CLCA2

CLCA2
  • 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
    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
    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