Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.970
Filtrar
1.
Int Heart J ; 65(3): 580-585, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38825499

RESUMO

Cardiac ryanodine receptor (RyR2) gain-of-function mutations cause catecholaminergic polymorphic ventricular tachycardia (CPVT). Conversely, RyR2 loss-of-function mutations cause a new disease entity, termed calcium release deficiency syndrome (CRDS), which may include RYR2-related long QT syndrome (LQTS). Importantly, unlike CPVT, patients with CRDS do not always exhibit exercise- or epinephrine-induced ventricular arrhythmias, which precludes a diagnosis of CRDS. Here we report a boy and his father, who both experienced exercise-induced cardiac events and harbor the same RYR2 E4107A variant. In the boy, an exercise stress test (EST) and epinephrine provocation test (EPT) did not induce any ventricular arrhythmias. QTc was slightly prolonged (QTc: 474 ms), and an EPT induced QTc prolongation (QTc-baseline: 466 ms, peak: 532 ms, steady-state: 527 ms). In contrast, in his father, QTc was not prolonged (QTc: 417 ms), and neither an EST nor EPT induced QTc prolongation. However, an EST induced multifocal premature ventricular contraction (PVC) bigeminy and bidirectional PVC couplets. Thus, they exhibited distinct clinical phenotypes: the boy exhibited LQTS (or CRDS) phenotype, whereas his father exhibited CPVT phenotype. These findings suggest that, in addition to the altered RyR2 function, other unidentified factors, such as other genetic, epigenetic, and environmental factors, and aging, may be involved in the diverse phenotypic manifestations. Considering that a single RYR2 variant can cause both CPVT and LQTS (or CRDS) phenotypes, in cascade screening of patients with CPVT and CRDS, an EST and EPT are not sufficient and genetic analysis is required to identify individuals who are at increased risk for life-threatening arrhythmias.


Assuntos
Síndrome do QT Longo , Fenótipo , Canal de Liberação de Cálcio do Receptor de Rianodina , Taquicardia Ventricular , Humanos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Masculino , Síndrome do QT Longo/genética , Síndrome do QT Longo/diagnóstico , Taquicardia Ventricular/genética , Taquicardia Ventricular/diagnóstico , Eletrocardiografia , Linhagem , Adulto , Teste de Esforço , Mutação
2.
J Am Heart Assoc ; 13(12): e033733, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38860414

RESUMO

BACKGROUND: Chronic sympathetic stimulation drives desensitization and downregulation of ß1 adrenergic receptor (ß1AR) in heart failure. We aim to explore the differential downregulation subcellular pools of ß1AR signaling in the heart. METHODS AND RESULTS: We applied chronic infusion of isoproterenol to induced cardiomyopathy in male C57BL/6J mice. We applied confocal and proximity ligation assay to examine ß1AR association with L-type calcium channel, ryanodine receptor 2, and SERCA2a ((Sarco)endoplasmic reticulum calcium ATPase 2a) and Förster resonance energy transfer-based biosensors to probe subcellular ß1AR-PKA (protein kinase A) signaling in ventricular myocytes. Chronic infusion of isoproterenol led to reduced ß1AR protein levels, receptor association with L-type calcium channel and ryanodine receptor 2 measured by proximity ligation (puncta/cell, 29.65 saline versus 14.17 isoproterenol, P<0.05), and receptor-induced PKA signaling at the plasma membrane (Förster resonance energy transfer, 28.9% saline versus 1.9% isoproterenol, P<0.05) and ryanodine receptor 2 complex (Förster resonance energy transfer, 30.2% saline versus 10.6% isoproterenol, P<0.05). However, the ß1AR association with SERCA2a was enhanced (puncta/cell, 51.4 saline versus 87.5 isoproterenol, P<0.05), and the receptor signal was minimally affected. The isoproterenol-infused hearts displayed decreased PDE4D (phosphodiesterase 4D) and PDE3A and increased PDE2A, PDE4A, and PDE4B protein levels. We observed a reduced role of PDE4 and enhanced roles of PDE2 and PDE3 on the ß1AR-PKA activity at the ryanodine receptor 2 complexes and myocyte shortening. Despite the enhanced ß1AR association with SERCA2a, the endogenous norepinephrine-induced signaling was reduced at the SERCA2a complexes. Inhibiting monoamine oxidase A rescued the norepinephrine-induced PKA signaling at the SERCA2a and myocyte shortening. CONCLUSIONS: This study reveals distinct mechanisms for the downregulation of subcellular ß1AR signaling in the heart under chronic adrenergic stimulation.


Assuntos
Canais de Cálcio Tipo L , Proteínas Quinases Dependentes de AMP Cíclico , Regulação para Baixo , Isoproterenol , Camundongos Endogâmicos C57BL , Miócitos Cardíacos , Receptores Adrenérgicos beta 1 , Canal de Liberação de Cálcio do Receptor de Rianodina , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático , Transdução de Sinais , Animais , Receptores Adrenérgicos beta 1/metabolismo , Masculino , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Isoproterenol/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/efeitos dos fármacos , Modelos Animais de Doenças , Camundongos , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/fisiopatologia , Cardiomiopatias/metabolismo , Cardiomiopatias/induzido quimicamente , Transferência Ressonante de Energia de Fluorescência
3.
Nat Commun ; 15(1): 5120, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38879623

RESUMO

Calmodulin transduces [Ca2+] information regulating the rhythmic Ca2+ cycling between the sarcoplasmic reticulum and cytoplasm during contraction and relaxation in cardiac and skeletal muscle. However, the structural dynamics by which calmodulin modulates the sarcoplasmic reticulum Ca2+ release channel, the ryanodine receptor, at physiologically relevant [Ca2+] is unknown. Using fluorescence lifetime FRET, we resolve different structural states of calmodulin and Ca2+-driven shifts in the conformation of calmodulin bound to ryanodine receptor. Skeletal and cardiac ryanodine receptor isoforms show different calmodulin-ryanodine receptor conformations, as well as binding and structural kinetics with 0.2-ms resolution, which reflect different functional roles of calmodulin. These FRET methods provide insight into the physiological calmodulin-ryanodine receptor structural states, revealing additional distinct structural states that complement cryo-EM models that are based on less physiological conditions. This technology will drive future studies on pathological calmodulin-ryanodine receptor interactions and dynamics with other important ryanodine receptor bound modulators.


Assuntos
Cálcio , Calmodulina , Transferência Ressonante de Energia de Fluorescência , Músculo Esquelético , Miocárdio , Canal de Liberação de Cálcio do Receptor de Rianodina , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Calmodulina/metabolismo , Calmodulina/química , Cálcio/metabolismo , Miocárdio/metabolismo , Cinética , Animais , Músculo Esquelético/metabolismo , Humanos , Conformação Proteica , Ligação Proteica , Retículo Sarcoplasmático/metabolismo
4.
J Cachexia Sarcopenia Muscle ; 15(3): 1003-1015, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38725372

RESUMO

BACKGROUND: Autosomal-recessive mutations in SPEG (striated muscle preferentially expressed protein kinase) have been linked to centronuclear myopathy with or without dilated cardiomyopathy (CNM5). Loss of SPEG is associated with defective triad formation, abnormal excitation-contraction coupling, calcium mishandling and disruption of the focal adhesion complex in skeletal muscles. To elucidate the underlying molecular pathways, we have utilized multi-omics tools and analysis to obtain a comprehensive view of the complex biological processes and molecular functions. METHODS: Skeletal muscles from 2-month-old SPEG-deficient (Speg-CKO) and wild-type (WT) mice were used for RNA sequencing (n = 4 per genotype) to profile transcriptomics and mass spectrometry (n = 4 for WT; n = 3 for Speg-CKO mice) to profile proteomics and phosphoproteomics. In addition, interactomics was performed using the SPEG antibody on pooled muscle lysates (quadriceps, gastrocnemius and triceps) from WT and Speg-CKO mice. Based on the multi-omics results, we performed quantitative real-time PCR, co-immunoprecipitation and immunoblot to verify the findings. RESULTS: We identified that SPEG interacts with myospryn complex proteins CMYA5, FSD2 and RyR1, which are critical for triad formation, and that SPEG deficiency results in myospryn complex abnormalities (protein levels decreased to 22 ± 3% for CMYA5 [P < 0.05] and 18 ± 3% for FSD2 [P < 0.01]). Furthermore, SPEG phosphorylates RyR1 at S2902 (phosphorylation level decreased to 55 ± 15% at S2902 in Speg-CKO mice; P < 0.05), and its loss affects JPH2 phosphorylation at multiple sites (increased phosphorylation at T161 [1.90 ± 0.24-fold], S162 [1.61 ± 0.37-fold] and S165 [1.66 ± 0.13-fold]; decreased phosphorylation at S228 and S231 [39 ± 6%], S234 [50 ± 12%], S593 [48 ± 3%] and S613 [66 ± 10%]; P < 0.05 for S162 and P < 0.01 for other sites). On analysing the transcriptome, the most dysregulated pathways affected by SPEG deficiency included extracellular matrix-receptor interaction (P < 1e-15) and peroxisome proliferator-activated receptor signalling (P < 9e-14). CONCLUSIONS: We have elucidated the critical role of SPEG in the triad as it works closely with myospryn complex proteins (CMYA5, FSD2 and RyR1), it regulates phosphorylation levels of various residues in JPH2 and S2902 in RyR1, and its deficiency is associated with dysregulation of several pathways. The study identifies unique SPEG-interacting proteins and their phosphorylation functions and emphasizes the importance of using a multi-omics approach to comprehensively evaluate the molecular function of proteins involved in various genetic disorders.


Assuntos
Camundongos Knockout , Proteínas Musculares , Músculo Esquelético , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Camundongos , Músculo Esquelético/metabolismo , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Proteômica/métodos , Fosforilação , Multiômica , Quinase de Cadeia Leve de Miosina
5.
Biochem Biophys Res Commun ; 720: 150105, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38754163

RESUMO

BACKGROUND: Dexmedetomidine (DEX), a highly selective α2-adrenoceptor agonist, can decrease the incidence of arrhythmias, such as catecholaminergic polymorphic ventricular tachycardia (CPVT). However, the underlying mechanisms by which DEX affects cardiac electrophysiological function remain unclear. METHODS: Ryanodine receptor (RyR2) heterozygous R2474S mice were used as a model for CPVT. WT and RyR2R2474S/+ mice were treated with isoproterenol (ISO) and DEX, and electrocardiograms were continuously monitored during both in vivo and ex vivo experiments. Dual-dye optical mapping was used to explore the anti-arrhythmic mechanism of DEX. RESULTS: DEX significantly reduced the occurrence and duration of ISO-induced of VT/VF in RyR2R2474S/+ mice in vivo and ex vivo. DEX remarkably prolonged action potential duration (APD80) and calcium transient duration (CaTD80) in both RyR2R2474S/+ and WT hearts, whereas it reduced APD heterogeneity and CaT alternans in RyR2R2474S/+ hearts. DEX inhibited ectopy and reentry formation, and stabilized voltage-calcium latency. CONCLUSION: DEX exhibited an antiarrhythmic effect through stabilizing membrane voltage and intracellular Ca2+. DEX can be used as a beneficial perioperative anesthetic for patients with CPVT or other tachy-arrhythmias.


Assuntos
Arritmias Cardíacas , Cálcio , Dexmedetomidina , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Dexmedetomidina/farmacologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Cálcio/metabolismo , Camundongos , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/genética , Potenciais da Membrana/efeitos dos fármacos , Isoproterenol/farmacologia , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/tratamento farmacológico , Antiarrítmicos/farmacologia , Masculino , Potenciais de Ação/efeitos dos fármacos , Camundongos Endogâmicos C57BL
7.
Nat Commun ; 15(1): 4115, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38750013

RESUMO

RyR1 is an intracellular Ca2+ channel important in excitable cells such as neurons and muscle fibers. Ca2+ activates it at low concentrations and inhibits it at high concentrations. Mg2+ is the main physiological RyR1 inhibitor, an effect that is overridden upon activation. Despite the significance of Mg2+-mediated inhibition, the molecular-level mechanisms remain unclear. In this work we determined two cryo-EM structures of RyR1 with Mg2+ up to 2.8 Å resolution, identifying multiple Mg2+ binding sites. Mg2+ inhibits at the known Ca2+ activating site and we propose that the EF hand domain is an inhibitory divalent cation sensor. Both divalent cations bind to ATP within a crevice, contributing to the precise transmission of allosteric changes within the enormous channel protein. Notably, Mg2+ inhibits RyR1 by interacting with the gating helices as validated by molecular dynamics. This structural insight enhances our understanding of how Mg2+ inhibition is overcome during excitation.


Assuntos
Cálcio , Microscopia Crioeletrônica , Magnésio , Canal de Liberação de Cálcio do Receptor de Rianodina , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Magnésio/metabolismo , Cálcio/metabolismo , Sítios de Ligação , Animais , Simulação de Dinâmica Molecular , Trifosfato de Adenosina/metabolismo , Humanos , Coelhos
8.
J Phys Chem B ; 128(19): 4670-4684, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38717304

RESUMO

Ryanodine receptor type 1 (RyR1) is a Ca2+-release channel central to skeletal muscle excitation-contraction (EC) coupling. RyR1's cryo-EM structures reveal a zinc-finger motif positioned within the cytoplasmic C-terminal domain (CTD). Yet, owing to limitations in cryo-EM resolution, RyR1 structures lack precision in detailing the metal coordination structure, prompting the need for an accurate model. In this study, we employed molecular dynamics (MD) simulations and the density functional theory (DFT) method to refine the binding characteristics of Zn2+ in the zinc-finger site of the RyR1 channel. Our findings also highlight substantial conformational changes in simulations conducted in the absence of Zn2+. Notably, we observed a loss of contact at the interface between protein domains proximal to the zinc-finger site, indicating a crucial role of Zn2+ in maintaining structural integrity and interdomain interactions within RyR1. Furthermore, this study provides valuable insights into the modulation of ATP, Ca2+, and caffeine binding, shedding light on the intricate relationship between Zn2+ coordination and the dynamic behavior of RyR1. Our integrative approach combining MD simulations and DFT calculations enhances our understanding of the molecular mechanisms governing ligand binding in RyR1.


Assuntos
Simulação de Dinâmica Molecular , Canal de Liberação de Cálcio do Receptor de Rianodina , Zinco , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Zinco/química , Zinco/metabolismo , Ligantes , Cálcio/química , Cálcio/metabolismo , Teoria da Densidade Funcional , Sítios de Ligação , Ligação Proteica , Dedos de Zinco , Cafeína/química , Cafeína/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Humanos
9.
Biochem Biophys Res Commun ; 723: 150163, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-38820626

RESUMO

Excitation-contraction coupling in skeletal muscle myofibers depends upon Ca2+ release from the sarcoplasmic reticulum through the ryanodine receptor/Ca2+-release channel RyR1. The RyR1 contains ∼100 Cys thiols of which ∼30 comprise an allosteric network subject to posttranslational modification by S-nitrosylation, S-palmitoylation and S-oxidation. However, the role and function of these modifications is not understood. Although aberrant S-nitrosylation of multiple unidentified sites has been associated with dystrophic diseases, malignant hyperthermia and other myopathic syndromes, S-nitrosylation in physiological situations is reportedly specific to a single (1 of ∼100) Cys in RyR1, Cys3636 in a manner gated by pO2. Using mice expressing a form of RyR1 with a Cys3636→Ala point mutation to prevent S-nitrosylation at this site, we showed that Cys3636 was the principal target of endogenous S-nitrosylation during normal muscle function. The absence of Cys3636 S-nitrosylation suppressed stimulus-evoked Ca2+ release at physiological pO2 (at least in part by altering the regulation of RyR1 by Ca2+/calmodulin), eliminated pO2 coupling, and diminished skeletal myocyte contractility in vitro and measures of muscle strength in vivo. Furthermore, we found that abrogation of Cys3636 S-nitrosylation resulted in a developmental defect reflected in diminished myofiber diameter, altered fiber subtypes, and altered expression of genes implicated in muscle development and atrophy. Thus, our findings establish a physiological role for pO2-coupled S-nitrosylation of RyR1 in skeletal muscle contractility and development and provide foundation for future studies of RyR1 modifications in physiology and disease.


Assuntos
Músculo Esquelético , Canal de Liberação de Cálcio do Receptor de Rianodina , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Animais , Músculo Esquelético/metabolismo , Camundongos , Cálcio/metabolismo , Cisteína/metabolismo , Processamento de Proteína Pós-Traducional , Desenvolvimento Muscular , Camundongos Transgênicos , Sinalização do Cálcio
10.
Nat Commun ; 15(1): 3528, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664444

RESUMO

Cardiac dysfunction is a hallmark of aging in humans and mice. Here we report that a two-week treatment to restore youthful Bridging Integrator 1 (BIN1) levels in the hearts of 24-month-old mice rejuvenates cardiac function and substantially reverses the aging phenotype. Our data indicate that age-associated overexpression of BIN1 occurs alongside dysregulated endosomal recycling and disrupted trafficking of cardiac CaV1.2 and type 2 ryanodine receptors. These deficiencies affect channel function at rest and their upregulation during acute stress. In vivo echocardiography reveals reduced systolic function in old mice. BIN1 knockdown using an adeno-associated virus serotype 9 packaged shRNA-mBIN1 restores the nanoscale distribution and clustering plasticity of ryanodine receptors and recovers Ca2+ transient amplitudes and cardiac systolic function toward youthful levels. Enhanced systolic function correlates with increased phosphorylation of the myofilament protein cardiac myosin binding protein-C. These results reveal BIN1 knockdown as a novel therapeutic strategy to rejuvenate the aging myocardium.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Envelhecimento , Miocárdio , Proteínas do Tecido Nervoso , Canal de Liberação de Cálcio do Receptor de Rianodina , Proteínas Supressoras de Tumor , Animais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Masculino , Envelhecimento/metabolismo , Camundongos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Miocárdio/metabolismo , Miocárdio/patologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Técnicas de Silenciamento de Genes , Endossomos/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/genética , Coração/fisiopatologia , Camundongos Endogâmicos C57BL , Humanos , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/genética , Sístole
11.
Stem Cell Res ; 77: 103411, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582058

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Mutação de Sentido Incorreto , Canal de Liberação de Cálcio do Receptor de Rianodina , Humanos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Miopatia da Parte Central/genética , Miopatia da Parte Central/patologia , Miopatia da Parte Central/metabolismo , Adulto , Linhagem Celular , Masculino , Diferenciação Celular , Feminino
12.
Stem Cell Res ; 77: 103410, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38583293

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes Induzidas , Hipertermia Maligna , Mutação de Sentido Incorreto , Canal de Liberação de Cálcio do Receptor de Rianodina , Humanos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Hipertermia Maligna/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Miopatia da Parte Central/genética , Miopatia da Parte Central/patologia , Masculino , Feminino , Linhagem Celular , Diferenciação Celular
13.
J Electrocardiol ; 84: 75-80, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38574633

RESUMO

In this case report, we describe a 14-year-old patient with a novel RyR2 gene mutation (c.6577G > T/p.Val2193Leu), identified through a comprehensive review of medical history, examination findings, and follow-up data. The pathogenic potential of this mutation, which results in the loss of some interatomic forces and compromises the closure of the RyR2 protein pore leading to calcium leakage, was analyzed using the I-TASSER Suite to predict the structural changes in the protein. This mutation manifested clinically as co-morbid catecholaminergic polymorphic ventricular tachycardia (CPVT) and benign epilepsy with centrotemporal spikes (BECTS), a combination not previously documented in the same patient. While seizures were successfully managed with levetiracetam, the patient's exercise-induced syncope episodes could not be controlled with metoprolol, highlighting the complexity and challenge in managing CPVT associated with this novel RyR2 variation.


Assuntos
Mutação , Canal de Liberação de Cálcio do Receptor de Rianodina , Taquicardia Ventricular , Humanos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Taquicardia Ventricular/genética , Taquicardia Ventricular/tratamento farmacológico , Adolescente , Masculino , Epilepsia Rolândica/genética , Epilepsia Rolândica/tratamento farmacológico , Eletrocardiografia
14.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(5): 581-585, 2024 May 10.
Artigo em Chinês | MEDLINE | ID: mdl-38684305

RESUMO

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.


Assuntos
Heterozigoto , Miopatia da Parte Central , Canal de Liberação de Cálcio do Receptor de Rianodina , Humanos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Masculino , Criança , Miopatia da Parte Central/genética , Sequenciamento do Exoma , Mutação , Testes Genéticos
16.
BMC Pulm Med ; 24(1): 194, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38649898

RESUMO

BACKGROUND: Patients with congenital myopathies may experience respiratory involvement, resulting in restrictive ventilatory dysfunction and respiratory failure. Pulmonary hypertension (PH) associated with this condition has never been reported in congenital ryanodine receptor type 1(RYR1)-related myopathy. CASE PRESENTATION: A 47-year-old woman was admitted with progressively exacerbated chest tightness and difficulty in neck flexion. She was born prematurely at week 28. Her bilateral lower extremities were edematous and muscle strength was grade IV-. Arterial blood gas analysis revealed hypoventilation syndrome and type II respiratory failure, while lung function test showed restrictive ventilation dysfunction, which were both worse in the supine position. PH was confirmed by right heart catheterization (RHC), without evidence of left heart disease, congenital heart disease, or pulmonary artery obstruction. Polysomnography indicated nocturnal hypoventilation. The ultrasound revealed reduced mobility of bilateral diaphragm. The level of creatine kinase was mildly elevated. Magnetic resonance imaging showed myositis of bilateral thigh muscle. Muscle biopsy of the left biceps brachii suggested muscle malnutrition and congenital muscle disease. Gene testing revealed a missense mutation in the RYR1 gene (exon33 c.C4816T). Finally, she was diagnosed with RYR1-related myopathy and received long-term non-invasive ventilation (NIV) treatment. Her symptoms and cardiopulmonary function have been greatly improved after 10 months. CONCLUSIONS: We report a case of RYR1-related myopathy exhibiting hypoventilation syndrome, type II respiratory failure and PH associated with restrictive ventilator dysfunction. Pulmonologists should keep congenital myopathies in mind in the differential diagnosis of type II respiratory failure, especially in patients with short stature and muscle weakness.


Assuntos
Hipertensão Pulmonar , Debilidade Muscular , Insuficiência Respiratória , Canal de Liberação de Cálcio do Receptor de Rianodina , Humanos , Feminino , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Pessoa de Meia-Idade , Debilidade Muscular/etiologia , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/genética , Insuficiência Respiratória/etiologia , Mutação de Sentido Incorreto , Imageamento por Ressonância Magnética , Doenças Musculares/genética , Doenças Musculares/diagnóstico , Doenças Musculares/complicações
17.
Proc Natl Acad Sci U S A ; 121(17): e2218204121, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38621141

RESUMO

Inherited arrhythmia syndromes (IASs) can cause life-threatening arrhythmias and are responsible for a significant proportion of sudden cardiac deaths (SCDs). Despite progress in the development of devices to prevent SCDs, the precise molecular mechanisms that induce detrimental arrhythmias remain to be fully investigated, and more effective therapies are desirable. In the present study, we screened a large-scale randomly mutagenized mouse library by electrocardiography to establish a disease model of IASs and consequently found one pedigree that exhibited spontaneous ventricular arrhythmias (VAs) followed by SCD within 1 y after birth. Genetic analysis successfully revealed a missense mutation (p.I4093V) of the ryanodine receptor 2 gene to be a cause of the arrhythmia. We found an age-related increase in arrhythmia frequency accompanied by cardiomegaly and decreased ventricular contractility in the Ryr2I4093V/+ mice. Ca2+ signaling analysis and a ryanodine binding assay indicated that the mutant ryanodine receptor 2 had a gain-of-function phenotype and enhanced Ca2+ sensitivity. Using this model, we detected the significant suppression of VA following flecainide or dantrolene treatment. Collectively, we established an inherited life-threatening arrhythmia mouse model from an electrocardiogram-based screen of randomly mutagenized mice. The present IAS model may prove feasible for use in investigating the mechanisms of SCD and assessing therapies.


Assuntos
Taquicardia Ventricular , Camundongos , Animais , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Arritmias Cardíacas/genética , Flecainida , Mutação de Sentido Incorreto , Morte Súbita Cardíaca , Mutação
18.
Proc Natl Acad Sci U S A ; 121(19): e2317753121, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38687794

RESUMO

Type 1 voltage-activated calcium channels (CaV1) in the plasma membrane trigger calcium release from the sarcoplasmic reticulum (SR) by two mechanisms. In voltage-induced calcium release (VICR), CaV1 voltage sensing domains are directly coupled to ryanodine receptors (RYRs), an SR calcium channel. In calcium-induced calcium release (CICR), calcium ions flowing through activated CaV1 channels bind and activate RYR channels. VICR is thought to occur exclusively in vertebrate skeletal muscle while CICR occurs in all other muscles (including all invertebrate muscles). Here, we use calcium-activated SLO-2 potassium channels to analyze CaV1-SR coupling in Caenorhabditis elegans body muscles. SLO-2 channels were activated by both VICR and external calcium. VICR-mediated SLO-2 activation requires two SR calcium channels (RYRs and IP3 Receptors), JPH-1/Junctophilin, a PDZ (PSD95, Dlg1, ZO-1 domain) binding domain (PBD) at EGL-19/CaV1's carboxy-terminus, and SHN-1/Shank (a scaffolding protein that binds EGL-19's PBD). Thus, VICR occurs in invertebrate muscles.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Canais de Cálcio , Cálcio , Proteínas de Membrana Transportadoras , Proteínas Musculares , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Cálcio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/metabolismo , Músculos/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Proteínas de Membrana/metabolismo , Sinalização do Cálcio/fisiologia
19.
Chem Pharm Bull (Tokyo) ; 72(4): 399-407, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38644198

RESUMO

Ryanodine receptor 2 (RyR2) is a large Ca2+-release channel in the sarcoplasmic reticulum (SR) of cardiac muscle cells. It serves to release Ca2+ from the SR into the cytosol to initiate muscle contraction. RyR2 overactivation is associated with arrhythmogenic cardiac disease, but few specific inhibitors have been reported so far. Here, we identified an RyR2-selective inhibitor 1 from the chemical compound library and synthesized it from glycolic acid. Synthesis of various derivatives to investigate the structure-activity relationship of each substructure afforded another two RyR2-selective inhibitors 6 and 7, among which 6 was the most potent. Notably, compound 6 also inhibited Ca2+ release in cells expressing the RyR2 mutants R2474S, R4497C and K4750Q, which are associated with cardiac arrhythmias such as catecholaminergic polymorphic ventricular tachycardia (CPVT). This inhibitor is expected to be a useful tool for research on the structure and dynamics of RyR2, as well as a lead compound for the development of drug candidates to treat RyR2-related cardiac disease.


Assuntos
Bloqueadores dos Canais de Cálcio , Canal de Liberação de Cálcio do Receptor de Rianodina , Humanos , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Descoberta de Drogas , Células HEK293 , Estrutura Molecular , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Relação Estrutura-Atividade , Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Antiarrítmicos/química , Antiarrítmicos/farmacologia , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/genética
20.
Arch Cardiovasc Dis ; 117(6-7): 382-391, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38670870

RESUMO

BACKGROUND: With the development of advanced sequencing techniques, genetic testing has emerged as a valuable tool for the work-up of non-ischaemic sudden cardiac arrest (SCA). AIMS: To evaluate the effectiveness of genetic testing in patients with unexplained SCA, according to clinical phenotype. METHODS: All patients who underwent molecular genetic testing for non-ischaemic SCA with no left ventricular cardiomyopathy between 2012 and 2021 in two French university hospitals were included. RESULTS: Of 66 patients (mean age 36.7±11.9years, 54.5% men), 21 (31.8%; 95% confidence interval 22.4-45.3%) carried a genetic variant: eight (12.1%) had a pathogenic or likely pathogenic (P/LP) variant and 13 (19.7%) had a variant of uncertain significance (VUS). Among 37 patients (56.1%) with no phenotypic clues, genetic testing identified a P/LP variant in five (13.5%), mainly in RYR2 (n=3) and SCN5A (n=2), and a VUS in nine (24.3%). None of the nine patients with phenotypic evidence of channelopathies had P/LP variants, but two had VUS in RYR2 and NKX2.5. Among the 20 patients with suspected arrhythmogenic cardiomyopathy, three P/LP variants (15.0%) and two VUS (10.0%) were found in DSC2, PKP2, SCN5A and DSG2, TRPM4, respectively. Genetic testing was performed sooner after cardiac arrest (P<0.001) and results were obtained more rapidly (P=0.02) after versus before 2016. CONCLUSION: This study highlights the utility of molecular genetic testing with a genetic variant of interest identified in one-third of patients with unexplained SCA. Genetic testing was beneficial even in patients without phenotypic clues, with one-fourth of patients carrying a P/LP variant that could have direct implications.


Assuntos
Morte Súbita Cardíaca , Predisposição Genética para Doença , Testes Genéticos , Fenótipo , Valor Preditivo dos Testes , Humanos , Masculino , Feminino , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/prevenção & controle , Adulto , Pessoa de Meia-Idade , França , Adulto Jovem , Fatores de Risco , Hospitais Universitários , Estudos Retrospectivos , Mutação , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Marcadores Genéticos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA