Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
J Biol Chem ; 298(1): 101412, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34793835

RESUMO

The N-terminal region (NTR) of ryanodine receptor (RyR) channels is critical for the regulation of Ca2+ release during excitation-contraction (EC) coupling in muscle. The NTR hosts numerous mutations linked to skeletal (RyR1) and cardiac (RyR2) myopathies, highlighting its potential as a therapeutic target. Here, we constructed two biosensors by labeling the mouse RyR2 NTR at domains A, B, and C with FRET pairs. Using fluorescence lifetime (FLT) detection of intramolecular FRET signal, we developed high-throughput screening (HTS) assays with these biosensors to identify small-molecule RyR modulators. We then screened a small validation library and identified several hits. Hits with saturable FRET dose-response profiles and previously unreported effects on RyR were further tested using [3H]ryanodine binding to isolated sarcoplasmic reticulum vesicles to determine effects on intact RyR opening in its natural membrane. We identified three novel inhibitors of both RyR1 and RyR2 and two RyR1-selective inhibitors effective at nanomolar Ca2+. Two of these hits activated RyR1 only at micromolar Ca2+, highlighting them as potential enhancers of excitation-contraction coupling. To determine whether such hits can inhibit RyR leak in muscle, we further focused on one, an FDA-approved natural antibiotic, fusidic acid (FA). In skinned skeletal myofibers and permeabilized cardiomyocytes, FA inhibited RyR leak with no detrimental effect on skeletal myofiber excitation-contraction coupling. However, in intact cardiomyocytes, FA induced arrhythmogenic Ca2+ transients, a cautionary observation for a compound with an otherwise solid safety record. These results indicate that HTS campaigns using the NTR biosensor can identify compounds with therapeutic potential.


Assuntos
Técnicas Biossensoriais , Canal de Liberação de Cálcio do Receptor de Rianodina , Animais , Cálcio/metabolismo , Transferência Ressonante de Energia de Fluorescência , Ensaios de Triagem em Larga Escala , Camundongos , Músculo Esquelético/química , Músculo Esquelético/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
2.
Medicine (Baltimore) ; 100(33): e26999, 2021 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-34414986

RESUMO

INTRODUCTION: Malignant hyperthermia (MH) and exertional rhabdomyolysis (ERM) have long been considered episodic phenotypes occurring in response to external triggers in otherwise healthy individuals with variants in RYR1. However, recent studies have demonstrated a clinical and histopathological continuum between patients with RYR1-related congenital myopathies and those with ERM or MH susceptibility. Furthermore, animal studies have shown non-neuromuscular features such as a mild bleeding disorder and an immunological gain-of-function associated with MH/ERM related RYR1 variants raising important questions for further research. Awareness of the neuromuscular disease spectrum and potential multisystem involvement in RYR1-related MH and ERM is essential to optimize the diagnostic work-up, improve counselling and and future treatment strategies for patients affected by these conditions. This study will examine in detail the nature and severity of continuous disease manifestations and their effect on daily life in patients with RYR1-related MH and ERM. METHODS: The study protocol consists of four parts; an online questionnaire study, a clinical observational study, muscle imaging, and specific immunological studies. Patients with RYR1-related MH susceptibility and ERM will be included. The imaging, immunological and clinical studies will have a cross-sectional design, while the questionnaire study will be performed three times during a year to assess disease impact, daily living activities, fatigue and pain. The imaging study consists of muscle ultrasound and whole-body magnetic resonance imaging studies. For the immunological studies, peripheral mononuclear blood cells will be isolated for in vitro stimulation with toll-like receptor ligands, to examine the role of the immune system in the pathophysiology of RYR1-related MH and ERM. DISCUSSION: This study will increase knowledge of the full spectrum of neuromuscular and multisystem features of RYR1-related MH and ERM and will establish a well-characterized baseline cohort for future studies on RYR1-related disorders. The results of this study are expected to improve recognition of RYR1-related symptoms, counselling and a more personalized approach to patients affected by these conditions. Furthermore, results will create new insights in the role of the immune system in the pathophysiology of MH and ERM. TRIAL REGISTRATION: This study was pre-registered at ClinicalTrials.gov (ID: NCT04610619).


Assuntos
Protocolos Clínicos , Hipertermia Maligna/etiologia , Rabdomiólise/etiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Estudos de Coortes , Estudos Transversais , Humanos , Hipertermia Maligna/genética , Estudos Prospectivos , Rabdomiólise/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Inquéritos e Questionários
3.
Microsc Microanal ; 26(1): 157-165, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31931893

RESUMO

The voltage-gated sodium channel [pore-forming subunit of the neuronal voltage-gated sodium channel (NaV1.6)] has recently been found in cardiac myocytes. Emerging studies indicate a role for NaV1.6 in ionic homeostasis as well as arrhythmogenesis. Little is known about the spatial organization of these channels in cardiac muscle, mainly due to the lack of high-fidelity antibodies. Therefore, we developed and rigorously validated a novel rabbit polyclonal NaV1.6 antibody and undertook super-resolution microscopy studies of NaV1.6 localization in cardiac muscle. We developed and validated a novel rabbit polyclonal antibody against a C-terminal epitope on the neuronal sodium channel 1.6 (NaV1.6). Raw sera showed high affinity in immuno-fluorescence studies, which was improved with affinity purification. The antibody was rigorously validated for specificity via multiple approaches. Lastly, we used this antibody in proximity ligation assay (PLA) and super-resolution STochastic Optical Reconstruction Microscopy (STORM) studies, which revealed enrichment of NaV1.6 in close proximity to ryanodine receptor (RyR2), a key calcium (Ca2+) cycling protein, in cardiac myocytes. In summary, our novel NaV1.6 antibody demonstrates high degrees of specificity and fidelity in multiple preparations. It enabled multimodal microscopic studies and revealed that over half of the NaV1.6 channels in cardiac myocytes are located within 100 nm of ryanodine receptor Ca2+ release channels.


Assuntos
Miocárdio/citologia , Canal de Sódio Disparado por Voltagem NAV1.6/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Animais , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Imagem Óptica
4.
Sci Rep ; 9(1): 7867, 2019 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-31133706

RESUMO

Pathological cardiac hypertrophy is a debilitating condition characterized by deleterious thickening of the myocardium, dysregulated Ca2+ signaling within cardiomyocytes, and contractile dysfunction. Importantly, the nanoscale organization, localization, and patterns of expression of critical Ca2+ handling regulators including dihydropyridine receptor (DHPR), ryanodine receptor 2 (RyR2), phospholamban (PLN), and sarco/endoplasmic reticulum Ca2+-ATPase 2A (SERCA2A) remain poorly understood, especially during pathological hypertrophy disease progression. In the current study, we induced cardiac pathological hypertrophy via transverse aortic constriction (TAC) on 8-week-old CD1 mice, followed by isolation of cardiac ventricular myocytes. dSTORM super-resolution imaging was then used to visualize proteins at nanoscale resolution at two time points and we quantified changes in protein cluster properties using Voronoi tessellation and 2D Fast Fourier Transform analyses. We showed a decrease in the density of DHPR and RyR2 clusters with pressure-overload cardiac hypertrophy and an increase in the density of SERCA2A protein clusters. PLN protein clusters decreased in density in 2-week TAC but returned to sham levels by 4-week TAC. Furthermore, 2D-FFT analysis revealed changes in molecular organization during pathological hypertrophy, with DHPR and RyR2 becoming dispersed while both SERCA2A and PLN sequestered into dense clusters. Our work reveals molecular adaptations that occur in critical SR proteins at a single molecule during pressure overload-induced cardiomyopathy. Nanoscale alterations in protein localization and patterns of expression of crucial SR proteins within the cardiomyocyte provided insights into the pathogenesis of cardiac hypertrophy, and specific evidence that cardiomyocytes undergo significant structural remodeling during the progression of pathological hypertrophy.


Assuntos
Cardiomegalia/patologia , Miócitos Cardíacos/patologia , Retículo Sarcoplasmático/patologia , Animais , Canais de Cálcio Tipo L/análise , Proteínas de Ligação ao Cálcio/análise , Cardiomegalia/diagnóstico por imagem , Células Cultivadas , Análise de Fourier , Camundongos , Microscopia , Imagem Óptica , Pressão , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/análise
5.
J Microsc ; 267(2): 160-175, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28370211

RESUMO

Multicolour fluorescence imaging by STimulated Emission Depletion (STED) superresolution microscopy with doughnut-shaped STED laser beams based on different wavelengths for each colour channel requires precise image registration. This is especially important when STED imaging is used for co-localisation studies of two or more native proteins in biological specimens to analyse nanometric subcellular spatial arrangements. We developed a robust postprocessing image registration protocol, with the aim to verify and ultimately optimise multicolour STED image quality. Importantly, this protocol will support any subsequent quantitative localisation analysis at nanometric scales. Henceforth, using an approach that registers each colour channel present during STED imaging individually, this protocol reliably corrects for optical aberrations and inadvertent sample drift. To achieve the latter goal, the protocol combines the experimental sample information, from corresponding STED and confocal images using the same optical beam path and setup, with that of an independent calibration sample. As a result, image registration is based on a strategy that maximises the cross-correlation between sequentially acquired images of the experimental sample, which are strategically combined by the protocol. We demonstrate the general applicability of the image registration protocol by co-staining of the ryanodine receptor calcium release channel in primary mouse cardiomyocytes. To validate this new approach, we identify user-friendly criteria, which - if fulfilled - support optimal image registration. In summary, we introduce a new method for image registration and rationally based postprocessing steps through a highly standardised protocol for multicolour STED imaging, which directly supports the reproducibility of protein co-localisation analyses. Although the reference protocol is discussed exemplarily for two-colour STED imaging, it can be readily expanded to three or more colours and STED channels.


Assuntos
Processamento de Imagem Assistida por Computador/métodos , Miócitos Cardíacos/enzimologia , Imagem Óptica/métodos , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Animais , Células Cultivadas , Camundongos
6.
Methods Enzymol ; 556: 455-74, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25857795

RESUMO

Skeletal muscle excitation-contraction coupling is triggered by the concerted action of two enormous Ca(2+) channel complexes, the dihydropyridine receptor and the type 1 ryanodine receptor. Recent advances in our understanding of the structure of these large Ca(2+) channels have been driven by fluorescence resonance energy transfer (FRET)-based analysis. A methodological challenge in conducting these FRET measurements is the ability to site-specifically label these huge ion channels with donor and acceptor fluorophores capable of undergoing energy transfer. In this chapter, we detail specific protocols for tagging large membrane proteins with these fluorescent probes using three orthogonal labeling methods: fluorescent protein fusions, biarsenical reagents directed to engineered tetracysteine tags, and Cy3/5 nitrilotriacetic acid conjugates that bind to poly-histidine tags.


Assuntos
Canais de Cálcio Tipo L/análise , Transferência Ressonante de Energia de Fluorescência/métodos , Músculo Esquelético/química , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Animais , Canais de Cálcio Tipo L/metabolismo , Corantes Fluorescentes/análise , Corantes Fluorescentes/metabolismo , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/metabolismo , Histidina/metabolismo , Humanos , Modelos Moleculares , Músculo Esquelético/metabolismo , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
7.
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
8.
Eur J Obstet Gynecol Reprod Biol ; 174: 123-7, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24405728

RESUMO

OBJECTIVE: Uterine artery occlusion by laparoscopy (UAOL) has been used for the treatment of uterine fibroids and beneficial effects to patients have been shown in clinical studies since 2000. Fibroid cells are more susceptible to apoptosis than myometrial cells under hypoxic conditions, but the molecular mechanisms underlying this effect remain unclear. The aim of this study was to investigate the role of intracellular calcium (Ca(2+)) release mediated by Ca(2+) channel inositol 1,4,5 trisphosphate receptor1 (IP3R1)/ryanodine receptor1 (RYR1) in the apoptosis of uterine fibroid cells under hypoxia. STUDY DESIGN: We compared the expressions of IP3R1 and RYR1 in fibroid and surrounding myometrial tissue from 20 patients before UAOL. After 6h treatment under hypoxia (1% O2) with or without Ca(2+) channel blockers (heparin or/and ruthenium red), the intracellular Ca(2+) concentration, cytochrome c (Cytc) protein and cell apoptosis were determined. RESULTS: IP3R1 and RYR1 mRNA and protein levels were significantly higher in fibroid than in myometrial tissues. Under hypoxic conditions, Ca(2+) concentration in fibroid cells was significantly higher than in myometrial cells (Ca(2+): 82.69±16.92nmol/L vs 46.14±9.11nmol/L, P<0.05), and Cytc increased similarly in fibroid cells. These increases in Ca(2+) concentration, Cytc and cell apoptosis were significantly reversed by calcium blocker in fibroid cells. CONCLUSION: This study demonstrated that intracellular calcium release mediated by IP3R1/RYR1 could induce apoptosis in uterine fibroid cells under hypoxic conditions, and was responsible for the susceptibility to apoptosis of fibroid cells under UAOL.


Assuntos
Cálcio/análise , Isquemia , Leiomioma/química , Miométrio/química , Neoplasias Uterinas/química , Útero/irrigação sanguínea , Adulto , Apoptose , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Hipóxia Celular , Células Cultivadas , Citocromos c/análise , Feminino , Expressão Gênica , Humanos , Receptores de Inositol 1,4,5-Trifosfato/análise , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Laparoscopia , Leiomioma/cirurgia , Pessoa de Meia-Idade , RNA Mensageiro/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/genética , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Artéria Uterina/cirurgia , Neoplasias Uterinas/cirurgia
9.
PLoS One ; 8(5): e64394, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23741324

RESUMO

Intracellular calcium signals are studied by laser-scanning confocal fluorescence microscopy. The required spatio-temporal resolution makes description of calcium signals difficult because of the low signal-to-noise ratio. We designed a new procedure of calcium spike analysis based on their fitting with a model. The accuracy and precision of calcium spike description were tested on synthetic datasets generated either with randomly varied spike parameters and Gaussian noise of constant amplitude, or with constant spike parameters and Gaussian noise of various amplitudes. Statistical analysis was used to evaluate the performance of spike fitting algorithms. The procedure was optimized for reliable estimation of calcium spike parameters and for dismissal of false events. A new algorithm was introduced that corrects the acquisition time of pixels in line-scan images that is in error due to sequential acquisition of individual pixels along the space coordinate. New software was developed in Matlab and provided for general use. It allows interactive dissection of temporal profiles of calcium spikes from x-t images, their fitting with predefined function(s) and acceptance of results on statistical grounds, thus allowing efficient analysis and reliable description of calcium signaling in cardiac myocytes down to the in situ function of ryanodine receptors.


Assuntos
Sinalização do Cálcio , Cálcio/análise , Modelos Estatísticos , Miócitos Cardíacos/metabolismo , Software , Algoritmos , Animais , Cálcio/metabolismo , Corantes Fluorescentes , Microscopia Confocal , Microscopia de Fluorescência , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Ratos , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Razão Sinal-Ruído
10.
Adv Exp Med Biol ; 740: 183-215, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22453943

RESUMO

In excitable tissues, the ryanodine receptor Ca(2+) release channel (RyR) protein complex regulates excitation-contraction coupling, exocytosis, gene expression and apoptosis. Defects in RyR function, in genetic or acquired pathologies, lead to massive disruptions of Ca(2+) release that can be lethal. Therefore, RyR has emerged as a putative therapeutic target and an increasing number of RyR-targeting drugs are currently being tested.Nonetheless this large-size channel is still a mystery in terms of structure, which hinders full characterization of the properties of this central protein. This chapter is dedicated to the methods available to examine RyR structure and function. The aim of the article is to concentrate on contemporary methodologies rather than focusing overtly on the progress that has been achieved using these techniques. Here we review a series of reliable approaches that are routinely employed to investigate this channel. Technical limitations are discussed, and technological developments are presented. This work is not a handbook, but it can be used as a resource and a starting point for the investigation of RyR at different levels of resolution.


Assuntos
Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Animais , Sítios de Ligação , Cálcio/metabolismo , Humanos , Bicamadas Lipídicas/química , Estrutura Terciária de Proteína , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/química
11.
Methods Mol Biol ; 798: 373-82, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22130848

RESUMO

The ryanodine receptor ion channels (RyRs) release Ca(2+) from the endo/sarcoplasmic reticulum in a variety of nonvertebrate and vertebrate species including flies, crustaceans, birds, fish, and amphibians. They are most abundant in skeletal and cardiac muscle, where in response to an action potential, the release of Ca(2+) ions from the sarcoplasmic reticulum through the RyRs into the cytoplasm leads to muscle contraction (i.e., excitation-contraction coupling). Here, we describe how to determine their cellular location using isoform-specific antibodies, their protein levels using an in vitro ((3)H)ryanodine-binding assay, and their cellular release of Ca(2+) using RyR-specific channel agonists and inhibitors.


Assuntos
Cálcio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Animais , Células HEK293 , Humanos , Imuno-Histoquímica , Camundongos , Músculos/metabolismo , Ligação Proteica/fisiologia , Isoformas de Proteínas/análise , Isoformas de Proteínas/imunologia , Transporte Proteico , Rianodina/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/imunologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
12.
Am J Physiol Cell Physiol ; 299(2): C264-78, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20445169

RESUMO

Ryanodine receptors (RyRs) regulate contractility in resistance-size cerebral artery smooth muscle, yet their molecular identity, subcellular location, and phenotype in this tissue remain unknown. Following rat resistance-size cerebral artery myocyte sarcoplasmic reticulum (SR) purification and incorporation into POPE-POPS-POPC (5:3:2; wt/wt) bilayers, unitary conductances of 110 +/- 8, 334 +/- 15, and 441 +/- 27 pS in symmetric 300 mM Cs(+) were usually detected. The most frequent (34/40 bilayers) conductance (334 pS) decreased to

Assuntos
Artérias Cerebrais/citologia , Artérias Cerebrais/fisiologia , Células Musculares/fisiologia , Subunidades Proteicas/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Animais , Células Cultivadas , Artérias Cerebrais/química , Feminino , Masculino , Células Musculares/química , Subunidades Proteicas/análise , Ratos , Ratos Sprague-Dawley , Canal de Liberação de Cálcio do Receptor de Rianodina/análise
13.
Mol Cell Biochem ; 341(1-2): 235-44, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20372981

RESUMO

Neurohumoral stimulation of Gq-coupled receptors has been proposed as a central mechanism in the pathogenesis of diabetic heart disease. The resulting contractile dysfunction is closely related to abnormal intracellular Ca(2+) handling with functional defects of the sarcoplasmic reticulum (SR). The present study was therefore designed to determine the role of G(q)-protein signaling via G(alpha)(11) and G(alpha)(q) in diabetes for the induction of functional and structural changes in the Ca(2+) release complex of the SR. An experimental type 1-diabetes was induced in wild type, G(alpha)(11) knockout, and G(alpha)(11/q)-knockout mice by injection of streptozotocin. Cardiac morphology and function was assessed in vivo by echocardiography. SR Ca(2+) leak was tested in vitro based on a (45)Ca(2+) assay and protein densities as well as gene expression of ryanodine receptor (RyR2), FKBP12.6, sorcin, and annexin A7 were analyzed by immunoblot and RT-PCR. In wild type animals 8 weeks of diabetes resulted in cardiac hypertrophy and SR Ca(2+) leak was increased. In addition, diabetic wild type animals showed reduced protein levels of FKBP12.6 and annexin A7. In G(alpha)(11)- and G(alpha)(11/q)-knockout animals, however, SR Ca(2+) release and cardiac phenotype remained unchanged upon induction of diabetes. Densities of the proteins that we presently analyzed were also unaltered in G(alpha)(11)-knockout mice. G(alpha)(11/q)-knockout animals even showed increased expression of sorcin and annexin A7. Thus, based on the present study we suggest a signaling pathway via the G(q)-proteins, G(alpha)(11) and G(alpha)(q), that could link increased neurohumoral stimulation in diabetes with defective RyR2 channel function by regulating protein expression of FKBP12.6, annexin A7, and sorcin.


Assuntos
Cálcio/metabolismo , Complicações do Diabetes/metabolismo , Diabetes Mellitus Experimental/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Miócitos Cardíacos/metabolismo , Retículo Sarcoplasmático/metabolismo , Animais , Anexina A7/análise , Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Diabetes Mellitus Experimental/patologia , Cardiopatias/etiologia , Cardiopatias/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Proteínas/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Proteínas de Ligação a Tacrolimo/análise
14.
Histochem Cell Biol ; 131(3): 371-82, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19002483

RESUMO

Ank1.5 is a muscle-specific isoform of ankyrin1 localized on the sarcoplasmic reticulum (SR) membrane that has been shown to interact with obscurin, a sarcomeric protein. We report here studies on the localization of obscurin and ank1.5 in embryonic and postnatal rodent skeletal muscles. Using two antibodies against epitopes in the N- and C-terminus of obscurin, two distinct patterns of localization were observed. Before birth, the antibodies against the N- and the C-terminus of obscurin stained the Z-disk and M-band, respectively. At the same time, ank1.5 was detected at the Z-disk, rising the possibility that obscurin molecules at M-band may not be able to interact with ank1.5. Localization of ank1.5 at Z-disks in E14 muscle fibers revealed that ank1.5 is among the earliest SR proteins to assemble, since its organization preceded that of other SR proteins, like SERCA and RyR. After birth, the antibody against the N-terminus of obscurin stained the M-band while that against the C-terminus stained both M-bands and the Z-disks. Starting from postnatal day 1, ank1.5 was found at the level of both M-bands and Z-disks. Altogether, from these results we infer that exposure of some obscurin epitopes changes during skeletal muscle development, resulting in distinct, antibody-specific, localization pattern. Why this occurs is not clear, yet these data indicate that the organization of obscurin at different locations in the sarcomere changes during muscle development and that this might affect the interaction with ank1.5.


Assuntos
Anquirinas/análise , Fatores de Troca do Nucleotídeo Guanina/análise , Proteínas Musculares/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Sarcômeros , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/análise , Retículo Sarcoplasmático/metabolismo , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Camundongos , Camundongos Endogâmicos , Músculo Esquelético/embriologia , Músculo Esquelético/crescimento & desenvolvimento , Proteínas Serina-Treonina Quinases , Ratos , Ratos Sprague-Dawley , Fatores de Troca de Nucleotídeo Guanina Rho
15.
Int J Sports Med ; 29(10): 795-802, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18401808

RESUMO

The aim of this study was to examine the effect of testosterone treatment on the expression of dihydropyridine and ryanodine receptors in skeletal muscle of mouse. Furthermore, the effects of training, a method also known to elevate the plasma testosterone level, were studied and compared to the effects of pure testosterone administration. Male mice were either administered with testosterone or trained with treadmill. After 6 weeks, hindlimb muscles were excised and the expression of receptors was measured by Western blotting. Furthermore, the alterations in myosin heavy chain phenotypes were studied. In general, both training and testosterone administration induced changes in the expression of both receptors and in myosin heavy chain composition. In testosterone treated mice the expression of dihydropyridine receptor in extensor digitorum longus was higher compared to the control ones (38.9 %, p = 0.026). In soleus the expression was quite the contrary (- 27.3 %, p = 0.044), as was the case with ryanodine receptor (- 51.4 %, p = 0.012). The amount of ryanodine receptors was higher in rectus femoris (144.0 %, p = 0.044) and plantaris (48.1 %, p = 0.037) in testosterone treated mice. In trained mice, the expression of ryanodine receptor was significantly higher in gastrocnemius (27.6 %, p = 0.018), soleus (57.2 %, p = 0.025), plantaris (28.5 %, p = 0.009) and extensor digitorum longus (94.8 %, p = 0.009) than in the control ones. No differences were observed in the dihydropyridine receptor level. To conclude, training has a more important role in skeletal muscle adaptation compared to increased plasma testosterone level. However, in postural muscles both treatments have comparable effects.


Assuntos
Cálcio/metabolismo , Músculo Esquelético/fisiologia , Testosterona/farmacologia , Animais , Canais de Cálcio Tipo L/análise , Canais de Cálcio Tipo L/efeitos dos fármacos , Dinamarca , Teste de Esforço , Masculino , Camundongos , Músculo Esquelético/efeitos dos fármacos , Cadeias Pesadas de Miosina/análise , Distribuição Aleatória , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/efeitos dos fármacos , Testosterona/sangue
16.
Anesth Analg ; 105(5): 1207-13, table of contents, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17959942

RESUMO

INTRODUCTION: Anesthetic preconditioning (APC) with volatile anesthetics improves recovery of contractile function and reduces calcium overload after ischemia/reperfusion (I/R). Mitochondrial and sarcolemmal K(ATP) channel openings have been implicated in APC-induced cardioprotection. In this study, we investigated the effect of APC on major calcium cycling proteins and its relation to K(ATP) channels. METHODS: Isolated perfused rat hearts were divided into seven groups: Time control (n = 10), ischemia control (n = 8), APC (n = 8), Mitochondrial K(ATP) inhibitor 5-hydroxydecanoate (5-HD, 200 microM, n = 8), Sarcolemmal K(ATP) inhibitor HMR1098 (HMR, 20 microM, n = 8), and APC plus 5-HD or APC plus HMR1098 (n = 8 each). APC was initiated by administering 1.5% isoflurane for 15 min, followed by a 15 min washout before 30 min of myocardial ischemia and 60 min of reperfusion. Ca2+-release channels (RyR2), Ca2+-adenosine triphosphatase (SERCA2a), phospholamban, plasma membrane Ca2+ ATPase, and sodium-calcium exchanger in the homogenate were determined by Western blot assay. RESULTS: APC improved contractile recovery (left ventricular developed pressure, +dP/dt, -dP/dt) after I/R, which was blocked by 5-HD and HMR. I/R depressed the density of RyR2, SERCA2a, and phospholamban, with no changes in the density of plasma membrane Ca2+ ATPase and sodium-calcium exchanger. APC reversed I/R-induced degradation of RyR2 and SERCA2a in the presence or absence of 5HD and HMR. CONCLUSIONS: I/R-induced depression in cardiac performance is associated with a down-regulation of the major sarcoplasmic reticulum Ca2+-cycling proteins. Anesthesia preconditioning with isoflurane prevents I/R-related degradation of the RyR2 and SERCA2a in the sarcoplasmic reticulum. However, this effect was independent of its activation of K(ATP) channels.


Assuntos
Cálcio/metabolismo , Precondicionamento Isquêmico Miocárdico/métodos , Isoflurano/uso terapêutico , Mitocôndrias Cardíacas/metabolismo , Canais de Potássio/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sarcolema/metabolismo , Animais , Técnicas In Vitro , Masculino , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/prevenção & controle , Ratos , Ratos Wistar , Canal de Liberação de Cálcio do Receptor de Rianodina/análise
17.
Proc Natl Acad Sci U S A ; 104(38): 14958-63, 2007 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-17848521

RESUMO

Single rat ventricular myocytes and human ventricle tissue sections were labeled with antibodies against the ryanodine receptor (RyR) and alpha-actinin to examine the 3D distribution of RyRs with confocal microscopy. Image contrast was maximized by refractive index matching and deconvolution. The RyR label formed discrete puncta representing clusters of RyRs or "couplons" around the edges of the myofilaments with a nearest-neighbor spacing of 0.66 +/- 0.06 microm in rat and 0.78 +/- 0.07 microm in human. Each bundle of myofibrils was served by approximately six couplons, which supplied a cross-sectional area of approximately 0.6 microm(2) in rat and approximately 0.8 microm(2) in human. Although the couplons were in reasonable registration with z-lines, there were discontinuities in the longitudinal position of sarcomeres so that dislocations in the order of RyR clusters occurred. There was approximately 53% longitudinal registration of RyR clusters, suggesting a nonrandom placement of couplons around the sarcomere. These data can explain the spherical propagation of Ca(2+) waves and provide quantitative 3D data sets needed for accurate modeling of cardiac Ca(2+)-induced Ca(2+) release. By quantifying labeling intensity in rat ventricular myocytes, a lower limit of 78 RyRs per cluster (on average) was obtained. By modeling the couplon as a disk wrapping around a t-tubule and fitting cluster images, 95% of couplons contained between 120 and 260 RyRs (assuming that RyRs are tight packed with a spacing of 29 nm). Assuming similar labeling efficiency in human, from the fluorescence intensity alone we estimate that human ventricular myocytes contain approximately 30% fewer RyRs per couplon than rat.


Assuntos
Miócitos Cardíacos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Actinina/química , Actinina/metabolismo , Animais , Células Cultivadas , Humanos , Microscopia Confocal , Miócitos Cardíacos/citologia , Miócitos Cardíacos/ultraestrutura , Ratos , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo
18.
Pflugers Arch ; 455(2): 297-308, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17562071

RESUMO

In several neuronal preparations, the ryanodine-sensitive calcium store was reported to participate in the generation of slow afterhyperpolarization currents (IsAHP) involved in spike frequency adaptation. We show that calcium release from the ryanodine-sensitive calcium store is a major determinant of the triggering of IsAHP in mouse CA1 pyramidal neurons. Whole-cell patch clamp recordings in hippocampus slices show that the intracellular calcium stores depletion using an inhibitor of the endoplasmic reticulum Ca2+-ATPase (5 microM cyclopiazonic acid), as well as the specific blockade of ryanodine receptors (100 microM ryanodine) both reduced the IsAHP by about 70%. Immunohistology, using an anti-RyR3 specific antibody, indicates that RyR3 expression is particularly enriched in the CA1 apical dendrites (considered as the most important site for sAHP generation). We show that our anti-RyR3 antibody acts as a functional RyR3 antagonist and induced a reduction in IsAHP by about 70%. The additional ryanodine application (100 micro M) did not further affect IsAHP, thus excluding RyR2 in IsAHP activation. Our results argue in favor of a specialized function of RyR3 in CA1 pyramidal cells in triggering IsAHP due to their localization in the apical dendrite.


Assuntos
Potenciais de Ação/fisiologia , Canais de Cálcio/fisiologia , Cálcio/metabolismo , Células Piramidais/fisiologia , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Potenciais de Ação/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Anticorpos/análise , Anticorpos/imunologia , Anticorpos/farmacologia , Eletrofisiologia , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Camundongos , Dados de Sequência Molecular , Plasticidade Neuronal/fisiologia , Técnicas de Patch-Clamp , Isoformas de Proteínas/análise , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/metabolismo , Células Piramidais/citologia , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/imunologia , Sinapses/fisiologia
19.
Am J Physiol Cell Physiol ; 293(1): C358-66, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17459948

RESUMO

The expression of carbonic anhydrase (CA) XIV was investigated in mouse skeletal muscles. Sarcoplasmic reticulum (SR) and sarcolemmal (SL) membrane fractions were isolated from wild-type (WT) and CA XIV knockout (KO) mice. The CA XIV protein of 54 kDa was present in SR and SL membrane fractions as shown by Western blot analysis. CA activity measurements of WT and KO membrane fractions showed that CA XIV accounts for approximately 50% and 66% of the total CA activities determined in the SR and SL fractions, respectively. This indicates the presence of at least one other membrane-associated CA isoform in these membranes, e.g., CA IV, CA IX, or CA XII. Muscle fibers of the extensor digitorum longus (EDL) muscle were immunostained with anti-CA XIV/FITC and anti-sarco(endo)plasmic reticulum Ca(2+)-ATPase 1/TRITC, with anti-CA XIV/FITC and anti-ryanodine receptor/TRITC, or with anti-CA XIV/FITC and anti-monocarboxylate transporter-4/TRITC. CA XIV was expressed in the plasma membrane and in the longitudinal SR but not in the terminal SR. Isometric contraction measurements of single twitches and tetani and a fatigue protocol applied to fiber bundles of the fast-twitch EDL and of the slow-twitch soleus muscle from WT and KO mice showed that the lack of SR membrane-associated CA XIV did not affect maximum force, rise and relaxation times, and fatigue behavior. Thus, it is concluded that a reduction of the total SR CA activity by approximately 50% in CA XIV KO mice does not lead to an impairment of SR function.


Assuntos
Anidrases Carbônicas/metabolismo , Fibras Musculares Esqueléticas/enzimologia , Músculo Esquelético/enzimologia , Equilíbrio Ácido-Base , Animais , ATPase de Ca(2+) e Mg(2+)/metabolismo , Anidrases Carbônicas/deficiência , Anidrases Carbônicas/genética , Glicosilação , Contração Isométrica , Camundongos , Camundongos Knockout , Transportadores de Ácidos Monocarboxílicos/análise , Fadiga Muscular , Fibras Musculares Esqueléticas/química , Proteínas Musculares/análise , Relaxamento Muscular , Força Muscular , Músculo Esquelético/química , Músculo Esquelético/citologia , Processamento de Proteína Pós-Traducional , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Sarcolema/enzimologia , Retículo Sarcoplasmático/enzimologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
20.
Biochemistry ; 46(14): 4272-9, 2007 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-17361990

RESUMO

Ryanodine receptor (RyR) mutations linked with some congenital skeletal and cardiac diseases are localized to three easily definable regions: region 1 (N-terminal domain), region 2 (central domain), and a rather broad region 3 containing the channel pore. As shown in our recent studies, the interdomain interaction between regions 1 and 2 plays a critical role in channel regulation and pathogenesis. Here we present evidence that within region 3 there is a similar channel regulation mechanism mediated by an interdomain interaction. DP15, a peptide corresponding to RyR1 residues 4820-4841, produced significant activation of [3H]ryanodine binding above threshold Ca2+ concentrations (>or=0.3 microM), but MH mutations (L4823P or L4837V) made in DP15 almost completely abolished its channel activating function. To identify the DP15 binding site(s) within RyR1, DP15 (labeled with a fluorescent probe Alexa Fluor 680 and a photoaffinity cross-linker APG) was cross-linked to RyR1, and the site of cross-linking was identified by gel analysis of fluorescently labeled proteolytic fragments with the aid of Western blotting with site-specific antibodies. The shortest fluorescently labeled band was a 96 kDa fragment which was stained with an antibody directed to the region of residues 4114-4142 of RyR1, indicating that the interaction between the region of residues 4820-4841 adjacent to the channel pore and the 96 kDa segment containing the region of residues 4114-4142 is involved in the mechanism of Ca2+-dependent channel regulation. In further support of this concept, anti-DP15 antibody and cardiac counterpart of DP15 produced channel activation similar to that of DP15.


Assuntos
Canais de Cálcio/química , Fragmentos de Peptídeos/metabolismo , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos/química , Anticorpos/metabolismo , Anticorpos/farmacologia , Sítios de Ligação , Western Blotting , Cálcio/metabolismo , Cálcio/farmacologia , Reagentes de Ligações Cruzadas/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Relação Dose-Resposta a Droga , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Peso Molecular , Músculo Esquelético/química , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/fisiologia , Ligação Proteica , Mapeamento de Interação de Proteínas , Estrutura Terciária de Proteína , Coelhos , Canal de Liberação de Cálcio do Receptor de Rianodina/análise , Canal de Liberação de Cálcio do Receptor de Rianodina/química , Canal de Liberação de Cálcio do Receptor de Rianodina/fisiologia , Retículo Sarcoplasmático
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...