Dihydropyridine receptor

二氢吡啶受体
  • 文章类型: Journal Article
    骨骼肌中的兴奋-收缩(EC)耦合表达是在1952年创造的(1)。该术语狭义地发展为仅包括三联体的过程,该过程在横管(T管)膜的去极化和肌浆网(SR)的Ca2释放之间进行干预。从1970年到1988年,阐明了EC耦合的基础。激活过程中释放Ca2的通道通过其与植物杀虫剂ryanodine的特异性结合而位于SR中。该通道被称为ryanodine受体(RyR)。RyR包含四个亚基,它们一起构成了穿越SR和T管膜之间间隙的“SR脚”结构。Ca2+通道,也称为二氢吡啶受体(DHPR),位于三元交界处的T形管膜中,对EC耦合至关重要。这两个渠道之间存在精确的关系。四个DHPR,组织为四分体,叠加在备用RyRs上。该结构与通过T管系统中膜内电荷的运动介导EC耦合的提议一致。推测DHPR充当电压传感器,通过蛋白质-蛋白质相互作用将信息传递到SR的RyRs,从而导致Ca2从SR释放。到1988年,在理解EC耦合方面取得了很大进展。然而,电压感测如何耦合到SRCa2+释放通道的打开的最终问题仍未解决。
    The expression excitation-contraction (EC) coupling in skeletal muscle was coined in 1952 (1). The term evolved narrowly to include only the processes at the triad that intervene between depolarization of the transverse tubular (T-tubular) membrane and Ca2+ release from the sarcoplasmic reticulum (SR). From 1970 to 1988, the foundation of EC coupling was elucidated. The channel through which Ca2+ was released during activation was located in the SR by its specific binding to the plant insecticide ryanodine. This channel was called the ryanodine receptor (RyR). The RyR contained four subunits that together constituted the \"SR foot\" structure that traversed the gap between the SR and the T-tubular membrane. Ca2+ channels, also called dihydropyridine receptors (DHPRs), were located in the T-tubular membrane at the triadic junction and shown to be essential for EC coupling. There was a precise relationship between the two channels. Four DHPRs, organized as tetrads, were superimposed on alternate RyRs. This structure was consistent with the proposal that EC coupling was mediated via a movement of intramembrane charge in the T-tubular system. The speculation was that the DHPR acted as a voltage sensor transferring information to the RyRs of the SR by protein-protein interaction causing the release of Ca2+ from the SR. A great deal of progress was made by 1988 toward understanding EC coupling. However, the ultimate question of how voltage-sensing is coupled to opening of the SR Ca2+ release channel remains unresolved.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    1970年代初,门控电流和不对称电荷运动的发现代表了我们对电压依赖性事件的生物物理基础的理解的显着飞跃,该事件是对神经和肌肉功能至关重要的电信号的基础。门控电流和电荷运动反映了一个基本过程,其中离子通道蛋白中的带电氨基酸残基响应膜电场的变化而移动,从而激活该蛋白的特定电压依赖性响应。在过去的50年中,对门控电流和不对称电荷运动的检测对于揭示导致可兴奋组织中的动作电位和骨骼肌中的兴奋-收缩(EC)耦合的多个分子和分子内过程至关重要。门控电流和不对称电荷运动的记录仍然是研究神经元传导和肌肉收缩的基本分子机制的重要组成部分。
    The discovery of gating currents and asymmetric charge movement in the early 1970s represented a remarkable leap forward in our understanding of the biophysical basis of voltage-dependent events that underlie electrical signalling that is vital for nerve and muscle function. Gating currents and charge movement reflect a fundamental process in which charged amino acid residues in an ion channel protein move in response to a change in the membrane electrical field and therefore activate the specific voltage-dependent response of that protein. The detection of gating currents and asymmetric charge movement over the past 50 years has been pivotal in unraveling the multiple molecular and intra-molecular processes which lead to action potentials in excitable tissues and excitation-contraction (EC) coupling in skeletal muscle. The recording of gating currents and asymmetric charge movement remains an essential component of investigations into the basic molecular mechanisms of neuronal conduction and muscle contraction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    X连锁遗传性神经肌肉疾病Duchenne型肌营养不良的特征是膜细胞骨架成分肌营养不良蛋白的原发性异常。骨骼肌中几乎完全不存在肌营养不良蛋白的Dp427-M同工型,使收缩纤维更容易进行性变性和肌膜渗漏。这反过来导致异常的钙稳态,增强的蛋白水解和受损的激发-收缩偶联。对患者活检标本和肌营养不良蛋白病遗传动物模型的基于生化和质谱的蛋白质组学研究表明,缺乏肌营养不良蛋白的自愿性肌肉中必需钙调节蛋白的浓度和/或生理功能发生了显着变化。异常包括与肌营养不良蛋白病相关的电压传感受体变化,钙释放通道,钙泵和钙结合蛋白。这篇综述文章概述了骨骼肌中肌膜肌营养不良蛋白-糖蛋白复合物和更广泛的肌营养不良蛋白复合物的重要性及其与去极化诱导的钙释放机制和兴奋-收缩-松弛周期的联系。除了慢性炎症,脂肪替代和反应性肌纤维化,X连锁肌营养不良症的主要病理生化标志表现为钙离子通过受损的质膜的慢性流入以及异常的细胞内钙通量和缓冲。因此,受损的钙处理蛋白应包括在杜氏肌营养不良症的改善的生物标志物特征中。
    The X-linked inherited neuromuscular disorder Duchenne muscular dystrophy is characterised by primary abnormalities in the membrane cytoskeletal component dystrophin. The almost complete absence of the Dp427-M isoform of dystrophin in skeletal muscles renders contractile fibres more susceptible to progressive degeneration and a leaky sarcolemma membrane. This in turn results in abnormal calcium homeostasis, enhanced proteolysis and impaired excitation-contraction coupling. Biochemical and mass spectrometry-based proteomic studies of both patient biopsy specimens and genetic animal models of dystrophinopathy have demonstrated significant changes in the concentration and/or physiological function of essential calcium-regulatory proteins in dystrophin-lacking voluntary muscles. Abnormalities include dystrophinopathy-associated changes in voltage sensing receptors, calcium release channels, calcium pumps and calcium binding proteins. This review article provides an overview of the importance of the sarcolemmal dystrophin-glycoprotein complex and the wider dystrophin complexome in skeletal muscle and its linkage to depolarisation-induced calcium-release mechanisms and the excitation-contraction-relaxation cycle. Besides chronic inflammation, fat substitution and reactive myofibrosis, a major pathobiochemical hallmark of X-linked muscular dystrophy is represented by the chronic influx of calcium ions through the damaged plasmalemma in conjunction with abnormal intracellular calcium fluxes and buffering. Impaired calcium handling proteins should therefore be included in an improved biomarker signature of Duchenne muscular dystrophy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Letter
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    In response to excitation of skeletal muscle fibers, trains of action potentials induce changes in the configuration of the dihydropyridine receptor (DHPR) anchored in the tubular membrane which opens the Ca2+ release channel in the sarcoplasmic reticulum membrane. The DHPR also functions as a voltage-gated Ca2+ channel that conducts L-type Ca2+ currents routinely recorded in mammalian muscle fibers, which role was debated for more than four decades. Recently, to allow a closer look into the role of DHPR Ca2+ influx in mammalian muscle, a knock-in (ki) mouse model (ncDHPR) carrying mutation N617D (adjacent to domain II selectivity filter E) in the DHPRα1S subunit abolishing Ca2+ permeation through the channel was generated [Dayal et al., 2017]. In the present study, the Mn2+ quenching technique was initially intended to be used on voltage-clamped muscle fibers from this mouse to determine whether Ca2+ influx through a pathway distinct from DHPR may occur to compensate for the absence of DHPR Ca2+ influx. Surprisingly, while N617D DHPR muscle fibers of the ki mouse do not conduct Ca2+, Mn2+ entry and subsequent quenching did occur because Mn2+ was able to permeate and produce L-type currents through N617D DHPR. N617D DHPR was also found to conduct Ba2+ and Ba2+ currents were strongly blocked by external Ca2+. Ba2+ permeation was smaller, current kinetics slower and Ca2+ block more potent than in wild-type DHPR. These results indicate that residue N617 when replaced by the negatively charged residue D is suitably located at entrance of the pore to trap external Ca2+ impeding in this way permeation. Because Ba2+ binds with lower affinity to D, Ba2+ currents occur, but with reduced amplitudes as compared to Ba2+ currents through wild-type channels. We conclude that mutations located outside the selectivity filter influence channel permeation and possibly channel gating in a fully differentiated skeletal muscle environment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Endocannabinoids play important roles in regulating CNS synaptic function and peripheral metabolism, but cannabinoids can also act acutely to modulate contraction strength in skeletal muscle. Nerve terminals and the skeletal muscle sarcolemma express components of the cannabinoid signaling system. Endocannabinoids, N-arachidonylethanolamine (anandamide, AEA) and 2-arachidonoyl-glycerol (2-AG), are produced by skeletal muscle. They may be involved in the acute regulation of neuromuscular transmission, by adjusting the parameters for quantal acetylcholine release from the motor nerve terminal. Downstream of neuromuscular transmission, cannabinoids may also act to limit the efficiency of excitation-contraction coupling. Improved understanding of the distinct signaling actions of particular cannabinoid compounds and their receptor/transduction systems will help advance our understanding of the role of endocannabinoids in skeletal muscle physiology. Cannabinoids might also offer the potential to develop new pharmacotherapeutics to treat neuromuscular disorders that affect muscle strength.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    The aim of this study was to investigate the effects of an enzymatic removal of glycogen on excitation-contraction coupling in mechanically skinned fibres of rat fast-twitch muscles, with a focus on the changes in the function of Na+-K+-pump and ryanodine receptor (RyR). Glycogen present in the skinned fibres and binding to microsomes was removed using glucoamylase (GA). Exposure of whole muscle to 20 U mL-1 GA for 6 min resulted in a 72% decrease in the glycogen content. Six minutes of GA treatment led to an 18 and a 22% reduction in depolarization- and action potential-induced forces in the skinned fibres, respectively. There was a minor but statistically significant increase in the repriming period, most likely because of an impairment of the Na+-K+-pump function. GA treatment exerted no effect on the maximum Ca2+ release rate from the RyR in the microsomes and the myofibrillar Ca2+ sensitivity in the skinned fibres. These results indicate that reduced glycogen per se can decrease muscle performance due to the impairment of SR Ca2+ release and suggest that although Na+-K+-pump function is adversely affected by reduced glycogen, the extent of the impairment is not sufficient to reduce Ca2+ release from the sarcoplasmic reticulum. This study provides direct evidence that glycogen above a certain amount is required for the preservation of the functional events preceding Ca2+ release from the sarcoplasmic reticulum.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    肌膜连接膜(JM)上的CaV1.1L型钙通道(LTCCs)与肌浆网(SR)的ryanodine受体(RyRs)的紧密物理关联对于骨骼肌的兴奋-收缩偶联(ECC)至关重要。然而,JM靶向LTCC的分子机制尚未被探索。Junctophilin1(JP1)和JP2通过桥接肌膜和SR膜来稳定JM。这里,我们研究了JP在LTCC本地化和功能中的作用。培养的肌管中JP1或JP2的敲除抑制了LTCC在JM处的聚集,并抑制了诱发的Ca2瞬变,而不破坏JM结构。免疫共沉淀和GST下拉测定表明,JPs与CaV1.1的近端C末端的12-aa残基发生物理相互作用。缺乏包括跨膜结构域(JP1ΔCT)的C末端的JP1突变体与肌膜/T小管膜相互作用,但不与SR膜相互作用。该突变体在体内成年小鼠肌肉中的表达对内源性JPs产生了明显的负作用,在不破坏JM形态的情况下损害三联体的LTCC-RyR偶联,并显著减少Ca2+瞬变而不影响SRCa2+含量。此外,与对照组相比,JP1ΔCT表达的肌肉的收缩力急剧下降。一起来看,JP通过物理相互作用将LTCC招募到JM,并确保骨骼肌三联组处具有稳健的ECC。
    Close physical association of CaV1.1 L-type calcium channels (LTCCs) at the sarcolemmal junctional membrane (JM) with ryanodine receptors (RyRs) of the sarcoplasmic reticulum (SR) is crucial for excitation-contraction coupling (ECC) in skeletal muscle. However, the molecular mechanism underlying the JM targeting of LTCCs is unexplored. Junctophilin 1 (JP1) and JP2 stabilize the JM by bridging the sarcolemmal and SR membranes. Here, we examined the roles of JPs in localization and function of LTCCs. Knockdown of JP1 or JP2 in cultured myotubes inhibited LTCC clustering at the JM and suppressed evoked Ca2+ transients without disrupting JM structure. Coimmunoprecipitation and GST pull-down assays demonstrated that JPs physically interacted with 12-aa residues in the proximal C terminus of the CaV1.1. A JP1 mutant lacking the C terminus including the transmembrane domain (JP1ΔCT) interacted with the sarcolemmal/T-tubule membrane but not the SR membrane. Expression of this mutant in adult mouse muscles in vivo exerted a dominant-negative effect on endogenous JPs, impairing LTCC-RyR coupling at triads without disrupting JM morphology, and substantially reducing Ca2+ transients without affecting SR Ca2+ content. Moreover, the contractile force of the JP1ΔCT-expressed muscle was dramatically reduced compared with the control. Taken together, JPs recruit LTCCs to the JM through physical interaction and ensure robust ECC at triads in skeletal muscle.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    骨骼肌收缩是由兴奋收缩(EC)耦合过程中释放的Ca2增加引起的,EC偶联缺陷与人肌病有关。EC偶联需要横向小管膜中的电压感应二氢吡啶受体(DHPR)与肌浆网(SR)中的Ca2释放通道ryanodine受体1(RyR1)之间的通讯。Stac3蛋白(SH3和富含半胱氨酸的结构域3)是EC偶联装置的重要组成部分,人类STAC3中的突变会导致使人衰弱的美洲原住民肌病(NAM),但Stac3如何作用于DHPR和/或RyR1的性质是未知的。使用电子显微镜,电生理学,和斑马鱼肌纤维的动态成像,我们发现DHPR水平显著降低,功能,和stac3突变体的稳定性。此外,在没有咖啡因敏感性改变的情况下,stac3NAM肌纤维在广泛的浓度范围内表现出咖啡因诱导的Ca2+释放增加以及内部存储中的Ca2+增加,这与增加的SR腔Ca2+一致。这些发现定义了Stac3在EC偶联和人类疾病中的关键作用。
    Skeletal muscle contractions are initiated by an increase in Ca2+ released during excitation-contraction (EC) coupling, and defects in EC coupling are associated with human myopathies. EC coupling requires communication between voltage-sensing dihydropyridine receptors (DHPRs) in transverse tubule membrane and Ca2+ release channel ryanodine receptor 1 (RyR1) in the sarcoplasmic reticulum (SR). Stac3 protein (SH3 and cysteine-rich domain 3) is an essential component of the EC coupling apparatus and a mutation in human STAC3 causes the debilitating Native American myopathy (NAM), but the nature of how Stac3 acts on the DHPR and/or RyR1 is unknown. Using electron microscopy, electrophysiology, and dynamic imaging of zebrafish muscle fibers, we find significantly reduced DHPR levels, functionality, and stability in stac3 mutants. Furthermore, stac3NAM myofibers exhibited increased caffeine-induced Ca2+ release across a wide range of concentrations in the absence of altered caffeine sensitivity as well as increased Ca2+ in internal stores, which is consistent with increased SR luminal Ca2+ These findings define critical roles for Stac3 in EC coupling and human disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Ca2+从肌浆网释放的生理过程称为兴奋-收缩耦合;它是由动作电位引发的,动作电位深入肌纤维,在肌纤维中被二氢吡啶受体感知,电压传感L型Ca2+通道位于横小管上。电压诱导的二氢吡啶受体的构象变化激活了肌浆网的ryanodine受体Ca2释放通道。释放的Ca2+与肌钙蛋白C结合,使收缩粗细的细丝相互作用。随后通过专门的Ca2泵(SERCA)将Ca2转运回肌浆网,为肌肉准备一个新的收缩周期。尽管其他蛋白质参与兴奋-收缩偶联,上述机制强调了两个Ca2+通道(二氢吡啶受体和ryanodine受体)发挥的独特作用,SERCACa2泵和膜隔室的精致空间组织赋予了负责该机制的蛋白质以快速有效地发挥作用。过去二十年的研究揭示了正常条件下兴奋-收缩耦合的细节,而基因组学的进步有助于识别神经肌肉疾病患者新基因的突变。虽然现在很清楚,许多患有先天性肌肉疾病的患者在编码直接参与Ca2+稳态的蛋白质的基因中携带突变,显然,突变也存在于编码不被认为直接参与Ca2+调节的蛋白质的基因中。该领域正在进行的研究现在集中在理解单个突变的功能效应上,以及了解未明确位于肌浆网中的蛋白质的作用,这些蛋白质仍然参与Ca2调节或兴奋-收缩偶联。未来的主要挑战是识别可以通过小分子进行药理学操纵的药物靶标。最终目的是改善先天性肌肉疾病患者的肌肉功能和生活质量。这篇综述的目的是概述有关Ca2失调及其对先天性肌肉疾病患者肌肉功能的影响的最新发现,这是由于涉及兴奋-收缩耦合的蛋白质突变以及更广泛的Ca2稳态。
    The physiological process by which Ca2+ is released from the sarcoplasmic reticulum is called excitation-contraction coupling; it is initiated by an action potential which travels deep into the muscle fiber where it is sensed by the dihydropyridine receptor, a voltage sensing L-type Ca2+channel localized on the transverse tubules. Voltage-induced conformational changes in the dihydropyridine receptor activate the ryanodine receptor Ca2+ release channel of the sarcoplasmic reticulum. The released Ca2+ binds to troponin C, enabling contractile thick-thin filament interactions. The Ca2+ is subsequently transported back into the sarcoplasmic reticulum by specialized Ca2+ pumps (SERCA), preparing the muscle for a new cycle of contraction. Although other proteins are involved in excitation-contraction coupling, the mechanism described above emphasizes the unique role played by the two Ca2+ channels (the dihydropyridine receptor and the ryanodine receptor), the SERCA Ca2+ pumps and the exquisite spatial organization of the membrane compartments endowed with the proteins responsible for this mechanism to function rapidly and efficiently. Research over the past two decades has uncovered the fine details of excitation-contraction coupling under normal conditions while advances in genomics have helped to identify mutations in novel genes in patients with neuromuscular disorders. While it is now clear that many patients with congenital muscle diseases carry mutations in genes encoding proteins directly involved in Ca2+ homeostasis, it has become apparent that mutations are also present in genes encoding for proteins not thought to be directly involved in Ca2+ regulation. Ongoing research in the field now focuses on understanding the functional effect of individual mutations, as well as understanding the role of proteins not specifically located in the sarcoplasmic reticulum which nevertheless are involved in Ca2+ regulation or excitation-contraction coupling. The principal challenge for the future is the identification of drug targets that can be pharmacologically manipulated by small molecules, with the ultimate aim to improve muscle function and quality of life of patients with congenital muscle disorders. The aim of this review is to give an overview of the most recent findings concerning Ca2+ dysregulation and its impact on muscle function in patients with congenital muscle disorders due to mutations in proteins involved in excitation-contraction coupling and more broadly on Ca2+ homeostasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号