STIM1

STIM1
  • 文章类型: Journal Article
    广泛理解STIM1的构象变化,该构象变化将ER钙存储消耗的感知从STIMER腔结构域传递到STIM细胞质区域,并最终传递到质膜中的ORAI通道。然而,驱动构象变化的STIM腔结构域二聚化的结构基础已被证明是难以捉摸的。最近发表的一项研究通过分子动力学模拟来解决这个问题。该报告指出了可能是腔域二聚化界面的一部分的STIM残基,并提供了有关STIM腔结构域的扭转运动如何触发细胞质SOAR/CAD结构域从其静止系链释放到STIMCC1片段的意外见解。
    The conformational change in STIM1 that communicates sensing of ER calcium-store depletion from the STIM ER-luminal domain to the STIM cytoplasmic region and ultimately to ORAI channels in the plasma membrane is broadly understood. However, the structural basis for the STIM luminal-domain dimerization that drives the conformational change has proven elusive. A recently published study has approached this question via molecular dynamics simulations. The report pinpoints STIM residues that may be part of a luminal-domain dimerization interface, and provides unexpected insight into how torsional movements of the STIM luminal domains might trigger release of the cytoplasmic SOAR/CAD domain from its resting tethers to the STIM CC1 segments.
    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
    慢性肾功能衰竭(CRF)是一种具有独特挑战的无法治愈的疾病。贫血是影响透析患者的常见并发症。促红细胞生成素(EPO)用于治疗贫血,但可能会导致不良反应。我们调查了存储操作的钙通道(SOC)信号的遗传多态性,一个重要的促红细胞生成素激活途径,可能诱导肾功能衰竭患者的EPO抵抗。共选择108名终末期肾病(ESRD)患者进行本研究。根据其促红细胞生成素抵抗指数(ERI)将患者分为两组:ERI>10的39例患者和ERI<10的69例患者。在我们的研究中,我们选择了STIM1中的四个标记单核苷酸多态性(tSNP)和ORAI1中的五个。进行聚合酶链反应,与抗EPO抗性的基因分型相关。STIM1中具有rs1561876的AG基因型,ORAI1中的rs6486795的TC基因型以及ORAI1中的rs12320939的TG或GG基因型的患者与促红细胞生成素抵抗的风险增加有关。总的来说,我们报道了STIM1的遗传多态性与EPO抗性之间的中度显着关系。我们还报道了ORAI1的遗传多态性与EPO抗性之间的高度显着关系。STIM1的(A-A-G)单倍型和(G-T-G-T-A,G-C-G-C-G,或ORAI1的G-T-T-C-G)单倍型与EPO抗性显着相关。
    Chronic renal failure (CRF) is an incurable disease with unique challenges. Anemia is a frequent complication affecting dialysis patients. Erythropoietin (EPO) is used to treat anemia, but a poor response may result. We investigated genetic polymorphisms of store-operated calcium channel (SOC) signaling, an important erythropoietin-activated pathway that may induce EPO resistance in patients with renal failure. A total of 108 end stage renal disease (ESRD) patients were selected for this study. Patients were divided into two groups according to their erythropoietin resistance index (ERI): 39 patients with an ERI>10 and 69 patients with an ERI<10. We selected four tagging single nucleotide polymorphisms (tSNPs) in STIM1 and five in ORAI1 in our study. A polymerase chain reaction was performed, and genotyping against EPO resistance was correlated. Patients with the AG genotype of rs1561876 in STIM1, the TC genotype of rs6486795 in ORAI1, and the TG or GG genotypes of rs12320939 in ORAI1 were associated with an increased risk of erythropoietin resistance. Overall, we reported a moderately significant relationship between genetic polymorphisms of STIM1 and EPO resistance. We also reported a highly significant relationship between genetic polymorphisms of ORAI1 and EPO resistance. The (A-A-G) haplotype of STIM1 and the (G-T-G-T-A, G-C-G-C-G, or G-T-T-C-G) haplotypes of ORAI1 were significantly associated with EPO resistance.
    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
    根据降解底物的类型,自噬分为非选择性或选择性。内质网(ER)-吞噬是选择性自噬的一种形式,用于将ER-驻留蛋白转运至自体溶酶体。FAM134B,具有序列相似性134的家族成员是众所周知的ER-吞噬受体。FAM134B的功能障碍导致几种疾病,包括病毒感染,炎症,神经退行性疾病和癌症,表明FAM134B在各种细胞内功能中起着至关重要的作用。然而,FAM134B介导的ER-phagy如何调节细胞内功能尚不清楚。在这项研究中,我们发现哺乳动物细胞中FAM134B敲低可加速细胞增殖。FAM134B敲低增加了STIM1的蛋白质量,STIM1是一种ERCa2传感器蛋白,介导了G1到S相转变中的存储操作的Ca2进入(SOCE)。FAM134B通过其C端胞质区域与STIM1结合。FAM134B敲除减少STIM1从ER到自体溶酶体的转运。最后,FAM134B敲除加速了G1向S期的转变。这些结果表明FAM134B可能通过ER-吞噬降解STIM1而参与细胞增殖。
    Autophagy is classified as non-selective or selective depending on the types of degrading substrates. Endoplasmic reticulum (ER)-phagy is a form of selective autophagy for transporting the ER-resident proteins to autolysosomes. FAM134B, a member of the family with sequence similarity 134, is a well-known ER-phagy receptor. Dysfunction of FAM134B results in several diseases including viral infection, inflammation, neurodegenerative disorder and cancer, indicating that FAM134B has crucial roles in various kinds of intracellular functions. However, how FAM134B-mediated ER-phagy regulates intracellular functions is not well understood. In this study, we found that FAM134B knockdown in mammalian cells accelerated cell proliferation. FAM134B knockdown increased the protein amount of STIM1, an ER Ca2+ sensor protein mediating the store-operated Ca2+ entry (SOCE) involved in G1 to S phase transition. FAM134B bound to STIM1 through its C-terminal cytosolic region. FAM134B knockdown reduced transport of STIM1 from the ER to autolysosomes. Finally, FAM134B knockdown accelerated G1 to S phase transition. These results suggest that FAM134B is involved in cell proliferation possibly through degradation of STIM1 via ER-phagy.
    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
    在细胞环境中,钙离子(Ca2+)的振荡与各种生理过程密切相关,如细胞增殖,新陈代谢,和生存。基质相互作用分子1(STIM1)蛋白在储存操作的钙进入(SOCE)过程中形成关键的调节成分。STIM1的结构属性对其功能至关重要,包含位于内质网(ER)腔和细胞质中的不同结构域。管腔内域能够及时检测不断下降的Ca2+浓度,提示激活细胞质结构域的结构修饰。这个激活的细胞质结构域经历构象改变并与膜成分接合,打开促进Ca2+从细胞外环境流入的通道。鉴于其多个领域和相互作用机制,STIM1在细胞生物学中起着基础性作用。本文综述了受STIM1结构和功能启发的光遗传学工具的设计。这些工具为研究和操纵具有精确时空控制的细胞内Ca2信号提供了开创性的方法。我们进一步探索这些工具的实际应用,跨越基础科学研究,临床研究,以及他们转化研究的潜力。
    In cellular contexts, the oscillation of calcium ions (Ca2+) is intricately linked to various physiological processes, such as cell proliferation, metabolism, and survival. Stromal interaction molecule 1 (STIM1) proteins form a crucial regulatory component in the store-operated calcium entry process. The structural attributes of STIM1 are vital for its functionality, encompassing distinct domains situated in the endoplasmic reticulum lumen and the cytoplasm. The intraluminal domain enables the timely detection of diminishing Ca2+ concentrations, prompting structural modifications that activate the cytoplasmic domain. This activated cytoplasmic domain undergoes conformational alterations and engages with membrane components, opening a channel that facilitates the influx of Ca2+ from the extracellular environment. Given its multiple domains and interaction mechanisms, STIM1 plays a foundational role in cellular biology. This review focuses on the design of optogenetic tools inspired by the structure and function of STIM1. These tools offer a groundbreaking approach for studying and manipulating intracellular Ca2+ signaling with precise spatiotemporal control. We further explore the practical applications of these tools, spanning fundamental scientific research, clinical studies, and their potential for translational research.
    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
    感觉到内质网(ER)钙(Ca2+)的降低,STIM1介导了一种普遍存在的Ca2流入过程,称为存储操作的Ca2进入(SOCE)。STIM1功能失调或SOCE异常与自身免疫性疾病密切相关。动脉粥样硬化,和各种形式的癌症。因此,揭示翻译后修饰的分子复杂性,如氧化,STIM1的功能至关重要。在最近的蛋白质组学筛查中,我们鉴定了三种蛋白质二硫键异构酶(PDIs)-脯氨酸4-羟化酶亚基β(P4HB),蛋白质二硫键异构酶A3(PDIA3),和含硫氧还蛋白结构域的蛋白5(TXNDC5)-作为STIM1的ER腔相互作用物。这里,我们证明了这些PDI与STIM1和STIM2动态关联。STIM1(STIM1-2CA)的两个保守半胱氨酸残基的突变降低了其在细胞和原位的Ca2亲和力。PDIA3或P4HB的敲除增加了野生型STIM1的Ca2亲和力,同时对STIM1-2CA突变体没有影响,表明PDIA3和P4HB通过作用于ER-管腔半胱氨酸残基来调节STIM1的Ca2+亲和力。这种STIM1的Ca2+敏感性的调制通过Ca2+成像实验进一步证实,这表明,这两个PDI的敲低不会影响STIM1介导的SOCE在完全存储耗尽时,但会导致SOCE振幅在部分存储耗尽时增强。因此,P4HB和PDIA3通过微调其Ca2结合亲和力动态调节STIM1激活,调节激活的STIM1水平以响应生理线索。本文报道的STIM1介导的Ca2+信号传导和氧化还原反应之间的协调可能对细胞生理学和病理学有影响。
    Sensing the lowering of endoplasmic reticulum (ER) calcium (Ca2+), STIM1 mediates a ubiquitous Ca2+ influx process called the store-operated Ca2+ entry (SOCE). Dysregulated STIM1 function or abnormal SOCE is strongly associated with autoimmune disorders, atherosclerosis, and various forms of cancers. Therefore, uncovering the molecular intricacies of post-translational modifications, such as oxidation, on STIM1 function is of paramount importance. In a recent proteomic screening, we identified three protein disulfide isomerases (PDIs)-Prolyl 4-hydroxylase subunit beta (P4HB), protein disulfide-isomerase A3 (PDIA3), and thioredoxin domain-containing protein 5 (TXNDC5)-as the ER-luminal interactors of STIM1. Here, we demonstrated that these PDIs dynamically associate with STIM1 and STIM2. The mutation of the two conserved cysteine residues of STIM1 (STIM1-2CA) decreased its Ca2+ affinity both in cellulo and in situ. Knockdown of PDIA3 or P4HB increased the Ca2+ affinity of wild-type STIM1 while showing no impact on the STIM1-2CA mutant, indicating that PDIA3 and P4HB regulate STIM1\'s Ca2+ affinity by acting on ER-luminal cysteine residues. This modulation of STIM1\'s Ca2+ sensitivity was further confirmed by Ca2+ imaging experiments, which showed that knockdown of these two PDIs does not affect STIM1-mediated SOCE upon full store depletion but leads to enhanced SOCE amplitudes upon partial store depletion. Thus, P4HB and PDIA3 dynamically modulate STIM1 activation by fine-tuning its Ca2+ binding affinity, adjusting the level of activated STIM1 in response to physiological cues. The coordination between STIM1-mediated Ca2+ signaling and redox responses reported herein may have implications for cell physiology and pathology.
    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
    血管平滑肌细胞(VSMC)的收缩功能通常随着年龄的增长而发生显著变化,导致严重的血管老化相关疾病。基质相互作用分子1(STIM1)在年龄介导的Ca2信号传导和血管收缩中的确切作用和机制尚不清楚。使用多肌电图系统研究了STIM1与年龄相关的血管功能障碍之间的联系,免疫组织化学分析,蛋白质印迹,和SA-β-gal染色。结果显示,胸主动脉中的血管收缩反应,肾内动脉,冠状动脉随着年龄的增长而减少。肾内和冠状动脉中的STIM1敲除降低了年轻小鼠的血管张力,而在胸主动脉中没有观察到变化。在使用硝苯地平的STIM1敲除组中,血管张力显着降低。在胸主动脉中,使用硝苯地平和thapsigargin后,血管收缩随着年龄的增长而显着降低,而在STIM1敲除后几乎消失。衰老VSMC的比例在老年小鼠中显着增加,在sm-STIM1KO老年小鼠中进一步增加。此外,衰老标志物p21、p16和IL-6的表达随着年龄的增长而显著增加,在STIM1敲低年龄组中p21表达进一步增加,但不是p16或IL-6。这些发现表明,不同的动脉表现出不同的器官特异性特征,并且STIM1下调可能通过p21途径的激活导致与年龄相关的血管收缩功能障碍。
    The contractile function of vascular smooth muscle cells (VSMCs) typically undergoes significant changes with advancing age, leading to severe vascular aging-related diseases. The precise role and mechanism of stromal interaction molecule-1 (STIM1) in age-mediated Ca2+ signaling and vasocontraction remain unclear. The connection between STIM1 and age-related vascular dysfunction was investigated using a multi-myograph system, immunohistochemical analysis, protein blotting, and SA-β-gal staining. Results showed that vasoconstrictor responses in the thoracic aorta, intrarenal artery, and coronary artery decreased with age. STIM1 knockdown in the intrarenal and coronary arteries reduced vascular tone in young mice, while no change was observed in the thoracic aorta. A significant reduction in vascular tone occurred in the STIM1 knockout group with nifedipine. In the thoracic aorta, vasoconstriction significantly decreased with age following the use of nifedipine and thapsigargin and almost disappeared after STIM1 knockdown. The proportion of senescent VSMCs increased significantly in aged mice and further increased in sm-STIM1 KO aged mice. Moreover, the expression of senescence markers p21, p16, and IL-6 significantly increased with age, with p21 expression further increased in the STIM1 knockdown aged group, but not p16 or IL-6. These findings indicate that different arteries exhibit distinct organ-specific features and that STIM1 downregulation may contribute to age-related vasoconstrictive dysfunction through activation of the p21 pathway.
    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
    肢带肌营养不良R1/2A(LGMDR1/2A)是由编码骨骼肌特异性Calpain3的CAPN3基因突变引起的,Ca2+-依赖性蛋白酶。钙蛋白酶3在三联体中的定位表明它有助于Ca2稳态。通过活细胞Ca2+测量,肌肉力学,免疫荧光,Capn3缺陷(C3KO)和野生型(WT)小鼠的电子显微镜(EM),我们确定了钙蛋白酶3的丢失是否改变了储存操作的钙输入(SOCE)活性。直接的Ca2流入测量显示Capn3的损失引起静息SOCE升高和静息胞浆Ca2增加,由EM观察到的钙进入单位(CEU)的高发生率支持。对C3KO和WT小鼠进行一次跑步机跑步以引起SOCE。在跑步机后运行的1HR内,在重复刺激期间,C3KO小鼠在指长伸肌中的力产生减少,而在指短屈肌肌纤维中的Ca2瞬变衰减更大。C3KO小鼠运动诱导的SOCE激活受损的惊人证据包括关键SOCE蛋白的共定位不良,基质相互作用分子1(STIM1)和ORAI1,并伴有C3KO肌肉中CEU的消失。这些结果表明,钙蛋白酶3是骨骼肌中SOCE的关键调节剂,并将SOCE失调鉴定为LGMDR1/2A病理的促成因素。
    Limb-Girdle Muscular Dystrophy R1/2A (LGMD R1/2A) is caused by mutations in the CAPN3 gene encoding Calpain 3, a skeletal-muscle specific, Ca2+-dependent protease. Localization of Calpain 3 within the triad suggests it contributes to Ca2+ homeostasis. Through live-cell Ca2+ measurements, muscle mechanics, immunofluorescence, and electron microscopy (EM) in Capn3 deficient (C3KO) and wild-type (WT) mice, we determined whether loss of Calpain 3 altered Store-Operated Calcium Entry (SOCE) activity. Direct Ca2+ influx measurements revealed loss of Capn3 elicits elevated resting SOCE and increased resting cytosolic Ca2+, supported by high incidence of calcium entry units (CEUs) observed by EM. C3KO and WT mice were subjected to a single bout of treadmill running to elicit SOCE. Within 1HR post-treadmill running, C3KO mice exhibited diminished force production in extensor digitorum longus muscles and a greater decay of Ca2+ transients in flexor digitorum brevis muscle fibers during repetitive stimulation. Striking evidence for impaired exercise-induced SOCE activation in C3KO mice included poor colocalization of key SOCE proteins, stromal-interacting molecule 1 (STIM1) and ORAI1, combined with disappearance of CEUs in C3KO muscles. These results demonstrate that Calpain 3 is a key regulator of SOCE in skeletal muscle and identify SOCE dysregulation as a contributing factor to LGMD R1/2A pathology.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    暂无摘要。
    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
    瞬时受体电位规范(TRPC)通道是钙通道,在视网膜中具有不同的表达谱和生理意义。由色素性视网膜炎(RP)引起的光感受器变性的大鼠视网膜的神经元和神经胶质细胞表现出高于健康视网膜中检测到的基础钙水平。内部视网膜细胞是最后退化的细胞,负责维持视觉皮层的活动,即使在光感受器完全丧失之后。我们认为TRPC1和TRPC5通道可能与高钙水平和内部视网膜变性的延迟有关。已知TRPC1在神经变性过程中介导保护作用,而TRPC5促进细胞死亡。为了理解这些渠道在RP中的含义,通过免疫荧光和邻近连接试验检测了TRPC1和TRPC5在健康视网膜(Sprague-Dawley大鼠)和退化视网膜(P23H-1,一种RP模型)中的共定位和随后的物理相互作用.在TRPC1和TRPC5物理相互作用的所有动物的最内层视网膜中存在重叠信号。这种相互作用随着光感受器损失的进展而显著增加。在健康和受损的视网膜中,两个通道都充当TRPC1/5异聚体,具有明显的TRPC1功能,以响应视网膜变性机制。此外,我们的研究结果支持TRPC5通道在Müller和视网膜神经节细胞中也与STIM1协同发挥作用.这些结果表明,TRPC1/5异聚体的增加可能有助于在外部视网膜变性过程中减缓内部视网膜的变性。
    Transient receptor potential canonical (TRPC) channels are calcium channels with diverse expression profiles and physiological implications in the retina. Neurons and glial cells of rat retinas with photoreceptor degeneration caused by retinitis pigmentosa (RP) exhibit basal calcium levels that are above those detected in healthy retinas. Inner retinal cells are the last to degenerate and are responsible for maintaining the activity of the visual cortex, even after complete loss of photoreceptors. We considered the possibility that TRPC1 and TRPC5 channels might be associated with both the high calcium levels and the delay in inner retinal degeneration. TRPC1 is known to mediate protective effects in neurodegenerative processes while TRPC5 promotes cell death. In order to comprehend the implications of these channels in RP, the co-localization and subsequent physical interaction between TRPC1 and TRPC5 in healthy retina (Sprague-Dawley rats) and degenerating (P23H-1, a model of RP) retina were detected by immunofluorescence and proximity ligation assays. There was an overlapping signal in the innermost retina of all animals where TRPC1 and TRPC5 physically interacted. This interaction increased significantly as photoreceptor loss progressed. Both channels function as TRPC1/5 heteromers in the healthy and damaged retina, with a marked function of TRPC1 in response to retinal degenerative mechanisms. Furthermore, our findings support that TRPC5 channels also function in partnership with STIM1 in Müller and retinal ganglion cells. These results suggest that an increase in TRPC1/5 heteromers may contribute to the slowing of the degeneration of the inner retina during the outer retinal degeneration.
    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
    储存操作的钙进入(SOCE)在维持细胞钙稳态中起着至关重要的作用。这种机制涉及蛋白质,如基质相互作用分子1(STIM1)和ORAI1。编码这些蛋白质的基因突变,特别是STIM1,可以导致各种疾病,包括与严重联合免疫缺陷相关的CRAC信道病。在这里,我们描述了一个新的纯合突变,NM_003156c.792-3C>G,在患有CRAC信道病临床特征的患者的STIM1中,包括免疫系统缺陷和肌肉无力.功能分析揭示了患者细胞中三种不同的剪接形式:野生型,外显子7跳绳,和内含子保留。钙内流分析显示患者细胞中SOCE受损,表明STIM1功能丧失。我们开发了一种改善STIM1剪接的反义寡核苷酸治疗,并强调了其作为治疗方法的潜力。我们的发现提供了对STIM1突变的复杂影响的见解,并阐明了患者多方面的临床表现。
    Store-operated calcium entry (SOCE) plays a crucial role in maintaining cellular calcium homeostasis. This mechanism involves proteins, such as stromal interaction molecule 1 (STIM1) and ORAI1. Mutations in the genes encoding these proteins, especially STIM1, can lead to various diseases, including CRAC channelopathies associated with severe combined immunodeficiency. Herein, we describe a novel homozygous mutation, NM_003156 c.792-3C > G, in STIM1 in a patient with a clinical profile of CRAC channelopathy, including immune system deficiencies and muscle weakness. Functional analyses revealed three distinct spliced forms in the patient cells: wild-type, exon 7 skipping, and intronic retention. Calcium influx analysis revealed impaired SOCE in the patient cells, indicating a loss of STIM1 function. We developed an antisense oligonucleotide treatment that improves STIM1 splicing and highlighted its potential as a therapeutic approach. Our findings provide insights into the complex effects of STIM1 mutations and shed light on the multifaceted clinical presentation of the patient.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号