CLCA2

CLCA2
  • 文章类型: Journal Article
    氯通道附件2(CLCA2)是一种跨膜蛋白,促进角质形成细胞的粘附和它们在高渗应激下的存活。在这里,我们显示CLCA2通过细胞外囊泡转运到角质形成细胞的细胞核。核定位在功能上是相关的,因为野生型CLCA2,但不是缺乏核定位信号的突变体,抑制角质形成细胞的迁移并保护它们免受高渗应激诱导的细胞死亡。在细胞核中,CLCA2结合并激活β-catenin,导致Wnt靶基因的表达增强。基于质谱的相互作用筛选和功能拯救研究确定RNA结合蛋白3是核CLCA2的关键效应物。这在体内可能是相关的,因为两种蛋白质共定位在人表皮中。一起,这些结果确定了在稳态和应激条件下,角质形成细胞中CLCA2的意外核功能,并表明细胞外囊泡及其核运输在关键细胞活动控制中的作用。
    Chloride channel accessory 2 (CLCA2) is a transmembrane protein, which promotes adhesion of keratinocytes and their survival in response to hyperosmotic stress. Here we show that CLCA2 is transported to the nucleus of keratinocytes via extracellular vesicles. The nuclear localization is functionally relevant, since wild-type CLCA2, but not a mutant lacking the nuclear localization signal, suppressed migration of keratinocytes and protected them from hyperosmotic stress-induced cell death. In the nucleus, CLCA2 bound to and activated β-catenin, resulting in enhanced expression of Wnt target genes. Mass-spectrometry-based interaction screening and functional rescue studies identified RNA binding protein 3 as a key effector of nuclear CLCA2. This is of likely relevance in vivo because both proteins co-localize in the human epidermis. Together, these results identify an unexpected nuclear function of CLCA2 in keratinocytes under homeostatic and stress conditions and suggest a role of extracellular vesicles and their nuclear transport in the control of key cellular activities.
    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
    细胞衰老,不可逆转的生长停滞状态,与各种年龄相关的病理有关,包括皮肤老化。在这项研究中,我们研究了CLCA2的作用,CLCA2是一种钙激活的氯离子通道辅助蛋白,细胞衰老及其对皮肤老化的影响。利用UVB和Nutlin3a诱导的衰老模型,我们观察到CLCA2在转录组和蛋白质组水平的上调,表明它参与了衰老途径。进一步的分析表明,CLCA2的耗竭导致衰老加速,以经典的衰老标记和独特的分泌组特征为特征。在3D皮肤等效模型中,用CLCA2敲低成纤维细胞构建的SE表现出让人联想到老化皮肤的特征,强调CLCA2在维持皮肤稳态中的重要性。我们的发现强调了CLCA2作为细胞衰老的新型调节剂及其对皮肤衰老机制的潜在影响。
    Cellular senescence, a state of irreversible growth arrest, is implicated in various age-related pathologies, including skin aging. In this study, we investigated the role of CLCA2, a calcium-activated chloride channel accessory protein, in cellular senescence and its implications for skin aging. Utilizing UVB and Nutlin3a-induced senescence models, we observed the upregulation of CLCA2 at both transcriptomic and proteomic levels, suggesting its involvement in senescence pathways. Further analysis revealed that the depletion of CLCA2 led to accelerated senescence onset, characterized by classic senescence markers and a unique secretome profile. In 3D skin equivalent models, SEs constructed with CLCA2 knockdown fibroblasts exhibited features reminiscent of aged skin, underscoring the importance of CLCA2 in maintaining skin homeostasis. Our findings highlight CLCA2 as a novel regulator of cellular senescence and its potential implications for skin aging mechanisms.
    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
    背景:过早卵巢功能不全(POI)是一种导致女性不孕的严重疾病。基因突变是引起POI的重要因素。据报道,TP63截短突变可通过增加生殖细胞凋亡而导致POI,然而,是什么因素介导这种细胞凋亡仍不清楚。
    方法:选取北京妇产医院93例POI患者,首都医科大学。对每位患者进行全外显子组测序(WES)。Sanger测序用于确认潜在的致病遗传变异。进行小基因测定以确定TP63变体的剪接效应。构建了TP63截短质粒。实时定量PCR,蛋白质印迹分析,双荧光素酶报告分析,免疫荧光染色,和细胞凋亡测定用于研究TP63截短突变导致POI的潜在机制。
    结果:通过93例散发性POI患者的WES,我们在TP63基因的剪接位点上发现了一个14bp的缺失.小基因分析表明,在TP63mRNA剪接过程中,14bp缺失变体导致外显子13跳跃,导致产生截短的TP63蛋白(TP63-mut)。过表达TP63-mut加速细胞凋亡。机械上,与TP63野生型蛋白相比,TP63-mut蛋白可以结合CLCA2的启动子区并激活CLCA2的转录数倍。使用特定的小干扰RNA(siRNA)沉默CLCA2或使用KU55933抑制剂抑制共济失调毛细血管扩张突变(ATM)途径减弱了由TP63-mut蛋白表达引起的细胞凋亡。
    结论:我们的发现揭示了CLCA2在POI发病机制中介导细胞凋亡的关键作用,并提示CLCA2是POI的潜在治疗靶点。
    BACKGROUND: Premature ovarian insufficiency (POI) is a severe disorder leading to female infertility. Genetic mutations are important factors causing POI. TP63-truncating mutation has been reported to cause POI by increasing germ cell apoptosis, however what factors mediate this apoptosis remains unclear.
    METHODS: Ninety-three patients with POI were recruited from Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Whole-exome sequencing (WES) was performed for each patient. Sanger sequencing was used to confirm potential causative genetic variants. A minigene assay was performed to determine splicing effects of TP63 variants. A TP63-truncating plasmid was constructed. Real-time quantitative PCR, western blot analyses, dual luciferase reporter assays, immunofluorescence staining, and cell apoptosis assays were used to study the underlying mechanism of a TP63-truncating mutation causing POI.
    RESULTS: By WES of 93 sporadic patients with POI, we found a 14-bp deletion covering the splice site in the TP63 gene. A minigene assay demonstrated that the 14-bp deletion variant led to exon 13 skipping during TP63 mRNA splicing, resulting in the generation of a truncated TP63 protein (TP63-mut). Overexpression of TP63-mut accelerated cell apoptosis. Mechanistically, the TP63-mut protein could bind to the promoter region of CLCA2 and activate the transcription of CLCA2 several times compared to that of the TP63 wild-type protein. Silencing CLCA2 using a specific small interfering RNA (siRNA) or inhibiting the Ataxia Telangiectasia Mutated (ATM) pathway using the KU55933 inhibitor attenuated cell apoptosis caused by TP63-mut protein expression.
    CONCLUSIONS: Our findings revealed a crucial role for CLCA2 in mediating apoptosis in POI pathogenesis, and suggested that CLCA2 is a potential therapeutic target for POI.
    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
    人类CLCA2通过与Orai1和STIM1相互作用来调节储存操作的钙进入(SOCE)。其表达为943aa型I跨膜蛋白,其在氨基酸708处被切割以产生可扩散的100kDa产物。N末端胞外域包含具有保守的HEXXH锌结合基序的水解酶样亚结构域,该基序被提议以自身蛋白水解方式切割前体。这里,我们检验了这一假设及其与SOCE的联系。我们首先研究了在分离的膜中,然后在纯化的蛋白质系统中进行自切割的条件。切割是锌依赖性的,并通过将HEXXH基序中的E突变为Q而废除,E165Q.切割效率随着CLCA2浓度的增加而增加,暗示它发生在反式。因此,通过共转染的野生型CLCA2切割E165Q突变体。此外,具有不同表位标签的CLCA2前体共免疫沉淀。在利用免疫纯化蛋白酶和靶标的无膜系统中,除非靶标首先变性,否则不会发生切割,暗示膜提供必要的结构或构象线索。出乎意料的是,裂解引起构象改变:免疫沉淀前体的N-末端抗体不能沉淀N-末端产物,除非产物首先用离子去污剂变性。E165Q突变消除了由野生型CLCA2引起的SOCE的刺激,确立了金属蛋白酶活性是该调节功能所必需的。
    Human CLCA2 regulates store-operated calcium entry (SOCE) by interacting with Orai1 and STIM1. It is expressed as a 943aa type I transmembrane protein that is cleaved at amino acid 708 to produce a diffusible 100 kDa product. The N-terminal ectodomain contains a hydrolase-like subdomain with a conserved HEXXH zinc-binding motif that is proposed to cleave the precursor autoproteolytically. Here, we tested this hypothesis and its link to SOCE. We first studied the conditions for autocleavage in isolated membranes and then in a purified protein system. Cleavage was zinc-dependent and abolished by mutation of the E in the HEXXH motif to Q, E165Q. Cleavage efficiency increased with CLCA2 concentration, implying that it occurs in trans. Accordingly, the E165Q mutant was cleaved by co-transfected wildtype CLCA2. Moreover, CLCA2 precursors with different epitope tags co-immunoprecipitated. In a membrane-free system utilizing immunopurified protease and target, no cleavage occurred unless the target was first denatured, implying that membranes provide essential structural or conformational cues. Unexpectedly, cleavage caused a conformational shift: an N-terminal antibody that immunoprecipitated the precursor failed to precipitate the N-terminal product unless the product was first denatured with an ionic detergent. The E165Q mutation abolished the stimulation of SOCE caused by wildtype CLCA2, establishing that the metalloprotease activity is required for this regulatory function.
    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
    宫颈癌是全球范围内威胁女性身心健康的重要恶性肿瘤。作为一种新的钙激活氯离子通道蛋白,钙激活氯通道(CLCA2)在肿瘤发生、发展中起着重要作用。但其在宫颈癌中的作用及确切调控机制尚不清楚。在我们的研究中,我们发现CLCA2在宫颈癌细胞中显著降低,CLCA2的过表达抑制细胞增殖,移民和入侵,促进宫颈癌细胞凋亡,CLCA2通过p38/JNK/ERK途径抑制EMT(上皮-间质转化)。体内结果与体外结果一致。总之,CLCA2的过表达在体内和体外抑制了宫颈癌的进展。这可能为CLCA2作为宫颈癌临床诊断和预后的新指标或作为潜在的药物治疗靶点提供理论依据。
    Cervical cancer is an important malignant tumor threatening the physical and mental health of women in the world. As a new calcium activated chloride channel protein, calcium activated chloride channel (CLCA2) plays an important role in tumorigenesis and development. But its role and exact regulatory mechanism in cervical cancer are still unclear. In our study, we found CLCA2 was significantly decreased in cervical cancer cells, and overexpression of CLCA2 inhibited the proliferation, migration and invasion, and promotes apoptosis of cervical cancer cells, and CLCA2 inhibited EMT (Epithelial-mesenchymal transition) through an p38 / JNK / ERK pathway. The results in vivo were consistent with those in vitro. In conclusion, overexpression of CLCA2 inhibited the progression of cervical cancer in vivo and in vitro. This may provide a theoretical basis for CLCA2 as a new indicator of clinical diagnosis and prognosis of cervical cancer or as a potential target of drug therapy.
    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
    背景:进行性心脏传导缺损(PCCD),也被称为Lenegre-Lev病,是最常见的心脏传导异常之一。先前的研究已经在一个有PCCD病史的68人家庭中筛选了导致心脏传导阻滞的已知突变位点,没有发现突变.
    目的:筛选PCCD家族致病基因并研究其与心脏传导阻滞疾病相关基因突变的功能。
    方法:对两名PCCD患者和一名非PCCD家族成员进行全外显子组测序(WES),以寻找相关致病基因。经过家庭共同隔离和初步功能分析,我们鉴定了突变基因CLCA2。为了研究这个基因的功能,我们使用CRISPR-Cas9技术构建了突变基因小鼠,基因型验证后进行心电图监测。
    结果:鉴定了CLCA2c.G1725T突变,并与表型共分离。分析表明,CLCA2c.G1725T突变是有害的,主要影响蛋白质的糖基化。免疫荧光染色显示CLCA2在窦房结(SAN)组织中高表达。对小鼠的心电图监测显示,CLCA2点突变可引起轻度传导阻滞和异位起搏器。
    结论:我们的研究结果表明,CLCA2基因的一个新的杂合错义突变c.G1725T可能与心脏传导阻滞疾病相关,该基因的突变可能导致窦房结病变和传导阻滞。
    BACKGROUND: Progressive cardiac conduction defect (PCCD), also known as Lenegre-Lev disease, is one of the most common heart conduction abnormalities. Previous studies have screened for known mutation sites that cause heart block in a 68-person family with a history of PCCD, revealed no mutations.
    OBJECTIVE: To screen pathogenic genes of the PCCD family and to study the function of the gene mutations related to heart block diseases.
    METHODS: Whole exome sequencing (WES) was performed on two PCCD patients and one non-PCCD family member to find the related pathogenic gene. After family co-segregation and preliminary functional analysis, we identified the mutant gene CLCA2. To study the function of this gene, we constructed mutant-gene mice using CRISPR-Cas9 technology, and electrocardiogram monitoring was performed after genotype verification.
    RESULTS: The CLCA2 c.G1725T mutation was identified and co-segregated with the phenotype. The analysis showed that the CLCA2 c.G1725T mutation is harmful and mainly affects protein glycosylation. Immunofluorescence staining revealed that CLCA2 was highly expressed in the sinoatrial node (SAN) tissues. Electrocardiogram monitoring of the mice revealed that CLCA2 point mutations induced mild conduction block and ectopic pacemakers.
    CONCLUSIONS: Our findings indicate that a novel heterozygous missense mutation c.G1725T of the CLCA2 gene may be associated with heart block disease and the mutation in this gene may lead to sinus node lesions and conduction blocking.
    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
    Prostate cancer (PCa) is the most common cancer among men. Metabolic syndrome (MeS) is associated with increased PCa aggressiveness and recurrence. Previously, we proposed C-terminal binding protein 1 (CTBP1), a transcriptional co-repressor, as a molecular link between these two conditions. Notably, CTBP1 depletion decreased PCa growth in MeS mice. The aim of this study was to investigate the molecular mechanisms that explain the link between MeS and PCa mediated by CTBP1. We found that CTBP1 repressed chloride channel accessory 2 (CLCA2) expression in prostate xenografts developed in MeS animals. CTBP1 bound to CLCA2 promoter and repressed its transcription and promoter activity in PCa cell lines. Furthermore, we found that CTBP1 formed a repressor complex with ZEB1, EP300 and HDACs that modulates the CLCA2 promoter activity. CLCA2 promoted PCa cell adhesion inhibiting epithelial-mesenchymal transition (EMT) and activating CTNNB1 together with epithelial marker (CDH1) induction, and mesenchymal markers (SNAI2 and TWIST1) repression. Moreover, CLCA2 depletion in PCa cells injected subcutaneously in MeS mice increased the circulating tumor cells foci compared to control. A microRNA (miRNA) expression microarray from PCa xenografts developed in MeS mice, showed 21 miRNAs modulated by CTBP1 involved in angiogenesis, extracellular matrix organization, focal adhesion and adherents junctions, among others. We found that miR-196b-5p directly targets CLCA2 by cloning CLCA2 3\'UTR and performing reporter assays. Altogether, we identified a new molecular mechanism to explain PCa and MeS link based on CLCA2 repression by CTBP1 and miR-196b-5p molecules that might act as key factors in the progression onset of this 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
    背景:据报道,CLCA2是一种肿瘤抑制因子,在乳腺癌中失调。然而,其在鼻咽癌肿瘤生长和转移中的作用鲜有报道。在这项研究中,我们研究了CLCA2影响NPC的功能和分子机制。
    方法:通过实时PCR(RT-PCR)检测人NPC细胞系和组织中CLCA2的表达,Westernblot和IHC。CLCA2在增殖中的生物学作用,在5-8F中评估了NPC细胞系的迁移和侵袭,S18、S26和SUNE-1细胞。细胞活力,通过MTS在体外评估迁移和侵袭,集落形成和transwell分析,分别。CLCA2在NPC的生长和转移中通过NPC异种移植肿瘤生长在体内进行评估,小鼠肺转移模型和贲门淋巴结转移模型。
    结果:CLCA2过表达显著降低增殖,NPC细胞的迁移和侵袭。相比之下,CLCA2的敲低引起相反的效果。CLCA2过表达抑制异种移植瘤生长和肺,在体内的the淋巴结(LN)转移。CLCA2通过抑制NPC细胞上皮间质转化(EMT)和激活FAK/ERK1/2信号通路抑制肿瘤转移。143例鼻咽癌标本的免疫组织化学染色显示CLCA2表达是一个独立的,患者总生存期和无远处转移生存期的有利预后因素。此外,抑制FAK和ERK1/2可逆转CLCA2沉默诱导的肿瘤细胞迁移。此外,针对氯通道的抑制剂抑制了NPC细胞的迁移,而CLCA2的存在可能会增强NPC细胞的迁移。
    结论:CLCA2抑制NPC增殖,迁移,通过抑制FAK/ERK信号传导进行侵袭和上皮间质转化。
    BACKGROUND: CLCA2 was reported as a tumor suppressor and disregulated in breast cancer. However, its function in tumor growth and metastasis in NPC has rarely been reported. In this study, we investigated the functional and molecular mechanisms by which CLCA2 influences NPC.
    METHODS: CLCA2 expression in human NPC cell lines and tissues was examined via real-time PCR (RT-PCR), Western blot and IHC. The biological roles of CLCA2 in proliferative, migration and invasion of NPC cell lines was evaluated in 5-8F, S18, S26 and SUNE-1 cells. Cell viability, migration and invasion were assessed in vitro by MTS, colony formation and transwell assay, respectively. CLCA2 in growth and metastasis of NPC were evaluated in vivo through NPC xenograft tumor growth, lung metastatic mice model and popliteal lymph node (LN) metastasis model.
    RESULTS: Overexpression of CLCA2 significantly decreased proliferation, migration and invasion of NPC cells. In contrast, knockdown of CLCA2 elicited the opposite effects. CLCA2 overexpression suppressed xenograft tumor growth and lung, popliteal lymph node (LN) metastasis in vivo. CLCA2 inhibited tumor metastasis through suppressing epithelial-Mesenchymal transition (EMT) and in-activating FAK/ERK1/2 signaling pathway in NPC cells. Immunohistochemical staining of 143 NPC samples revealed that CLCA2 expression was an independent, favorable prognostic factor for overall survival and distant metastasis-free survival of patients. In addition, inhibition of FAK and ERK1/2 reversed CLCA2 silencing-induced tumor cell migration. Furthermore, inhibitors against chloride channels suppressed NPC cellular migration which could have been enhanced by the presence of CLCA2.
    CONCLUSIONS: CLCA2 suppress NPC proliferation, migration, invasion and epithelial-mesenchymal transition through inhibiting FAK/ERK signaling.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号