Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Circ Res ; 128(3): 419-432, 2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33342222

RESUMO

RATIONALE: The cardiac sodium channel NaV1.5 has a fundamental role in excitability and conduction. Previous studies have shown that sodium channels cluster together in specific cellular subdomains. Their association with intracellular organelles in defined regions of the myocytes, and the functional consequences of that association, remain to be defined. OBJECTIVE: To characterize a subcellular domain formed by sodium channel clusters in the crest region of the myocytes and the subjacent subsarcolemmal mitochondria. METHODS AND RESULTS: Through a combination of imaging approaches including super-resolution microscopy and electron microscopy we identified, in adult cardiac myocytes, a NaV1.5 subpopulation in close proximity to subjacent subsarcolemmal mitochondria; we further found that subjacent subsarcolemmal mitochondria preferentially host the mitochondrial NCLX (Na+/Ca2+ exchanger). This anatomic proximity led us to investigate functional changes in mitochondria resulting from sodium channel activity. Upon TTX (tetrodotoxin) exposure, mitochondria near NaV1.5 channels accumulated more Ca2+ and showed increased reactive oxygen species production when compared with interfibrillar mitochondria. Finally, crosstalk between NaV1.5 channels and mitochondria was analyzed at a transcriptional level. We found that SCN5A (encoding NaV1.5) and SLC8B1 (which encode NaV1.5 and NCLX, respectively) are negatively correlated both in a human transcriptome data set (Genotype-Tissue Expression) and in human-induced pluripotent stem cell-derived cardiac myocytes deficient in SCN5A. CONCLUSIONS: We describe an anatomic hub (a couplon) formed by sodium channel clusters and subjacent subsarcolemmal mitochondria. Preferential localization of NCLX to this domain allows for functional coupling where the extrusion of Ca2+ from the mitochondria is powered, at least in part, by the entry of sodium through NaV1.5 channels. These results provide a novel entry-point into a mechanistic understanding of the intersection between electrical and structural functions of the heart.


Assuntos
Cálcio/metabolismo , Mitocôndrias Cardíacas/metabolismo , Proteínas Mitocondriais/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Animais , Sinalização do Cálcio , Linhagem Celular , Feminino , Humanos , Cinética , Masculino , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Mitocôndrias Cardíacas/ultraestrutura , Proteínas Mitocondriais/genética , Miócitos Cardíacos/ultraestrutura , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Imagem Individual de Molécula , Trocador de Sódio e Cálcio/genética , Superóxidos/metabolismo
2.
Int J Mol Sci ; 20(17)2019 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-31438494

RESUMO

Human variants in plakophilin-2 (PKP2) associate with most cases of familial arrhythmogenic cardiomyopathy (ACM). Recent studies show that PKP2 not only maintains intercellular coupling, but also regulates transcription of genes involved in Ca2+ cycling and cardiac rhythm. ACM penetrance is low and it remains uncertain, which genetic and environmental modifiers are crucial for developing the cardiomyopathy. In this study, heterozygous PKP2 knock-out mice (PKP2-Hz) were used to investigate the influence of exercise, pressure overload, and inflammation on a PKP2-related disease progression. In PKP2-Hz mice, protein levels of Ca2+-handling proteins were reduced compared to wildtype (WT). PKP2-Hz hearts exposed to voluntary exercise training showed right ventricular lateral connexin43 expression, right ventricular conduction slowing, and a higher susceptibility towards arrhythmias. Pressure overload increased levels of fibrosis in PKP2-Hz hearts, without affecting the susceptibility towards arrhythmias. Experimental autoimmune myocarditis caused more severe subepicardial fibrosis, cell death, and inflammatory infiltrates in PKP2-Hz hearts than in WT. To conclude, PKP2 haploinsufficiency in the murine heart modulates the cardiac response to environmental modifiers via different mechanisms. Exercise upon PKP2 deficiency induces a pro-arrhythmic cardiac remodeling, likely based on impaired Ca2+ cycling and electrical conduction, versus structural remodeling. Pathophysiological stimuli mainly exaggerate the fibrotic and inflammatory response.


Assuntos
Cálcio/metabolismo , Cardiomiopatias/metabolismo , Haploinsuficiência/fisiologia , Doença Autoimune do Sistema Nervoso Experimental/etiologia , Doença Autoimune do Sistema Nervoso Experimental/metabolismo , Placofilinas/metabolismo , Animais , Western Blotting , Cardiomiopatias/etiologia , Cardiomiopatias/patologia , Ecocardiografia , Eletrocardiografia , Fibrose/etiologia , Fibrose/metabolismo , Fibrose/patologia , Haploinsuficiência/genética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doença Autoimune do Sistema Nervoso Experimental/patologia , Placofilinas/genética , Reação em Cadeia da Polimerase
3.
Swiss Med Wkly ; 154(7): 3615, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38980543

RESUMO

AIM OF THE STUDY: The aim of this study is to provide an analysis of the career trajectory of the recipients of a Swiss National MD-PhD grant thirty years after the creation of the Swiss interuniversity MD-PhD programme. METHODS: The study surveyed 277 recipients of a Swiss National MD-PhD grant using an online questionnaire in April 2022. There were twenty questions about participants' demographics, the duration of their MD-PhD training, their career trajectory, current position, research and clinical activity, the impact of the support on the recipients' careers, and their satisfaction with various aspects of the grant. RESULTS: The study showed that 141 out of the 277 grant recipients contacted returned the survey (51% response rate). The gender distribution of the participants was 33% women, 63% men, 4% unknown, which is almost the same as that of all grantees (35% women, 65% men). One hundred and fourteen (81%) respondents had completed their MD-PhD thesis and were graduates, while 27 (19%) were still MD-PhD students. The mean duration of the MD-PhD training was 4.27 years, with a slight upward trend over time. A large proportion of graduates, 81%, remained scientifically active after the grant, most of them in academic settings. Of the grantees who had completed their MD-PhD at least eight years before the survey, 55% had a paid research position with 40% combining research and clinical roles, and 15% doing research only. Seventy-six per cent remained clinically active, 54% occupied leadership positions, and 25% were professors. Most grantees believed that the grant had had a positive impact on their career trajectory. The main challenges included a delay in clinical training, a limited number of clinical positions with dedicated research time after the MD-PhD period, and sub-optimal recognition by hospital hierarchies. CONCLUSION: The data collected for this study confirm that the competitive Swiss National MD-PhD Grants Programme excels in supporting promising physician scientists who remain active in both research and clinical contexts in the long term. The individual grants are perceived as a distinction that acts as the basis for a successful career in academic medicine. Continued support and alternative funding sources, however, will be essential to ensure the programme's sustainability.


Assuntos
Escolha da Profissão , Humanos , Suíça , Feminino , Masculino , Inquéritos e Questionários , Adulto , Organização do Financiamento/estatística & dados numéricos , Pesquisa Biomédica/estatística & dados numéricos , Educação de Pós-Graduação em Medicina/estatística & dados numéricos
4.
Circ Arrhythm Electrophysiol ; 13(7): e008241, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32536203

RESUMO

BACKGROUND: Mutations in the gene encoding the cardiac voltage-gated sodium channel Nav1.5 cause various cardiac arrhythmias. This variety may arise from different determinants of Nav1.5 expression between cardiomyocyte domains. At the lateral membrane and T-tubules, Nav1.5 localization and function remain insufficiently characterized. METHODS: We used novel single-molecule localization microscopy and computational modeling to define nanoscale features of Nav1.5 localization and distribution at the lateral membrane, the lateral membrane groove, and T-tubules in cardiomyocytes from wild-type (N=3), dystrophin-deficient (mdx; N=3) mice, and mice expressing C-terminally truncated Nav1.5 (ΔSIV; N=3). We moreover assessed T-tubules sodium current by recording whole-cell sodium currents in control (N=5) and detubulated (N=5) wild-type cardiomyocytes. RESULTS: We show that Nav1.5 organizes as distinct clusters in the groove and T-tubules which density, distribution, and organization partially depend on SIV and dystrophin. We found that overall reduction in Nav1.5 expression in mdx and ΔSIV cells results in a nonuniform redistribution with Nav1.5 being specifically reduced at the groove of ΔSIV and increased in T-tubules of mdx cardiomyocytes. A T-tubules sodium current could, however, not be demonstrated. CONCLUSIONS: Nav1.5 mutations may site-specifically affect Nav1.5 localization and distribution at the lateral membrane and T-tubules, depending on site-specific interacting proteins. Future research efforts should elucidate the functional consequences of this redistribution.


Assuntos
Membrana Celular/metabolismo , Ativação do Canal Iônico , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Imagem Individual de Molécula , Animais , Membrana Celular/ultraestrutura , Simulação por Computador , Distrofina/genética , Distrofina/metabolismo , Potenciais da Membrana , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Modelos Cardiovasculares , Miócitos Cardíacos/ultraestrutura , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Transporte Proteico
5.
Channels (Austin) ; 14(1): 268-286, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32815768

RESUMO

The cardiac voltage-gated sodium channel Nav1.5 conducts the rapid inward sodium current crucial for cardiomyocyte excitability. Loss-of-function mutations in its gene SCN5A are linked to cardiac arrhythmias such as Brugada Syndrome (BrS). Several BrS-associated mutations in the Nav1.5 N-terminal domain (NTD) exert a dominant-negative effect (DNE) on wild-type channel function, for which mechanisms remain poorly understood. We aim to contribute to the understanding of BrS pathophysiology by characterizing three mutations in the Nav1.5 NTD: Y87C-here newly identified-, R104W, and R121W. In addition, we hypothesize that the calcium sensor protein calmodulin is a new NTD binding partner. Recordings of whole-cell sodium currents in TsA-201 cells expressing WT and variant Nav1.5 showed that Y87C and R104W but not R121W exert a DNE on WT channels. Biotinylation assays revealed reduction in fully glycosylated Nav1.5 at the cell surface and in whole-cell lysates. Localization of Nav1.5 WT channel with the ER did not change in the presence of variants, as shown by transfected and stained rat neonatal cardiomyocytes. We demonstrated that calmodulin binds the Nav1.5 NTD using in silico modeling, SPOTS, pull-down, and proximity ligation assays. Calmodulin binding to the R121W variant and to a Nav1.5 construct missing residues 80-105, a predicted calmodulin-binding site, is impaired. In conclusion, we describe the new natural BrS Nav1.5 variant Y87C and present first evidence that calmodulin binds to the Nav1.5 NTD, which seems to be a determinant for the DNE.


Assuntos
Calmodulina/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Animais , Western Blotting , Síndrome de Brugada/metabolismo , Calmodulina/genética , Linhagem Celular , Células Cultivadas , Eletroforese em Gel de Poliacrilamida , Eletrofisiologia , Éxons/genética , Humanos , Masculino , Pessoa de Meia-Idade , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Ligação Proteica , Ratos , Adulto Jovem
6.
Front Physiol ; 10: 1487, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31920695

RESUMO

T-tubules are invaginations of the lateral membrane of striated muscle cells that provide a large surface for ion channels and signaling proteins, thereby supporting excitation-contraction coupling. T-tubules are often remodeled in heart failure. To better understand the electrical behavior of T-tubules of cardiac cells in health and disease, this study addresses two largely unanswered questions regarding their electrical properties: (1) the delay of T-tubular membrane depolarization and (2) the effects of T-tubular sodium current on T-tubular potentials. Here, we present an elementary computational model to determine the delay in depolarization of deep T-tubular membrane segments as the narrow T-tubular lumen provides resistance against the extracellular current. We compare healthy tubules to tubules with constrictions and diseased tubules from mouse and human, and conclude that constrictions greatly delay T-tubular depolarization, while diseased T-tubules depolarize faster than healthy ones due to tubule widening. Increasing the tubule length non-linearly delays the depolarization. We moreover model the effect of T-tubular sodium current on intraluminal T-tubular potentials. We observe that extracellular potentials become negative during the sodium current transient (up to -40 mV in constricted T-tubules), which feedbacks on sodium channel function (self-attenuation) in a manner resembling ephaptic effects that have been described for intercalated discs where opposing membranes are very close together. The intraluminal potential and sodium current self-attenuation however greatly depend on sodium current conductance. These results show that (1) the changes in passive electrical properties of remodeled T-tubules cannot explain the excitation-contraction coupling defects in diseased cells; and (2) the sodium current may modulate intraluminal potentials. Such extracellular potentials might also affect excitation-contraction coupling and macroscopic conduction.

7.
Sci Data ; 5: 180170, 2018 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-30129933

RESUMO

Mice are used universally as model organisms for studying heart physiology, and a plethora of genetically modified mouse models exist to study cardiac disease. Transcriptomic data for whole-heart tissue are available, but not yet for isolated ventricular cardiomyocytes. Our lab therefore collected comprehensive RNA-seq data from wildtype murine ventricular cardiomyocytes as well as from knockout models of the ion channel regulators CASK, dystrophin, and SAP97. We also elucidate ion channel expression from wild-type cells to help forward the debate about which ion channels are expressed in cardiomyocytes. Researchers studying the heart, and especially cardiac arrhythmias, may benefit from these cardiomyocyte-specific transcriptomic data to assess expression of genes of interest.

8.
Sci Data ; 5: 180216, 2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30299437

RESUMO

This corrects the article DOI: 10.1038/sdata.2018.170.

9.
Cardiovasc Res ; 113(3): 259-275, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28069669

RESUMO

This review presents an extensively integrated model of the cardiac intercalated disc (ID), a highly orchestrated structure that connects adjacent cardiomyocytes. Classically, three main structures are distinguished: gap junctions (GJs) metabolically and electrically connect cytoplasm of adjacent cardiomyocytes; adherens junctions (AJs) connect the actin cytoskeleton of adjacent cells; and desmosomes function as cell anchors and connect intermediate filaments. Furthermore, ion channels reside in the ID. Mutations in ID proteins have been associated with cardiac arrhythmias such as Brugada syndrome and arrhythmogenic cardiomyopathy. However, rather than being independent, all ID components work together intensively by multifunctional proteins such as ZO-1, Ankyrin G, and ß-catenin, integrating mechanical and electrical functions. GJs form a plaque surrounded by the perinexus in which free connexons reside; the connexome integrates NaV channels, the desmosome and GJs; and the area composita hosts AJs and desmosomes, also integrated as adhering junctions. Furthermore, the transitional junction connects sarcomeres to the plasma membrane. Lastly, this review integrates all these findings in comprehensible figures, illustrating the interdependencies of ID proteins.


Assuntos
Arritmias Cardíacas/metabolismo , Comunicação Celular , Junções Intercelulares/metabolismo , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/metabolismo , Transdução de Sinais , Junções Aderentes/metabolismo , Junções Aderentes/patologia , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Arritmias Cardíacas/fisiopatologia , Desmossomos/metabolismo , Desmossomos/patologia , Junções Comunicantes/metabolismo , Junções Comunicantes/patologia , Predisposição Genética para Doença , Humanos , Junções Intercelulares/genética , Junções Intercelulares/patologia , Canais Iônicos/metabolismo , Mecanotransdução Celular , Proteínas de Membrana/genética , Mutação , Miócitos Cardíacos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA