity, with onset during the neonatal period, associated with bilateral ptosis and ophthalmoparesis, and unusual muscle biopsy features at light and electron microscopic levels. RESULTS: Muscle biopsy histochemistry revealed a peculiar morphological pattern characterized by numerous internalized myonuclei in up to 51% of fibres and large areas of myofibrillar disorganization with undefined borders. Ultrastructurally, such areas frequently occupied the whole myofibre cross section and extended to a moderate number of sarcomeres in length. Molecular genetic investigations identified recessive mutations in the ryanodine receptor (RYR1) gene in six compound heterozygous patients and one homozygous patient. Nine mutations are novel and four have already been reported either as pathogenic recessive mutations or as changes affecting a residue associated with dominant malignant hyperthermia susceptibility. Only two mutations were located in the C-terminal transmembrane domain whereas the others were distributed throughout the cytoplasmic region of RyR1. CONCLUSION: Our data enlarge the spectrum of RYR1 mutations and highlight their clinical and morphological heterogeneity. A congenital myopathy featuring ptosis and external ophthalmoplegia, concomitant with the novel histopathological phenotype showing fibres with large, poorly delimited areas of myofibrillar disorganization and internal nuclei, is highly suggestive of an RYR1-related congenital myopathy.


Asunto(s)
Mutación , Miofibrillas/ultraestructura , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/metabolismo , Miopatía del Núcleo Central/patología , Canal Liberador de Calcio Receptor de Rianodina/genética , Adolescente , Adulto , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Niño , Femenino , Genes Recesivos , Humanos , Inmunohistoquímica , Masculino , Microscopía Electrónica de Transmisión , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Adulto Joven
11.
Neuromuscul Disord ; 19(5): 344-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19303294

RESUMEN

Mutations in the skeletal muscle ryanodine receptor (RYR1) gene have been associated with a wide range of phenotypes including the malignant hyperthermia (MH) susceptibility trait, Central Core Disease (CCD) and other congenital myopathies characterized by early onset and predominant proximal weakness. We report a patient presenting at 77 years with a predominant axial myopathy associated with prominent involvement of spine extensors, confirmed on MRI and muscle biopsy, compatible with a core myopathy. RYR1 mutational analysis revealed a novel heterozygous missense mutation (c.119G>T; p.Gly40Val) affecting the RYR1 N-terminus, previously predominantly associated with MH susceptibility. This case expands the spectrum of RYR1-related phenotypes and suggests that MH-related RYR1 mutations may give rise to overt neuromuscular symptoms later in life, with clinical features not typically found in CCD due to C-terminal hotspot mutations. Late-onset congenital myopathies may be under-recognised and diagnosis requires a high degree of clinical suspicion.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Músculo Esquelético/metabolismo , Mutación/genética , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Factores de Edad , Edad de Inicio , Anciano , Señalización del Calcio/genética , Análisis Mutacional de ADN , Progresión de la Enfermedad , Genes Dominantes/genética , Marcadores Genéticos , Genotipo , Heterocigoto , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Masculino , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miopatía del Núcleo Central/patología
12.
Am J Hum Genet ; 79(5): 859-68, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17033962

RESUMEN

Epigenetic regulation of gene expression is a source of genetic variation, which can mimic recessive mutations by creating transcriptional haploinsufficiency. Germline epimutations and genomic imprinting are typical examples, although their existence can be difficult to reveal. Genomic imprinting can be tissue specific, with biallelic expression in some tissues and monoallelic expression in others or with polymorphic expression in the general population. Mutations in the skeletal-muscle ryanodine-receptor gene (RYR1) are associated with malignant hyperthermia susceptibility and the congenital myopathies central core disease and multiminicore disease. RYR1 has never been thought to be affected by epigenetic regulation. However, during the RYR1-mutation analysis of a cohort of patients with recessive core myopathies, we discovered that 6 (55%) of 11 patients had monoallelic RYR1 transcription in skeletal muscle, despite being heterozygous at the genomic level. In families for which parental DNA was available, segregation studies showed that the nonexpressed allele was maternally inherited. Transcription analysis in patients' fibroblasts and lymphoblastoid cell lines indicated biallelic expression, which suggests tissue-specific silencing. Transcription analysis of normal human fetal tissues showed that RYR1 was monoallelically expressed in skeletal and smooth muscles, brain, and eye in 10% of cases. In contrast, 25 normal adult human skeletal-muscle samples displayed only biallelic expression. Finally, the administration of the DNA methyltransferase inhibitor 5-aza-deoxycytidine to cultured patient skeletal-muscle myoblasts reactivated the transcription of the silenced allele, which suggests hypermethylation as a mechanism for RYR1 silencing. Our data indicate that RYR1 undergoes polymorphic, tissue-specific, and developmentally regulated allele silencing and that this unveils recessive mutations in patients with core myopathies. Furthermore, our data suggest that imprinting is a likely mechanism for this phenomenon and that similar mechanisms could play a role in human phenotypic heterogeneity.


Asunto(s)
Epigénesis Genética , Miopatía del Núcleo Central/genética , Mutación Puntual , Canal Liberador de Calcio Receptor de Rianodina/genética , Alelos , Animales , Azacitidina/análogos & derivados , Azacitidina/farmacología , Secuencia de Bases , Estudios de Casos y Controles , Células Cultivadas , Islas de CpG , Metilación de ADN , Cartilla de ADN/genética , Decitabina , Femenino , Feto/metabolismo , Silenciador del Gen , Genes Recesivos , Impresión Genómica , Humanos , Ácidos Hidroxámicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Miopatía del Núcleo Central/metabolismo , Linaje , Polimorfismo de Nucleótido Simple , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Distribución Tisular
13.
Hum Mutat ; 27(10): 977-89, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16917943

RESUMEN

The RYR1 gene encodes the skeletal muscle isoform ryanodine receptor and is fundamental to the process of excitation-contraction coupling and skeletal muscle calcium homeostasis. Mapping to chromosome 19q13.2, the gene comprises 106 exons and encodes a protein of 5,038 amino acids. Mutations in the gene have been found in association with several diseases: the pharmacogenetic disorder, malignant hyperthermia (MH); and three congenital myopathies, including central core disease (CCD), multiminicore disease (MmD), and in an isolated case of a congenital myopathy characterized on histology by cores and rods. The majority of gene mutations reported are missense changes identified in cases of MH and CCD. In vitro analysis has confirmed that alteration of normal calcium homeostasis is a functional consequence of some of these changes. Genotype-phenotype correlation studies performed using data from MH and CCD patients have also suggested that mutations may be associated with a range of disease severity phenotypes. This review aims to summarize the current understanding of RYR1 mutations reported in association with MH and CCD and the present viewpoint on the use of mutation data to aid clinical diagnosis of these conditions.


Asunto(s)
Hipertermia Maligna/genética , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Calcio/metabolismo , Genotipo , Humanos , Hipertermia Maligna/metabolismo , Mutación/genética , Miopatía del Núcleo Central/metabolismo , Fenotipo , Canal Liberador de Calcio Receptor de Rianodina/fisiología
14.
Biochem J ; 395(2): 259-66, 2006 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-16372898

RESUMEN

More than 80 mutations in the skeletal muscle ryanodine receptor gene have been found to be associated with autosomal dominant forms of malignant hyperthermia and central core disease, and with recessive forms of multi-minicore disease. Studies on the functional effects of pathogenic dominant mutations have shown that they mostly affect intracellular Ca2+ homoeostasis, either by rendering the channel hypersensitive to activation (malignant hyperthermia) or by altering the amount of Ca2+ released subsequent to physiological or pharmacological activation (central core disease). In the present paper, we show, for the first time, data on the functional effect of two recently identified recessive ryanodine receptor 1 amino acid substitutions, P3527S and V4849I, as well as that of R999H, another substitution that was identified in two siblings that were affected by multi-minicore disease. We studied the intracellular Ca2+ homoeostasis of EBV (Epstein-Barr virus)-transformed lymphoblastoid cells from the affected patients, their healthy relatives and control individuals. Our results show that the P3527S substitution in the homozygous state affected the amount of Ca2+ released after pharmacological activation with 4-chloro-m-cresol and caffeine, but did not affect the size of the thapsigargin-sensitive Ca2+ stores. The other substitutions had no effect on either the size of the intracellular Ca2+ stores, or on the amount of Ca2+ released after ryanodine receptor activation; however, both the P3527S and V4849I substitutions had a small but significant effect on the resting Ca2+ concentration.


Asunto(s)
Sustitución de Aminoácidos/genética , Linfocitos/metabolismo , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Cafeína/farmacología , Señalización del Calcio/efectos de los fármacos , Estudios de Casos y Controles , Células Cultivadas , Cresoles/farmacocinética , Análisis Mutacional de ADN , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Herpesvirus Humano 4/genética , Humanos , Linfocitos/citología , Mutación/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Tapsigargina/farmacología
15.
J Clin Invest ; 115(8): 2033-8, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16075044

RESUMEN

Here we review the current knowledge about the mutations of the gene encoding the cardiac ryanodine receptor (RyR2) that cause cardiac arrhythmias. Similarities between the mutations identified in the RyR2 gene and those found in the gene RyR1 that cause malignant hyperthermia and central core disease are discussed. In vitro functional characterization of RyR1 and RyR2 mutants is reviewed, with a focus on the contribution that in vitro expression studies have made to our understanding of related human diseases.


Asunto(s)
Hipertermia Maligna/genética , Mutación , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Humanos , Hipertermia Maligna/metabolismo , Miopatía del Núcleo Central/metabolismo
16.
Cell Calcium ; 37(2): 121-7, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15589992

RESUMEN

Malignant hyperthermia (MH) and central core disease (CCD) are inherited human disorders of skeletal muscle Ca2+ homeostasis. Both MH and CCD are linked to mutations and/or deletions in the gene encoding the skeletal muscle ryanodine receptor (RyR1), the intracellular Ca2+ release channel, which is essential to excitation-contraction (EC) coupling. Our knowledge on how mutations in RyR1 disrupt intracellular Ca2+ homeostasis and EC coupling, eventually leading to MH and CCD has been recently improved, thanks to multidisciplinary studies ranging from clinical, single channel recordings, patch-clamp experiments, and molecular biology. This review presents a brief historical perspective, on how pioneer studies resulted in associating MH and CCD to RyR1. The review is also focused on discussing novel results in regard to pathophysiological consequences of specific MH/CCD RyR1 mutant proteins, which are representative of the different cellular mechanisms that are linked to either phenotype.


Asunto(s)
Calcio/metabolismo , Hipertermia Maligna/metabolismo , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Humanos
17.
Biophys J ; 87(5): 3193-204, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15347586

RESUMEN

Malignant hyperthermia (MH) and central core disease (CCD) are disorders of skeletal muscle Ca2+ homeostasis that are linked to mutations in the type 1 ryanodine receptor (RyR1). Certain RyR1 mutations result in an MH-selective phenotype (MH-only), whereas others result in a mixed phenotype (MH + CCD). We characterized effects on Ca2+ handling and excitation-contraction (EC) coupling of MH-only and MH + CCD mutations in RyR1 after expression in skeletal myotubes derived from RyR1-null (dyspedic) mice. Compared to wild-type RyR1-expressing myotubes, MH + CCD- and MH-only-expressing myotubes exhibited voltage-gated Ca2+ release (VGCR) that activated at more negative potentials and displayed a significantly higher incidence of spontaneous Ca2+ oscillations. However, maximal VGCR was reduced only for MH + CCD mutants (Y4795C, R2435L, and R2163H) in which spontaneous Ca2+ oscillations occurred with significantly longer duration (Y4795C and R2435L) or higher frequency (R2163H). Notably, myotubes expressing these MH + CCD mutations in RyR1 exhibited both increased [Ca2+]i and reduced sarcoplasmic reticulum (SR) Ca2+ content. We conclude that MH-only mutations modestly increase basal release-channel activity in a manner insufficient to alter net SR Ca2+ content ("compensated leak"), whereas the mixed MH + CCD phenotype arises from mutations that enhance basal activity to a level sufficient to promote SR Ca2+ depletion, elevate [Ca2+]i, and reduce maximal VGCR ("decompensated leak").


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Fiebre/metabolismo , Activación del Canal Iónico , Fibras Musculares Esqueléticas/metabolismo , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Células Cultivadas , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética , Retículo Sarcoplasmático/metabolismo , Relación Estructura-Actividad
18.
Biochem Biophys Res Commun ; 322(4): 1256-66, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15336973

RESUMEN

Ca2+ ions play a pivotal role in a wide array of cellular processes ranging from fertilization to cell death. In skeletal muscle, a mechanical interaction between plasma membrane dihydropyridine receptors (DHPRs, L-type Ca2+ channels) and Ca2+ release channels (ryanodine receptors, RyR1s) of the sarcoplasmic reticulum orchestrates a complex, bi-directional Ca2+ signaling process that converts electrical impulses in the sarcolemma into myoplasmic Ca2+ transients during excitation-contraction coupling. Mutations in the genes that encode the two proteins that coordinate this electrochemical conversion process (the DHPR and RyR1) result in a variety of skeletal muscle disorders including malignant hyperthermia (MH), central core disease (CCD), multiminicore disease, nemaline rod myopathy, and hypokalemic periodic paralysis. Although RyR1 and DHPR disease mutations are thought to alter excitability and Ca2+ homeostasis in skeletal muscle, only recently has research begun to probe the molecular mechanisms by which these genetic defects lead to distinct clinical and histopathological manifestations. This review focuses on recent advances in determining the impact of MH and CCD mutations in RyR1 on muscle Ca2+ signaling and how these effects contribute to disease-specific aspects of these disorders.


Asunto(s)
Señalización del Calcio , Hipertermia Maligna/genética , Hipertermia Maligna/metabolismo , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Calcio/metabolismo , Humanos , Músculo Esquelético/metabolismo , Mutación , Contracción Miocárdica , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
19.
Biochem Biophys Res Commun ; 322(4): 1280-5, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15336975

RESUMEN

Ryanodine receptors (RyR) are the Ca2+ release channels of sarcoplasmic reticulum that provide the majority of the [Ca2+] necessary to induce contraction of cardiac and skeletal muscle cells. In their cellular environment, RyRs are exquisitely regulated by a variety of cytosolic factors and accessory proteins so that their output signal (Ca2+) induces cell contraction without igniting signaling pathways that eventually lead to contractile dysfunction or pathological cellular remodeling. Here we review how dysfunction of RyRs, most commonly expressed as enhanced Ca2+ release at rest (skeletal muscle) or during diastole (cardiac muscle), appears to be the fundamental mechanism underlying several genetic or acquired syndromes. In skeletal muscle, malignant hyperthermia and central core disease result from point mutations in RYR1, the skeletal isoform of RyRs. In cardiac muscle, RYR2 mutations lead to catecholaminergic polymorphic ventricular tachycardia and other cardiac arrhythmias. Lastly, an altered phosphorylation of the RyR2 protein may be involved in some forms of congestive heart failure.


Asunto(s)
Cardiopatías/genética , Cardiopatías/metabolismo , Músculo Esquelético , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Calcio/metabolismo , Insuficiencia Cardíaca , Humanos , Hipertermia Maligna/genética , Hipertermia Maligna/metabolismo , Músculo Esquelético/metabolismo , Mutación , Miocardio/metabolismo , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Taquicardia Ventricular
20.
Neuromuscul Disord ; 12(10): 930-8, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12467748

RESUMEN

Central core disease is a congenital myopathy with muscle weakness defined pathologically by the presence of extensive areas in muscle fibres that are devoid of oxidative enzyme activity. The gene responsible has been shown to be the ryanodine receptor 1 on chromosome 19q13 and mutations have now been identified in several patients. Some cases with the morphological defect remain molecularly undefined, particularly those studied before molecular studies were available. We have studied three families with congenital onset, each with a dominantly inherited mutation in a C-terminal exon of the ryanodine receptor 1. They illustrate the spectrum of pathology that can be observed in patients with the myopathic features of central core disease. We show that extensive fibrosis and fat may be present, type 1 fibre uniformity may occur in the absence of cores; cores may be central or peripheral, single or multiple; and that an appearance of multiple focal minicores might cause a diagnostic pathological dilemma. In addition, we show the value of immunocytochemistry in identifying cores, in particular the use of antibodies to desmin and gamma-filamin.


Asunto(s)
Miopatía del Núcleo Central/patología , Adulto , Biopsia , Niño , Preescolar , Proteínas Contráctiles/metabolismo , Femenino , Filaminas , Ligamiento Genético , Hematoxilina , Humanos , Inmunohistoquímica , Lactante , Masculino , Proteínas de Microfilamentos/metabolismo , Microscopía Electrónica , Mutación , Miopatía del Núcleo Central/metabolismo , Miopatía del Núcleo Central/fisiopatología , NADH Tetrazolio Reductasa/metabolismo , Linaje , Canal Liberador de Calcio Receptor de Rianodina/genética , Cadena B de alfa-Cristalina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA