Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 148
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(30): e2122140119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35867837

RESUMEN

Ryanodine receptors (RyRs) are main regulators of intracellular Ca2+ release and muscle contraction. The Y522S mutation of RyR1 causes central core disease, a weakening myopathy, and malignant hyperthermia, a sudden and potentially fatal response to anesthetics or heat. Y522 is in the core of the N-terminal subdomain C of RyR1 and the mechanism of how this mutation orchestrates malfunction is unpredictable for this 2-MDa ion channel, which has four identical subunits composed of 15 distinct cytoplasmic domains each. We expressed and purified the RyR1 rabbit homolog, Y523S, from HEK293 cells and reconstituted it in nanodiscs under closed and open states. The high-resolution cryogenic electron microscopic (cryo-EM) three-dimensional (3D) structures show that the phenyl ring of Tyr functions in a manner analogous to a "spacer" within an α-helical bundle. Mutation to the much smaller Ser alters the hydrophobic network within the bundle, triggering rearrangement of its α-helices with repercussions in the orientation of most cytoplasmic domains. Examining the mutation-induced readjustments exposed a series of connected α-helices acting as an ∼100 Å-long lever: One end protrudes toward the dihydropyridine receptor, its molecular activator (akin to an antenna), while the other end reaches the Ca2+ activation site. The Y523S mutation elicits channel preactivation in the absence of any activator and full opening at 1.5 µM free Ca2+, increasing by ∼20-fold the potency of Ca2+ to activate the channel compared with RyR1 wild type (WT). This study identified a preactivated pathological state of RyR1 and a long-range lever that may work as a molecular switch to open the channel.


Asunto(s)
Hipertermia Maligna , Músculo Esquelético , Miopatía del Núcleo Central , Canal Liberador de Calcio Receptor de Rianodina , Animales , Calcio/metabolismo , Microscopía por Crioelectrón , Células HEK293 , Humanos , Hipertermia Maligna/genética , Músculo Esquelético/metabolismo , Mutación , Miopatía del Núcleo Central/genética , Conejos , Canal Liberador de Calcio Receptor de Rianodina/química , Canal Liberador de Calcio Receptor de Rianodina/genética
2.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(5): 581-585, 2024 May 10.
Artículo en Zh | MEDLINE | ID: mdl-38684305

RESUMEN

OBJECTIVE: To explore the clinical features and genetic etiology of a child with Central core disease (CCD). METHODS: A child with CCD who was treated at the Children's Hematology Department of the First Affiliated Hospital of Zhengzhou University in February 2022 was selected as the study subject. Muscle biopsy was performed. Peripheral blood samples were collected from the child and his parents for the extraction of genomic DNA. The child was subjected to whole exome sequencing (WES), and candidate variant was verified by Sanger sequencing. RESULTS: The child, a 12-year-old boy, had manifested motor retardation, facial weakness, ptosis, pectus carinatum, scoliosis, etc. Muscle biopsy showed that the central nucleus muscle fibers and atrophic muscle fibers were mainly type I. WES revealed that the child has harbored c.10561G>A (p.G3521S) and c.3448T>C (p.C1150R) compound heterozygous variants of the RYR1 gene. Sanger sequencing confirmed that they were inherited from his mother and father, respectively. Based on the guidelines from the American College of Medical Genetics and Genomics, both variants were considered as likely pathogenic (PS4+PM1+PM2_Supporting+PP3;PM1+PM2_Supporting+PM3+PP3). CONCLUSION: By combining his clinical manifestation and results of muscle pathology and genetic testing, the child was diagnosed with CCD, which may be attributed to the c.10561G>A (p.G3521S) and c.3448T>C (p.C1150R) compound heterozygous variants of the RYR1 gene.


Asunto(s)
Heterocigoto , Miopatía del Núcleo Central , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Canal Liberador de Calcio Receptor de Rianodina/genética , Masculino , Niño , Miopatía del Núcleo Central/genética , Secuenciación del Exoma , Mutación , Pruebas Genéticas
3.
Am J Med Genet A ; 191(6): 1646-1651, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36965156

RESUMEN

Ryanodine receptor type 1-related disorder (RYR1-RD) is the most common subgroup of congenital myopathies with a wide phenotypic spectrum ranging from mild hypotonia to lethal fetal akinesia. Genetic testing for myopathies is imperative as the diagnosis informs counseling regarding prognosis and recurrence risk, treatment options, monitoring, and clinical management. However, diagnostic challenges exist as current options are limited to clinical suspicion prompting testing including: single gene sequencing or familial variant testing, multi-gene panels, exome, genome sequencing, and invasive testing including muscle biopsy. The timing of diagnosis is of great importance due to the association of RYR1-RD with malignant hyperthermia (MH). MH is a hypermetabolic crisis that occurs secondary to excessive calcium release in muscles, leading to systemic effects that can progress to shock and death if unrecognized. Given the association of MH with pathogenic variants in RYR1, a diagnosis of RYR1-RD necessitates an awareness of medical team to avoid potentially triggering agents. We describe a case of a unique fetal presentation with bilateral diaphragmatic eventrations who had respiratory failure, dysmorphic facial features, and profound global hypotonia in the neonatal period. The diagnosis was made at several months of age, had direct implications on her clinical care related to anticipated need to long-term ventilator support, and ultimately death secondary an arrhythmia as a result of suspected MH. Our report reinforces the importance of having high suspicion for a genetic syndrome and pursuing early, rapid exome or genome sequencing as first line testing in critically ill neonatal intensive care unit patients and further evaluating the pathogenicity of a variant of uncertain significance in the setting of a myopathic phenotype.


Asunto(s)
Hipertermia Maligna , Miopatía del Núcleo Central , Femenino , Humanos , Embarazo , Miopatía del Núcleo Central/diagnóstico , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Hipotonía Muscular , Mapeo Cromosómico , Presentación en Trabajo de Parto , Hipertermia Maligna/diagnóstico , Hipertermia Maligna/genética , Mutación
4.
Muscle Nerve ; 63(3): 304-310, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33146414

RESUMEN

BACKGROUND: The diagnosis of uncommon pediatric neuromuscular disease (NMD) is challenging due to genetic and phenotypic heterogeneity, yet is important to guide treatment, prognosis, and recurrence risk. Patients with diagnostically challenging presentations typically undergo extensive testing with variable molecular diagnostic yield. Given the advancement in next generation sequencing (NGS), we investigated the value of clinical whole exome sequencing (ES) in uncommon pediatric NMD. METHODS: A retrospective cohort study of 106 pediatric NMD patients with a combination of ES, chromosomal microarray (CMA), and candidate gene testing was completed at a large tertiary referral center. RESULTS: A molecular diagnosis was achieved in 37/79 (46%) patients with ES, 4/44 (9%) patients with CMA, and 15/74 (20%) patients with candidate gene testing. In 2/79 (3%) patients, a dual molecular diagnosis explaining the neuromuscular disease process was identified. A total of 42 patients (53%) who received ES remained without a molecular diagnosis at the conclusion of the study. CONCLUSIONS: Due to NGS, molecular diagnostic yield of rare neurological diseases is at an all-time high. We show that ES has a higher diagnostic rate compared to other genetic tests in a complex pediatric neuromuscular disease cohort and should be considered early in the diagnostic journey for select NMD patients with challenging presentations in which a clinical diagnosis is not evident.


Asunto(s)
Secuenciación del Exoma , Enfermedades Neuromusculares/diagnóstico , Adolescente , Biopsia , Niño , Preescolar , Estudios de Cohortes , Electromiografía , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Análisis por Micromatrices , Miopatías Mitocondriales/diagnóstico , Miopatías Mitocondriales/genética , Miopatías Mitocondriales/patología , Técnicas de Diagnóstico Molecular , Distrofias Musculares/diagnóstico , Distrofias Musculares/genética , Distrofias Musculares/patología , Distrofia Muscular de Cinturas/diagnóstico , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Miopatía del Núcleo Central/diagnóstico , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/patología , Miositis/diagnóstico , Miositis/genética , Miositis/patología , Conducción Nerviosa , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/patología , Estudios Retrospectivos , Análisis de Secuencia de ADN , Atrofias Musculares Espinales de la Infancia/diagnóstico , Atrofias Musculares Espinales de la Infancia/genética , Atrofias Musculares Espinales de la Infancia/patología , Ataxias Espinocerebelosas/diagnóstico , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología
5.
Muscle Nerve ; 60(1): 80-87, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31004442

RESUMEN

INTRODUCTION: The objective of this study was to obtain a 6-month natural history of motor function performance in individuals with RYR1- related myopathy (RYR1-RM) by using the Motor Function Measure-32 (MFM-32) and graded functional tests (GFT) while facilitating preparation for interventional trials. METHODS: In total, 34 participants completed the MFM-32 and GFTs at baseline and 6-month visits. RESULTS: Motor deficits according to MFM-32 were primarily observed in the standing and transfers domain (D1; mean 71%). Among the GFTs, participants required the most time to ascend/descend stairs (>7.5 s). Functional movement, determined by GFT grades, was strongly correlated with MFM-32 (D1; r ≥ 0.770, P < 0.001). Motor Function Measure-32 and GFT scores did not reflect any change in performance between baseline and 6-month visits. DISCUSSION: The MFM-32 and GFTs detected motor impairment in RYR1-RM, which remained stable over 6 months. Thus, these measures may be suitable for assessing change in motor function in response to therapeutic intervention. Muscle Nerve 60: 80-87, 2019.


Asunto(s)
Movimiento/fisiología , Miopatías Estructurales Congénitas/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/genética , Adolescente , Adulto , Niño , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miopatías Estructurales Congénitas/genética , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/fisiopatología , Oftalmoplejía/genética , Oftalmoplejía/fisiopatología , Canal Liberador de Calcio Receptor de Rianodina/deficiencia , Adulto Joven
6.
Muscle Nerve ; 58(2): 235-244, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29669168

RESUMEN

INTRODUCTION: Congenital myopathies are muscle diseases characterized by specific histopathologic features, generalized hypotonia from birth, and perinatal complications, although some cases develop during childhood or, rarely, in adulthood. We undertook this study to characterize congenital myopathies among patients registered at our institution. METHODS: Clinical, histopathologic, and genetic features were evaluated in 34 patients recruited for this study. RESULTS: The majority of patients experienced a childhood onset, and no disease-related mortality was recorded during follow-up. Functional outcomes were no better for those with late-onset disease, indicating later disease progression can be significant. Nemaline myopathy was the most frequent pathology, followed by central core disease and centronuclear myopathy. Among the 18 (54.5%) genetically confirmed patients, NEB and RYR1 mutations were the most common, followed by DNM2 mutations. DISCUSSION: This study shows features not previously reported and suggests that congenital myopathy should be considered an important issue among adult patients. Muscle Nerve 58: 235-244, 2018.


Asunto(s)
Miotonía Congénita/patología , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Dinamina II , Dinaminas/genética , Femenino , Humanos , Lactante , Masculino , Fibras Musculares Esqueléticas/patología , Proteínas Musculares/genética , Mutación , Miopatías Nemalínicas/genética , Miopatías Nemalínicas/patología , Miopatías Estructurales Congénitas/congénito , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Miopatía del Núcleo Central/congénito , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/patología , Miotonía Congénita/genética , República de Corea , Estudios Retrospectivos , Canal Liberador de Calcio Receptor de Rianodina/genética , Resultado del Tratamiento , Adulto Joven
7.
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
8.
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
9.
Hum Mol Genet ; 23(4): 980-91, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24105469

RESUMEN

Core myopathies (CM), the main non-dystrophic myopathies in childhood, remain genetically unexplained in many cases. Heart disease is not considered part of the typical CM spectrum. No congenital heart defect has been reported, and childhood-onset cardiomyopathy has been documented in only two CM families with homozygous mutations of the TTN gene. TTN encodes titin, a giant protein of striated muscles. Recently, heterozygous TTN truncating mutations have also been reported as a major cause of dominant dilated cardiomyopathy. However, relatively few TTN mutations and phenotypes are known, and titin pathophysiological role in cardiac and skeletal muscle conditions is incompletely understood. We analyzed a series of 23 families with congenital CM and primary heart disease using TTN M-line-targeted sequencing followed in selected patients by whole-exome sequencing and functional studies. We identified seven novel homozygous or compound heterozygous TTN mutations (five in the M-line, five truncating) in 17% patients. Heterozygous parents were healthy. Phenotype analysis identified four novel titinopathies, including cardiac septal defects, left ventricular non-compaction, Emery-Dreifuss muscular dystrophy or arthrogryposis. Additionally, in vitro studies documented the first-reported absence of a functional titin kinase domain in humans, leading to a severe antenatal phenotype. We establish that CM are associated with a large range of heart conditions of which TTN mutations are a major cause, thereby expanding the TTN mutational and phenotypic spectrum. Additionally, our results suggest titin kinase implication in cardiac morphogenesis and demonstrate that heterozygous TTN truncating mutations may not manifest unless associated with a second mutation, reassessing the paradigm of their dominant expression.


Asunto(s)
Codón sin Sentido , Conectina/genética , Cardiopatías/genética , Miopatía del Núcleo Central/genética , Adolescente , Conectina/metabolismo , Consanguinidad , Femenino , Genes Recesivos , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Cardiopatías/metabolismo , Cardiopatías/patología , Heterocigoto , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Miopatía del Núcleo Central/metabolismo , Miopatía del Núcleo Central/patología , Linaje , Fenotipo , Adulto Joven
10.
Muscle Nerve ; 54(3): 432-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26799446

RESUMEN

INTRODUCTION: Ryanodine receptor 1 (RYR1), myosin heavy chain 7 (MYH7), and selenoprotein N1 (SEPN1) mutations are associated with core myopathies. RYR1 mutations cause most cases of central core disease (CCD). METHODS: We screened 8 Chinese patients with clinicopathological diagnosis of CCD. Genetic analysis was carried out by targeted next generation sequencing (NGS) to identify causative genes. Variants were assessed for pathogenicity using bioinformatic approaches, and NGS results were confirmed by Sanger sequencing. RESULTS: One novel (p.L4578V) and heterozygous missense mutations in RYR1 were identified in 7 patients. Two patients carried a novel mutation, 1 had p.M4640R, 3 had p.R4861H, and 1 had p.R4861C. All patients had mild to moderate severity phenotypes. Histopathological findings demonstrated central cores and type I fiber predominance. CONCLUSIONS: NGS is an efficient strategy to identify variants in RYR1 in CCD. However, genetic results revealed by NGS must be combined with clinicopathologic features to validate the diagnosis. Muscle Nerve, 2016 Muscle Nerve 54: 432-438, 2016.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Mutación/genética , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Adolescente , Adulto , Pueblo Asiatico/genética , Niño , Preescolar , Análisis Mutacional de ADN , Electromiografía , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Microscopía Electrónica de Transmisión , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Miopatía del Núcleo Central/patología , Adulto Joven
11.
Proc Natl Acad Sci U S A ; 109(2): 610-5, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22203976

RESUMEN

The type 1 ryanodine receptor (RyR1) is expressed widely in the brain, with high levels in the cerebellum, hippocampus, and hypothalamus. We have shown that L-type Ca(2+) channels in terminals of hypothalamic magnocellular neurons are coupled to RyRs, as they are in skeletal muscle, allowing voltage-induced Ca(2+) release (VICaR) from internal Ca(2+) stores without Ca(2+) influx. Here we demonstrate that RyR1 plays a role in VICaR in nerve terminals. Furthermore, in heterozygotes from the Ryr1(I4895T/WT) (IT/+) mouse line, carrying a knock-in mutation corresponding to one that causes a severe form of human central core disease, VICaR is absent, demonstrating that type 1 RyR mediates VICaR and that these mice have a neuronal phenotype. The absence of VICaR was shown in two ways: first, depolarization in the absence of Ca(2+) influx elicited Ca(2+)syntillas (scintilla, spark, in a nerve terminal, a SYNaptic structure) in WT, but not in mutant terminals; second, in the presence of extracellular Ca(2+), IT/+ terminals showed a twofold decrease in global Ca(2+) transients, with no change in plasmalemmal Ca(2+) current. From these studies we draw two conclusions: (i) RyR1 plays a role in VICaR in hypothalamic nerve terminals; and (ii) a neuronal alteration accompanies the myopathy in IT/+ mice, and, possibly in humans carrying the corresponding RyR1 mutation.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Calcio/metabolismo , Hipotálamo/citología , Miopatía del Núcleo Central/genética , Neuronas/metabolismo , Terminales Presinápticos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Animales , Fluorescencia , Técnicas de Sustitución del Gen , Hipotálamo/metabolismo , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
12.
Biochemistry ; 53(5): 932-46, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24447242

RESUMEN

Ryanodine receptors (RyRs) are large ion channels that are responsible for the release of Ca(2+) from the sarcoplasmic/endoplasmic reticulum. Calmodulin (CaM) is a Ca(2+) binding protein that can affect the channel open probability at both high and low Ca(2+) concentrations, shifting the Ca(2+) dependencies of channel opening in an isoform-specific manner. Here we analyze the binding of CaM and its individual domains to three different RyR regions using isothermal titration calorimetry. We compared binding to skeletal muscle (RyR1) and cardiac (RyR2) isoforms, under both Ca(2+)-loaded and Ca(2+)-free conditions. CaM can bind all three regions in both isoforms, but the binding modes differ appreciably in two segments. The results highlight a Ca(2+)/CaM and apoCaM binding site in the C-terminal fifth of the channel. This binding site is the target for malignant hyperthermia and central core disease mutations in RyR1, which affect the energetics and mode of CaM binding.


Asunto(s)
Calmodulina/química , Canal Liberador de Calcio Receptor de Rianodina/química , Animales , Sitios de Unión , Calmodulina/genética , Humanos , Hipertermia Maligna/genética , Músculo Esquelético/metabolismo , Mutación , Miocardio/metabolismo , Miopatía del Núcleo Central/genética , Unión Proteica , Isoformas de Proteínas/química , Conejos , Ratas , Termodinámica
13.
J Neurol Neurosurg Psychiatry ; 85(10): 1149-52, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24828896

RESUMEN

BACKGROUND: Autosomal dominant (AD) central core disease (CCD) is a congenital myopathy characterised by the presence of cores in the muscle fibres which correspond to broad areas of myofibrils disorganisation, Z-line streaming and lack of mitochondria. Heterozygous mutations in the RYR1 gene were observed in the large majority of AD-CCD families; however, this gene was excluded in some of AD-CCD families. OBJECTIVE: To enlarge the genetic spectrum of AD-CCD demonstrating mutations in an additional gene. PATIENTS AND METHODS: Four affected AD family members over three generations, three of whom were alive and participate in the study: the mother and two of three siblings. The symptoms began during the early childhood with mild delayed motor development. Later they developed mainly tibialis anterior weakness, hypertrophy of calves and significant weakness (amyotrophic) of quadriceps. No cardiac or ocular involvement was noted. RESULTS: The muscle biopsies sections showed a particular pattern: eccentric cores in type 1 fibres, associated with type 1 predominance. Most cores have abrupt borders. Electron microscopy confirmed the presence of both unstructured and structured cores. Exome sequencing analysis identified a novel heterozygous missense mutation p.Leu1723Pro in MYH7 segregating with the disease and affecting a conserved residue in the myosin tail domain. CONCLUSIONS: We describe MYH7 as an additional causative gene for AD-CCD. These findings have important implications for diagnosis and future investigations of AD-congenital myopathies with cores, without cardiomyopathy, but presenting a particular involvement of distal and quadriceps muscles.


Asunto(s)
Miosinas Cardíacas/genética , Predisposición Genética a la Enfermedad/genética , Mutación Missense/genética , Miopatía del Núcleo Central/genética , Cadenas Pesadas de Miosina/genética , Adulto , Anciano , Femenino , Heterocigoto , Humanos , Masculino , Fibras Musculares de Contracción Lenta/diagnóstico por imagen , Fibras Musculares de Contracción Lenta/patología , Fibras Musculares de Contracción Lenta/ultraestructura , Miopatía del Núcleo Central/diagnóstico por imagen , Miopatía del Núcleo Central/patología , Linaje , Radiografía
14.
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
15.
Beijing Da Xue Xue Bao Yi Xue Ban ; 46(5): 691-7, 2014 Oct 18.
Artículo en Zh | MEDLINE | ID: mdl-25331388

RESUMEN

OBJECTIVE: To detect hot spot mutation of RYR1 gene in 15 cases of congenital myopathy with different subtypes, and to discuss the value of RYR1 gene hot spot mutation detection in the diagnosis of the disease. METHODS: Clinical data were collected in all the patients, including clinical manifestations and signs, serum creatine kinase, electromyography. Fourteen of the patients accepted the muscle biopsy. Hot spot mutation in the C-terminal of RYR1 gene (extron 96-106) had been detected in all the 15 patients. RESULTS: All the patients presented with motor development delay, and they could walk at the age of 1 to 3.5 years,but were always easy to fall and could not run or jump. There were no progressive deteriorations. Physical examination showed different degrees of muscle weakness and hypotonia.High arched palates were noted in 3 patients. The serum levels of creatine kinase were mildly elevated in 3 cases, and normal in 12 cases. Electromyography showed "myogenic" features in 11 patients, being normal in the other 4 patients. Muscle biopsy pathologic diagnosis was the central core disease in 3 patients, the central nuclei in 2 patients, the congenital fiber type disproportion in 2 patients, the nameline myopathy in 3 patient, the multiminicore disease in 1 patient, and nonspecific minimal changes in the other 3 patients; one patient was diagnosed with central core disease according to positive family history and gene mutation. In the family case (Patient 2) of central core disease, the c.14678G>A (p.Arg4893Gln) mutation in 102 extron of RYR1 was identified in three members of the family, which had been reported to be a pathogenic mutation. The c.14596A>G(p.Lys4866Gln) mutation in 101 extron was found in one patient with central core disease(Patient 1), and the c.14719G>A(p.Gly4907Ser) mutation in 102 extron was found in another case of the central core disease(Patient 3).The same novel mutation was verified in one of the patients' (Patient 3) asymptomatic father. CONCLUSION: Congenital myopathies in the different subtype have the similar clinical manifestations, signs, enzyme detection and electromyography changes. Muscle biopsy plays an important role in the selection of genes to be detected. Hot spot mutation in C-terminal of the RYR1 gene can only be identified in patients with central core disease, so we suggest this hot spot gene mutation screening apply to the suspicious patient with central core disease only.


Asunto(s)
Miopatías Estructurales Congénitas/genética , Miopatía del Núcleo Central/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Biopsia , Análisis Mutacional de ADN , Electromiografía , Humanos , Mutación , Linaje
16.
Stem Cell Res ; 77: 103411, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582058

RESUMEN

RYR1 variants are a common cause of congenital myopathies, including multi-minicore disease (MmD) and central core disease (CCD). Here, we generated iPSC lines from two CCD patients with dominant RYR1 missense variants that affect the transmembrane (pore) and SPRY3 protein domains (p.His4813Tyr and p.Asn1346Lys, respectively). Both lines had typical iPSC morphology, expressed canonical pluripotency markers, exhibited trilineage differentiation potential, and had normal karyotypes. Together with existing RYR1 iPSC lines, these represent important tools to study and develop treatments for RYR1-related myopathies.


Asunto(s)
Células Madre Pluripotentes Inducidas , Mutación Missense , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/patología , Miopatía del Núcleo Central/metabolismo , Adulto , Línea Celular , Masculino , Diferenciación Celular , Femenino
17.
Stem Cell Res ; 77: 103410, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38583293

RESUMEN

RYR1 variants are the most common genetic cause of congenital myopathies, and typically cause central core disease (CCD) and/or malignant hyperthermia (MH). Here, we generated iPSC lines from two patients with CCD and MH caused by dominant RYR1 variants within the central region of the protein (p.Val2168Met and p.Arg2508Cys). Both lines displayed typical iPSC morphology, uniform expression of pluripotency markers, trilineage differentiation potential, and had normal karyotypes. These are the first RYR1 iPSC lines from patients with both CCD and MH. As these are common CCD/MH variants, these lines should be useful to study these conditions and test therapeutics.


Asunto(s)
Células Madre Pluripotentes Inducidas , Hipertermia Maligna , Mutación Missense , Canal Liberador de Calcio Receptor de Rianodina , Humanos , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Hipertermia Maligna/genética , Células Madre Pluripotentes Inducidas/metabolismo , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/patología , Masculino , Femenino , Línea Celular , Diferenciación Celular
18.
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
19.
Anesth Analg ; 116(2): 420-3, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23267001

RESUMEN

A healthy 6-year-old boy developed lower extremity rigidity, trismus, and fever after playing in a splash pool. On arrival in the emergency department, he appeared to be seizing. An endotracheal tube was emergently placed using succinylcholine. Cardiac arrest followed. He could not be resuscitated. Postmortem genetic analysis found a novel RYR1 variant. Family testing revealed the same variant in his father who also had muscle contracture testing diagnostic for susceptibility to malignant hyperthermia and central core disease diagnosed histologically. Because there was no exposure to volatile anesthetics before the onset of symptoms, this is a case of "awake" malignant hyperthermia worsened by succinylcholine.


Asunto(s)
Servicios Médicos de Urgencia , Servicio de Urgencia en Hospital , Hipertermia Maligna/fisiopatología , Niño , Diazepam/efectos adversos , Resultado Fatal , Humanos , Hipnóticos y Sedantes/efectos adversos , Intubación Intratraqueal , Hígado/química , Lorazepam , Masculino , Hipertermia Maligna/patología , Relajantes Musculares Centrales/efectos adversos , Rigidez Muscular/inducido químicamente , Miopatía del Núcleo Central/genética , Fármacos Neuromusculares Despolarizantes/efectos adversos , Síndrome de Dificultad Respiratoria/inducido químicamente , Canal Liberador de Calcio Receptor de Rianodina/genética , Succinilcolina/efectos adversos
20.
Paediatr Anaesth ; 23(9): 834-41, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23617272

RESUMEN

The core myopathies are a subset of myopathies that present in infancy with hypotonia and muscle weakness. They were formerly considered a rare type of congenital myopathy but are now recognized as being more prevalent. Due to their genetic linkage to mutations in the ryanodine receptor gene (RYR1), core myopathies (in particular, central core disease) carry a high risk of malignant hyperthermia susceptibility. In this review article, we describe the phenotypical, genetic, and histopathological characteristics of core myopathies and further describe the currently understood nature of their risk of malignant hyperthermia. We also review the level of suspicion a clinician should exhibit with a child who has a possible core myopathy or other congenital myopathy presenting for an anesthetic prior to a definitive genetic analysis. For this review article, we performed literature searches using the key words anesthesiology, core myopathies, pediatric neurology, malignant hyperthermia, genetics, ryanodine receptor, and molecular biology. We also relied on literature accumulated by the two authors, who served as hotline consultants for the Malignant Hyperthermia Hotline of the Malignant Hyperthermia Association of the United States (MHAUS) for the past 12 years.


Asunto(s)
Hipertermia Maligna/fisiopatología , Miopatía del Núcleo Central/fisiopatología , Anestesia , Anestésicos/efectos adversos , Niño , Susceptibilidad a Enfermedades , Humanos , Hipertermia Maligna/complicaciones , Hipertermia Maligna/genética , Miopatía del Núcleo Central/complicaciones , Miopatía del Núcleo Central/genética , Planificación de Atención al Paciente , Canal Liberador de Calcio Receptor de Rianodina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA