Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Gen Physiol ; 156(4)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38385988

RESUMO

We have previously demonstrated that type II ryanodine receptors (RyR2) tetramers can be rapidly rearranged in response to a phosphorylation cocktail. The cocktail modified downstream targets indiscriminately, making it impossible to determine whether phosphorylation of RyR2 was an essential element of the response. Here, we used the ß-agonist isoproterenol and mice homozygous for one of the following clinically relevant mutations: S2030A, S2808A, S2814A, or S2814D. We measured the length of the dyad using transmission electron microscopy (TEM) and directly visualized RyR2 distribution using dual-tilt electron tomography. We found that the S2814D mutation, by itself, significantly expanded the dyad and reorganized the tetramers, suggesting a direct link between the phosphorylation state of the tetramer and its microarchitecture. S2808A and S2814A mutant mice, as well as wild types, had significant expansions of their dyads in response to isoproterenol, while S2030A mutants did not. In agreement with functional data from these mutants, S2030 and S2808 were necessary for a complete ß-adrenergic response, unlike S2814 mutants. Additionally, all mutants had unique effects on the organization of their tetramer arrays. Lastly, the correlation of structural with functional changes suggests that tetramer-tetramer contacts play an important functional role. We thus conclude that both the size of the dyad and the arrangement of the tetramers are linked to the state of the channel tetramer and can be dynamically altered by a ß-adrenergic receptor agonist.


Assuntos
Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Camundongos , Isoproterenol/farmacologia , Mutação , Fosforilação , Canal de Liberação de Cálcio do Receptor de Rianodina/química
2.
Front Cell Dev Biol ; 12: 1298007, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38304423

RESUMO

Atrial fibrillation (AF), the most common arrhythmia, has been associated with different electrophysiological, molecular, and structural alterations in atrial cardiomyocytes. Therefore, more studies are required to elucidate the genetic and molecular basis of AF. Various genome-wide association studies (GWAS) have strongly associated different single nucleotide polymorphisms (SNPs) with AF. One of these GWAS identified the rs13376333 risk SNP as the most significant one from the 1q21 chromosomal region. The rs13376333 risk SNP is intronic to the KCNN3 gene that encodes for small conductance calcium-activated potassium channels type 3 (SK3). However, the functional electrophysiological effects of this variant are not known. SK channels represent a unique family of K+ channels, primarily regulated by cytosolic Ca2+ concentration, and different studies support their critical role in the regulation of atrial excitability and consequently in the development of arrhythmias like AF. Since different studies have shown that both upregulation and downregulation of SK3 channels can lead to arrhythmias by different mechanisms, an important goal is to elucidate whether the rs13376333 risk SNP is a gain-of-function (GoF) or a loss-of-function (LoF) variant. A better understanding of the functional consequences associated with these SNPs could influence clinical practice guidelines by improving genotype-based risk stratification and personalized treatment. Although research using native human atrial cardiomyocytes and animal models has provided useful insights, each model has its limitations. Therefore, there is a critical need to develop a human-derived model that represents human physiology more accurately than existing animal models. In this context, research with human induced pluripotent stem cells (hiPSC) and subsequent generation of cardiomyocytes derived from hiPSC (hiPSC-CMs) has revealed the underlying causes of various cardiovascular diseases and identified treatment opportunities that were not possible using in vitro or in vivo studies with animal models. Thus, the ability to generate atrial cardiomyocytes and atrial tissue derived from hiPSCs from human/patients with specific genetic diseases, incorporating novel genetic editing tools to generate isogenic controls and organelle-specific reporters, and 3D bioprinting of atrial tissue could be essential to study AF pathophysiological mechanisms. In this review, we will first give an overview of SK-channel function, its role in atrial fibrillation and outline pathophysiological mechanisms of KCNN3 risk SNPs. We will then highlight the advantages of using the hiPSC-CM model to investigate SNPs associated with AF, while addressing limitations and best practices for rigorous hiPSC studies.

3.
Biomed Opt Express ; 14(11): 5710-5719, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-38021136

RESUMO

Three-dimensional (3D) structured illumination microscopy (SIM) improves spatial resolution by a factor of two in both lateral and axial directions. However, the adoption of 3D SIM is limited by low imaging speed, susceptibility to out-of-focus light, and likelihood of reconstruction errors. Here we present a novel approach for 3D SIM using a spinning disk. The disk generates a 3D lattice illumination pattern on the sample and optically reconstructs super-resolved images in real time. This technique achieves a 2-times resolution improvement with a speed up to 100 frames per second while physically rejecting 90% of the background signal.

4.
J Gen Physiol ; 155(11)2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37728575

RESUMO

Earlier work has shown that ventricular ryanodine receptors (RyR2) within a cluster rearrange on phosphorylation as well as with a number of other stimuli. Using dSTORM, we investigated the effects of 300 nmol/liter isoproterenol on RyR2 clusters. In rat ventricular cardiomyocytes, there was a symmetrical enlargement of RyR2 cluster areas, a decrease in the edge-to-edge nearest neighbor distance, and distribution changes that suggested movement to increase the cluster areas by coalescence. The surface area covered by the phosphorylated clusters was significantly greater than in the control cells, as was the cluster density. This latter change was accompanied by a decreased cluster fragmentation, implying that new tetramers were brought into the sarcoplasmic reticulum. We propose a possible mechanism to explain these changes. We also visualized individual RyR2 tetramers and confirmed our earlier electron-tomographic finding that the tetramers are in a disorganized but non-random array occupying about half of the cluster area. Multiclusters, cluster groups defined by the maximum distance between their members, were analyzed for various distances. At 100 nm, the areas occupied by the multiclusters just exceeded those of the single clusters, and more than half of the multiclusters had only a single subcluster that could initiate a spark. Phosphorylation increased the size of the multiclusters, markedly so for distances >100 nm. There was no relationship between the number of subclusters in a group and the area covered by it. We conclude that isoproterenol induces rapid, significant, changes in the molecular architecture of excitation-contraction coupling.


Assuntos
Miócitos Cardíacos , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Ratos , Isoproterenol/farmacologia , Acoplamento Excitação-Contração , Análise por Conglomerados
5.
Opt Lett ; 48(15): 3933-3936, 2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37527086

RESUMO

Among various super-resolution microscopic techniques, structured illumination microscopy (SIM) stands out for live-cell imaging because of its higher imaging speed. However, conventional SIM lacks optical sectioning capability. Here we demonstrate a new, to the best of our knowledge, approach using a phase-modulated spinning disk (PMSD) that enhances the optical sectioning capability of SIM. The PMSD consists of a pinhole array for confocal imaging and a transparent polymer layer for light phase modulation. The light phase modulation was designed to cancel the zeroth-order diffracted beam and create a sharp lattice illumination pattern using the interference of four first-order diffracted beams. In the detection optical path, the PMSD serves as a spatial filter to physically reject about 80% of the out-of-focus signals, an approach that allows for real-time optical reconstruction of super-resolved images with enhanced contrast. Furthermore, the simplicity of the design makes it easy to upgrade a conventional fluorescence microscope to a PMSD SIM system.

6.
bioRxiv ; 2023 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-37292875

RESUMO

We have previously demonstrated that type II ryanodine receptors (RyR2) tetramers can be rapidly rearranged in response to a phosphorylation cocktail. The cocktail modified downstream targets indiscriminately making it impossible to determine whether phosphorylation of RyR2 was an essential element of the response. We therefore used the ß-agonist isoproterenol and mice with one of the homozygous mutations, S2030A +/+ , S2808A +/+ , S2814A +/+ , or S2814D +/+ , to address this question and to elucidate the role of these clinically relevant mutations. We measured the length of the dyad using transmission electron microscopy (TEM) and directly visualized RyR2 distribution using dual-tilt electron tomography. We found that: 1) The S2814D mutation, by itself, significantly expanded the dyad and reorganized the tetramers suggesting a direct link between the phosphorylation state of the tetramer and the microarchitecture. 2) All of the wild-type, as well as the S2808A and S2814A mice, had significant expansions of their dyads in response to ISO, while S2030A did not. 3) In agreement with functional data from the same mutants, S2030 and S2808 were necessary for a complete ß-adrenergic response, whereas S2814 was not. 4) All the mutated residues had unique effects on the organization of their tetramer arrays. 5) The correlation of structure with function suggests that tetramer-tetramer contacts play an important functional role. We conclude that both the size of the dyad and the arrangement of the tetramers are linked to the state of the channel tetramer and can be dynamically altered by a ß-adrenergic receptor agonist. Summary: Analysis of RyR2 mutants suggests a direct link between the phosphorylation state of the channel tetramer and the microarchitecture of the dyad. All phosphorylation site mutations produced significant and unique effects on the structure of the dyad and its response to isoproterenol.

7.
Front Cardiovasc Med ; 9: 967659, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36061558

RESUMO

Cardiovascular diseases are the leading cause of mortality and reduced quality of life globally. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a personalized platform to study inherited heart diseases, drug-induced cardiac toxicity, and cardiac regenerative therapy. However, the immaturity of CMs obtained by current strategies is a major hurdle in utilizing hiPSC-CMs at their fullest potential. Here, the major findings and limitations of current maturation methodologies to enhance the utility of hiPSC-CMs in the battle against a major source of morbidity and mortality are reviewed. The most recent knowledge of the potential signaling pathways involved in the transition of fetal to adult CMs are assimilated. In particular, we take a deeper look on role of nutrient sensing signaling pathways and the potential role of cap-independent translation mediated by the modulation of mTOR pathway in the regulation of cardiac gap junctions and other yet to be identified aspects of CM maturation. Moreover, a relatively unexplored perspective on how our knowledge on the effects of preterm birth on cardiovascular development can be actually utilized to enhance the current understanding of CM maturation is examined. Furthermore, the interaction between the evolving neonatal human heart and brown adipose tissue as the major source of neonatal thermogenesis and its endocrine function on CM development is another discussed topic which is worthy of future investigation. Finally, the current knowledge regarding transcriptional mediators of CM maturation is still limited. The recent studies have produced the groundwork to better understand CM maturation in terms of providing some of the key factors involved in maturation and development of metrics for assessment of maturation which proves essential for future studies on in vitro PSC-CMs maturation.

8.
Curr Protoc ; 1(12): e320, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34958715

RESUMO

Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a potentially lethal inherited cardiac arrhythmia condition, triggered by physical or acute emotional stress, that predominantly expresses early in life. Gain-of-function mutations in the cardiac ryanodine receptor gene (RYR2) account for the majority of CPVT cases, causing substantial disruption of intracellular calcium (Ca2+ ) homeostasis particularly during the periods of ß-adrenergic receptor stimulation. However, the highly variable penetrance, patient outcomes, and drug responses observed in clinical practice remain unexplained, even for patients with well-established founder RyR2 mutations. Therefore, investigation of the electrophysiological consequences of CPVT-causing RyR2 mutations is crucial to better understand the pathophysiology of the disease. The development of strategies for reprogramming human somatic cells to human induced pluripotent stem cells (hiPSCs) has provided a unique opportunity to study inherited arrhythmias, due to the ability of hiPSCs to differentiate down a cardiac lineage. Employment of genome editing enables generation of disease-specific cell lines from healthy and diseased patient-derived hiPSCs, which subsequently can be differentiated into cardiomyocytes. This paper describes the means for establishing an hiPSC-based model of CPVT in order to recapitulate the disease phenotype in vitro and investigate underlying pathophysiological mechanisms. The framework of this approach has the potential to contribute to disease modeling and personalized medicine using hiPSC-derived cardiomyocytes. © 2021 Wiley Periodicals LLC.


Assuntos
Células-Tronco Pluripotentes Induzidas , Taquicardia Ventricular , Humanos , Miócitos Cardíacos , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Taquicardia Ventricular/genética
9.
Iran J Pathol ; 15(3): 167-174, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32754211

RESUMO

BACKGROUND & OBJECTIVE: Most colorectal cancers (CRCs) arise from adenomatous polyps, and clinical management of this type of polyp is highly dependent on the reliability and validity of the pathological diagnosis. The aim of this study was to examine the interobserver agreement of five pathologists in assessing dysplasia in adenomatous polyps. METHODS: In this study, a total of 146 adenomatous polyps of patients undergoing colonoscopy were selected from hospitals of Shahid Beheshti University of Medical Sciences, Tehran, Iran between 2017 and 2018. Five pathologists independently classified adenomatous polyps according to histologic type, nuclear pseudostratification, mitotic activity, nuclear polarity, nuclear pleomorphism, nuclear shape, nucleolus, chromatin pattern, cytology grade, architectural features, dysplasia, and final diagnosis. The overall kappa statistic (k) was used to assess agreement among pathologists. RESULTS: The mean age of the patients was 62.06 ± 13.06 (mean ± SD) with a male-to-female ratio of 2.2:1. The most common site of resection was the sigmoid colon (28.1%). The highest agreement was found for dysplasia grade (k=0.415) and histologic type (k=0.401), whereas the lowest agreement was found for mitotic activity (k=0.185), nuclear shape (k=0.187), and nucleolus (k=0.196). CONCLUSION: Our findings indicate that agreement among pathologists in assessing dysplasia in adenomatous polyps is within fair to moderate levels of agreement. Therefore, there is a vital need to better clarify the current diagnostic criteria.

10.
Elife ; 92020 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-31916935

RESUMO

The effects of the immunophilins, FKBP12 and FKBP12.6, and phosphorylation on type II ryanodine receptor (RyR2) arrangement and function were examined using correlation microscopy (line scan confocal imaging of Ca2+ sparks and dual-tilt electron tomography) and dSTORM imaging of permeabilized Wistar rat ventricular myocytes. Saturating concentrations (10 µmol/L) of either FKBP12 or 12.6 significantly reduced the frequency, spread, amplitude and Ca2+ spark mass relative to control, while the tomograms revealed both proteins shifted the tetramers into a largely side-by-side configuration. Phosphorylation of immunophilin-saturated RyR2 resulted in structural and functional changes largely comparable to phosphorylation alone. dSTORM images of myocyte surfaces demonstrated that both FKBP12 and 12.6 significantly reduced RyR2 cluster sizes, while phosphorylation, even of immunophilin-saturated RyR2, increased them. We conclude that both RyR2 cluster size and the arrangement of tetramers within clusters is dynamic and respond to changes in the cellular environment. Further, these changes affect Ca2+ spark formation.


Assuntos
Miócitos Cardíacos/metabolismo , Processamento de Proteína Pós-Traducional , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Cálcio/metabolismo , Ventrículos do Coração , Fosforilação , Estrutura Quaternária de Proteína , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Retículo Sarcoplasmático/metabolismo , Proteína 1A de Ligação a Tacrolimo/farmacologia , Proteínas de Ligação a Tacrolimo/farmacologia
11.
Biol Reprod ; 96(6): 1288-1302, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28486663

RESUMO

The endoplasmic reticulum (ER) in Sertoli cells is a component of unique adhesion junctions (ectoplasmic specializations-ESs) and is closely associated with structures termed tubulobulbar complexes (TBCs) that internalize intercellular junctions during sperm release and during the translocation of spermatocytes through the blood-testis barrier. A role for the ER in Ca2+ regulation at ESs and TBCs has been suspected, but evidence for this function has proved elusive. Using electron microscopy, we define two new ER-plasma membrane (PM) contact sites in apical Sertoli cell processes. One of these sites occurs at TBCs where flattened lamellar cisternae of ER envelope the swollen bulb regions of the complexes, and where the gap between adjacent membranes is 12 nm. The other is at the periphery of apical processes where the gap between membranes is 13-14 nm. Using immunolocalization at the light and electron microscopic levels, we demonstrate that Ca2+ regulatory machinery is present at the ESs attached to spermatid heads, and at ER-PM contacts. Sarco/endoplasmic reticulum Ca2+-ATPase 2 (ATP2A2, SERCA2) is present at ESs; transient receptor potential channel subfamily M member 6 (TRPM6), Homer1 (HOMER1), and inositol 1,4,5-trisphosphate receptor (ITPR, IP3R) are present at ER-PM contacts associated with TBC bulbs; and stromal interacting molecule 1 (STIM1), Orai1 (ORAI1), and ATP2A2 are present at the ER-PM contacts around the margins of Sertoli cell apical processes. In Sertoli cells, the molecular machinery associated with ER generated Ca2+ fluxes is present in regions and structures directly related to junction remodeling-a process necessary for sperm release.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Retículo Endoplasmático/fisiologia , Junções Intercelulares/fisiologia , Células de Sertoli/fisiologia , Animais , Regulação da Expressão Gênica , Masculino , Ratos , Ratos Sprague-Dawley , Células de Sertoli/ultraestrutura
12.
J Biol Chem ; 291(45): 23490-23505, 2016 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-27621312

RESUMO

Cardiac ryanodine receptor (Ryr2) Ca2+ release channels and cellular metabolism are both disrupted in heart disease. Recently, we demonstrated that total loss of Ryr2 leads to cardiomyocyte contractile dysfunction, arrhythmia, and reduced heart rate. Acute total Ryr2 ablation also impaired metabolism, but it was not clear whether this was a cause or consequence of heart failure. Previous in vitro studies revealed that Ca2+ flux into the mitochondria helps pace oxidative metabolism, but there is limited in vivo evidence supporting this concept. Here, we studied heart-specific, inducible Ryr2 haploinsufficient (cRyr2Δ50) mice with a stable 50% reduction in Ryr2 protein. This manipulation decreased the amplitude and frequency of cytosolic and mitochondrial Ca2+ signals in isolated cardiomyocytes, without changes in cardiomyocyte contraction. Remarkably, in the context of well preserved contractile function in perfused hearts, we observed decreased glucose oxidation, but not fat oxidation, with increased glycolysis. cRyr2Δ50 hearts exhibited hyperphosphorylation and inhibition of pyruvate dehydrogenase, the key Ca2+-sensitive gatekeeper to glucose oxidation. Metabolomic, proteomic, and transcriptomic analyses revealed additional functional networks associated with altered metabolism in this model. These results demonstrate that Ryr2 controls mitochondrial Ca2+ dynamics and plays a specific, critical role in promoting glucose oxidation in cardiomyocytes. Our findings indicate that partial RYR2 loss is sufficient to cause metabolic abnormalities seen in heart disease.


Assuntos
Sinalização do Cálcio , Glucose/metabolismo , Contração Miocárdica , Miocárdio/metabolismo , Complexo Piruvato Desidrogenase/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Cálcio/metabolismo , Deleção de Genes , Metaboloma , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/citologia , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Oxirredução , Proteoma , Piruvatos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/genética
13.
Circ Res ; 115(2): 252-62, 2014 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-24786399

RESUMO

RATIONALE: Single-tilt tomograms of the dyads in rat ventricular myocytes indicated that type 2 ryanodine receptors (RYR2s) were not positioned in a well-ordered array. Furthermore, the orientation and packing strategy of purified type 1 ryanodine receptors in lipid bilayers is determined by the free Mg2+ concentration. These observations led us to test the hypothesis that RYR2s within the mammalian dyad have multiple and complex arrangements. OBJECTIVES: To determine the arrangement of RYR2 tetramers in the dyads of mammalian cardiomyocytes and the effects of physiologically and pathologically relevant factors on this arrangement. METHODS AND RESULTS: We used dual-tilt electron tomography to produce en-face views of dyads, enabling a direct examination of RYR2 distribution and arrangement. Rat hearts fixed in situ; isolated rat cardiomyocytes permeabilized, incubated with 1 mmol/L Mg2+, and then fixed; and sections of human ventricle, all showed that the tetramer packing within a dyad was nonuniform containing a mix of checkerboard and side-by-side arrangements, as well as isolated tetramers. Both phosphorylation and 0.1 mmol/L Mg2+ moved the tetramers into a predominantly checkerboard configuration, whereas the 4 mmol/L Mg2+ induced a dense side-by-side arrangement. These changes occurred within 10 minutes of application of the stimuli. CONCLUSIONS: The arrangement of RYR2 tetramers within the mammalian dyad is neither uniform nor static. We hypothesize that this is characteristic of the dyad in vivo and may provide a mechanism for modulating the open probabilities of the individual tetramers.


Assuntos
Acoplamento Excitação-Contração , Ventrículos do Coração/química , Miócitos Cardíacos/química , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Animais , Sinalização do Cálcio/efeitos dos fármacos , Tomografia com Microscopia Eletrônica , Ativação Enzimática/efeitos dos fármacos , Ventrículos do Coração/citologia , Ventrículos do Coração/ultraestrutura , Humanos , Magnésio/farmacologia , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/ultraestrutura , Fosforilação , Proteínas Quinases/fisiologia , Processamento de Proteína Pós-Traducional , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Retículo Sarcoplasmático/fisiologia
14.
Cardiovasc Res ; 98(2): 169-76, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23400762

RESUMO

This review highlights recent and ongoing discoveries that are transforming the previously held view of dyad structure and function. New data show that dyads vary greatly in both structure and in their associated molecules. Dyads can contain varying numbers of type 2 ryanodine receptor (RYR2) clusters that range in size from one to hundreds of tetramers and they can adopt numerous orientations other than the expected checkerboard. The association of Ca(v)1.2 with RYR2, which defines the couplon, is not absolute, leading to a number of scenarios such as dyads without couplons and those in which only a fraction of the clusters are in couplons. Different dyads also vary in the transporters and exchangers with which they are associated producing functional differences that amplify their structural diversity. The essential role of proteins, such as junctophilin-2, calsequestrin, triadin, and junctin that maintain both the functional and structural integrity of the dyad have recently been elucidated giving a new mechanistic understanding of heart diseases, such as arrhythmias, hypertension, failure, and sudden cardiac death.


Assuntos
Miócitos Cardíacos/ultraestrutura , Sarcolema/ultraestrutura , Retículo Sarcoplasmático/ultraestrutura , Animais , Cálcio/metabolismo , Canais de Cálcio Tipo L/fisiologia , Calsequestrina/fisiologia , Acoplamento Excitação-Contração , Humanos , Proteínas de Membrana/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Trocador de Sódio e Cálcio/fisiologia
15.
Cardiovasc Res ; 96(3): 372-80, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22869620

RESUMO

AIMS: The molecular mechanisms controlling heart function and rhythmicity are incompletely understood. While it is widely accepted that the type 2 ryanodine receptor (Ryr2) is the major Ca(2+) release channel in excitation-contraction coupling, the role of these channels in setting a consistent beating rate remains controversial. Gain-of-function RYR2 mutations in humans and genetically engineered mouse models are known to cause Ca(2+) leak, arrhythmias, and sudden cardiac death. Embryonic stem-cell derived cardiomyocytes lacking Ryr2 display slower beating rates, but no supporting in vivo evidence has been presented. The aim of the present study was to test the hypothesis that RYR2 loss-of-function would reduce heart rate and rhythmicity in vivo. METHODS AND RESULTS: We generated inducible, tissue-specific Ryr2 knockout mice with acute ∼50% loss of RYR2 protein in the heart but not in other tissues. Echocardiography, working heart perfusion, and in vivo ECG telemetry demonstrated that deletion of Ryr2 was sufficient to cause bradycardia and arrhythmia. Our results also show that cardiac Ryr2 knockout mice exhibit functional and structural hallmarks of heart failure, including sudden cardiac death. CONCLUSION: These results illustrate that the RYR2 channel plays an essential role in pacing heart rate. Moreover, we find that RYR2 loss-of-function can lead to fatal arrhythmias typically associated with gain-of-function mutations. Given that RYR2 levels can be reduced in pathological conditions, including heart failure and diabetic cardiomyopathy, we predict that RYR2 loss contributes to disease-associated bradycardia, arrhythmia, and sudden death.


Assuntos
Arritmias Cardíacas/metabolismo , Relógios Biológicos , Frequência Cardíaca , Miocárdio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Arritmias Cardíacas/diagnóstico por imagem , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Bradicardia/genética , Bradicardia/metabolismo , Bradicardia/fisiopatologia , Débito Cardíaco , Morte Súbita Cardíaca/etiologia , Regulação para Baixo , Eletrocardiografia Ambulatorial/métodos , Acoplamento Excitação-Contração , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Miocárdica , RNA Mensageiro/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/deficiência , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Telemetria , Fatores de Tempo , Ultrassonografia , Função Ventricular
16.
Protoplasma ; 249 Suppl 1: S31-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22057630

RESUMO

The couplons of the cardiomyocyte form nanospaces within the cell that place the L-type calcium channel (Ca(v)1.2), situated on the plasmalemma, in opposition to the type 2 ryanodine receptor (RyR2), situated on the sarcoplasmic reticulum. These two molecules, which form the basis of excitation-contraction coupling, are separated by a very limited space, which allows a few Ca(2+) ions passing through Ca(v)1.2 to activate the RyR2 at concentration levels that would be deleterious to the whole cell. The limited space also allows Ca(2+) inactivation of Ca(v)1.2. We have found that not all couplons are the same and that their properties are likely determined by their molecular partners which, in turn, determine their excitability. In particular, there are a class of couplons that lie outside the RyR2-Ca(v)1.2 dyad; in this case, the RyR2 is close to caveolin-3 rather than Ca(v)1.2. These extra-dyadic couplons are probably controlled by the multitude of molecules associated with caveolin-3 and may modulate contractile force under situations such as stress. It has long been assumed that like the skeletal muscle, the RyR2 in the couplon are arranged in a structured array with the RyR2 interacting with each other via domain 6 of the RyR2 molecule. This arrangement was thought to provide local control of RyR2 excitability. Using 3D electron tomography of the couplon, we show that the RyR2 in the couplon do not form an ordered pattern, but are scattered throughout it. Relatively few are in a checkerboard pattern--many RyR2 sit edge-to-edge, a configuration which might preclude their controlling each other's excitability. The discovery of this structure makes many models of cardiac couplon function moot and is a current avenue of further research.


Assuntos
Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sarcolema/ultraestrutura , Retículo Sarcoplasmático/ultraestrutura , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Caveolina 3/metabolismo , Humanos , Contração Muscular , Ratos , Sarcolema/fisiologia , Retículo Sarcoplasmático/fisiologia
17.
Biophys J ; 99(12): 3923-9, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21156134

RESUMO

We analyzed the distribution of ryanodine receptor (RyR) and Cav1.2 clusters in adult rat ventricular myocytes using three-dimensional object-based colocalization metrics. We found that ∼75% of the Cav1.2 clusters and 65% of the RyR clusters were within couplons, and both were roughly two and a half times larger than their extradyadic counterparts. Within a couplon, Cav1.2 was concentrated near the center of the underlying RyR cluster and accounted for ∼67% of its size. These data, together with previous findings from binding studies, enable us to estimate that a couplon contains 74 RyR tetramers and 10 copies of the α-subunit of Cav1.2. Extradyadic clusters of RyR contained ∼30 tetramers, whereas the extradyadic Cav1.2 clusters contained, on average, only four channels. Between 80% and 85% of both RyR and Cav1.2 molecules are within couplons. RyR clusters were in the closest proximity, with a median nearest-neighbor distance of 552 nm; comparable values for Cav1.2 clusters and couplons were 619 nm and 735 nm, respectively. Extradyadic RyR clusters were significantly closer together (624 nm) and closer to the couplons (674 nm) than the couplons were to each other. In contrast, the extradyadic clusters of Cav1.2 showed no preferential localization and were broadly distributed. These results provide a wealth of morphometric data that are essential for understanding intracellular Ca2+ regulation and modeling Ca2+ dynamics.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Animais , Análise por Conglomerados , Ventrículos do Coração , Masculino , Miócitos Cardíacos/citologia , Ratos , Ratos Wistar , Coloração e Rotulagem
18.
Dev Biol ; 338(1): 15-27, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19879257

RESUMO

Integrin-mediated adhesion to the ECM is essential for normal development of animal tissues. During muscle development, integrins provide the structural stability required to construct such a highly tensile, force generating tissue. Mutations that disrupt integrin-mediated adhesion in skeletal muscles give rise to a myopathy in humans and mice. To determine if this is due to defects in formation or defects in maintenance of muscle tissue, we used an inducible, targeted RNAi based approach to disrupt integrin-mediated adhesion in fully formed adult fly muscles. A decrease in integrin-mediated adhesion in adult muscles led to a progressive loss of muscle function due to a failure to maintain normal sarcomeric cytoarchitecture. This defect was due to a gradual, age dependent disorganization of the sarcomeric actin, Z-line, and M-line. Electron microscopic analysis showed that reduction in integrin-mediated adhesion resulted in detachment of actin filaments from the Z-lines, separation of the Z-lines from the membrane, and eventually to disintegration of the Z-lines. Our results show that integrin-mediated adhesion is essential for maintaining sarcomeric integrity and illustrate that the seemingly stable adhesive contacts underlying sarcomeric architecture are inherently dynamic.


Assuntos
Drosophila melanogaster/citologia , Integrinas/metabolismo , Sarcômeros/metabolismo , Actinina/metabolismo , Actinas/metabolismo , Envelhecimento/metabolismo , Animais , Adesão Celular , Diferenciação Celular , Drosophila melanogaster/enzimologia , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/ultraestrutura , Voo Animal , Técnicas de Silenciamento de Genes , Modelos Biológicos , Músculos/metabolismo , Músculos/patologia , Especificidade de Órgãos , Fenótipo , Sarcômeros/patologia , Sarcômeros/ultraestrutura , Talina/metabolismo , Fatores de Tempo
19.
Biophys J ; 96(11): 4651-60, 2009 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-19486687

RESUMO

Ryanodine receptors (RyRs) are located primarily on the junctional sarcoplasmic reticulum (SR), adjacent to the transverse tubules and on the cell surface near the Z-lines, but some RyRs are on junctional SR adjacent to axial tubules. Neither the size of the axial junctions nor the numbers of RyRs that they contain have been determined. RyRs may also be located on the corbular SR and on the free or network SR. Because determining and quantifying the distribution of RyRs is critical for both understanding and modeling calcium dynamics, we investigated the distribution of RyRs in healthy adult rat ventricular myocytes, using electron microscopy, electron tomography, and immunofluorescence. We found RyRs in only three regions: in couplons on the surface and on transverse tubules, both of which are near the Z-line, and in junctions on most of the axial tubules--axial junctions. The axial junctions averaged 510 nm in length, but they occasionally spanned an entire sarcomere. Numerical analysis showed that they contain as much as 19% of a cell's RyRs. Tomographic analysis confirmed the axial junction's architecture, which is indistinguishable from junctions on transverse tubules or on the surface, and revealed a complexly structured tubule whose lumen was only 26 nm at its narrowest point. RyRs on axial junctions colocalize with Ca(v)1.2, suggesting that they play a role in excitation-contraction coupling.


Assuntos
Monócitos/metabolismo , Monócitos/ultraestrutura , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/ultraestrutura , Retículo Sarcoplasmático/metabolismo , Retículo Sarcoplasmático/ultraestrutura , Animais , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo L/ultraestrutura , Imunofluorescência , Átrios do Coração/metabolismo , Átrios do Coração/ultraestrutura , Ventrículos do Coração/ultraestrutura , Imageamento Tridimensional , Masculino , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Ratos , Ratos Wistar , Sarcômeros/metabolismo , Sarcômeros/ultraestrutura , Tomografia
20.
Proc Natl Acad Sci U S A ; 106(18): 7636-41, 2009 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-19383796

RESUMO

Heart muscle excitation-contraction (E-C) coupling is governed by Ca(2+) release units (CRUs) whereby Ca(2+) influx via L-type Ca(2+) channels (Cav1.2) triggers Ca(2+) release from juxtaposed Ca(2+) release channels (RyR2) located in junctional sarcoplasmic reticulum (jSR). Although studies suggest that the jSR protein triadin anchors cardiac calsequestrin (Casq2) to RyR2, its contribution to E-C coupling remains unclear. Here, we identify the role of triadin using mice with ablation of the Trdn gene (Trdn(-/-)). The structure and protein composition of the cardiac CRU is significantly altered in Trdn(-/-) hearts. jSR proteins (RyR2, Casq2, junctin, and junctophilin 1 and 2) are significantly reduced in Trdn(-/-) hearts, whereas Cav1.2 and SERCA2a remain unchanged. Electron microscopy shows fragmentation and an overall 50% reduction in the contacts between jSR and T-tubules. Immunolabeling experiments show reduced colocalization of Cav1.2 with RyR2 and substantial Casq2 labeling outside of the jSR in Trdn(-/-) myocytes. CRU function is impaired in Trdn(-/-) myocytes, with reduced SR Ca(2+) release and impaired negative feedback of SR Ca(2+) release on Cav1.2 Ca(2+) currents (I(Ca)). Uninhibited Ca(2+) influx via I(Ca) likely contributes to Ca(2+) overload and results in spontaneous SR Ca(2+) releases upon beta-adrenergic receptor stimulation with isoproterenol in Trdn(-/-) myocytes, and ventricular arrhythmias in Trdn(-/-) mice. We conclude that triadin is critically important for maintaining the structural and functional integrity of the cardiac CRU; triadin loss and the resulting alterations in CRU structure and protein composition impairs E-C coupling and renders hearts susceptible to ventricular arrhythmias.


Assuntos
Arritmias Cardíacas/fisiopatologia , Cálcio/metabolismo , Proteínas de Transporte/fisiologia , Coração/fisiopatologia , Proteínas Musculares/fisiologia , Contração Miocárdica , Retículo Sarcoplasmático/metabolismo , Animais , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Canais de Cálcio Tipo L/metabolismo , Proteínas de Transporte/genética , Coração/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Mutantes , Proteínas Musculares/genética , Contração Miocárdica/genética , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Retículo Sarcoplasmático/ultraestrutura , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA