Bves

BVES
  • 文章类型: Case Reports
    POPDC1也被称为BVES,是一种高度保守的跨膜蛋白,重要的横纹肌功能和稳态。POPDC1基因的致病变异与25型肢带型肌营养不良(LGMDR25)相关。在本研究中,我们对具有LGMD临床特征的单个家族进行了三全外显子组测序(WES),然后进行了Sanger测序.所有POPDC1错义变体的蛋白质建模(POPDC1Pro134Leu,POPDC1Ile193Ser,使用分子动力学(MD)模拟进行与LGMDR25相关的POPDC1Ser201Phe)。我们在POPDC1基因中鉴定了纯合错义变体(c.401C>T;p.Pro134Leu)。改变了3D结构,破坏性波动,不太紧凑,在POPDC1蛋白模型的所有三种变体中均观察到不稳定性。相比之下,POPDC1Ser201Phe蛋白动力学比其他变体更不稳定。对新发现的变体的功能研究将为该疾病的潜在机制增加关键答案。
    POPDC1 also known as BVES, is a highly conserved transmembrane protein, important for striated muscle function and homeostasis. Pathogenic variants in the POPDC1 gene are associated with limb-girdle muscular dystrophy type 25 (LGMDR25). In the present study, we performed trio-whole exome sequencing (WES) followed by Sanger sequencing on a single family having LGMD clinical features. Protein modeling of all POPDC1 missense variants (POPDC1Pro134Leu , POPDC1Ile193Ser , and POPDC1Ser201Phe ) associated with LGMDR25 were performed using Molecular Dynamics (MD) simulation. We identified a homozygous missense variant (c.401C>T; p.Pro134Leu) in the POPDC1 gene. Altered 3D structure, disruptive fluctuation, less compactness, and instability were observed in all the three variants of POPDC1 protein models. In comparison, POPDC1Ser201Phe protein dynamics were more unstable than other variants. Functional study of newly identified variant would add key answers to underlying mechanisms of the 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
    肿瘤细胞从原发灶脱离是肝细胞癌(HCC)转移的早期事件,其中细胞粘附分子起着重要作用。机械拥挤的作用越来越引起人们的关注。先前的研究发现,过度拥挤可以诱导活细胞挤压以维持上皮细胞稳态,通常,活的挤压细胞最终会通过称为“失巢”的过程死亡,提示肿瘤细胞对失巢凋亡具有抗性,可能会通过细胞挤出引发原发肿瘤的转移。我们已经证明跨膜粘附分子血管心外膜物质(BVES)抑制是HCC转移的早期事件。然而,其抑制是否与肝癌细胞挤压有关,特别是在肝癌转移中,仍然未知。本研究旨在探讨BVES在肿瘤细胞挤压在肝癌转移中的作用,以及潜在的机制。
    通过硅胶室观察到细胞挤出,培养皿倒置,和三维细胞培养模型。聚合酶链反应,西方印迹,免疫组织化学,免疫荧光,免疫共沉淀,和RhoA活性测定用于探索BVES调节细胞挤压的潜在机制。建立了原位异种移植模型,以研究BVES和细胞挤出在体内HCC转移中的作用。
    在HCC细胞和组织中观察到肿瘤细胞挤出。BVES在HCC和挤压肿瘤细胞中的表达均降低。BVES过表达导致体外和体内HCC细胞挤出减少。此外,我们的数据显示,BVES与ZO-1和GEFT共同定位,调节ZO-1的表达和定位,和GEFT分布,从而调节RhoA活性。
    本研究表明,肝癌中BVES下调增强了肿瘤细胞的挤压,从而促进HCC转移,这有助于更全面地了解肿瘤转移,并为开发新的HCC治疗策略提供了线索。视频摘要。
    Tumor cells detachment from primary lesions is an early event for hepatocellular carcinoma (HCC) metastasis, in which cell adhesion molecules play an important role. The role of mechanical crowding has attracted increasing attention. Previous studies have found that overcrowding can induce live cells extrusion to maintain epithelial cell homeostasis, and normally, live extruded cells eventually die through a process termed anoikis, suggesting the potential of tumor cells resistant to anoikis might initiate metastasis from primary tumors by cell extrusion. We have demonstrated transmembrane adhesion molecule blood vessel epicardial substance (BVES) suppression as an early event in HCC metastasis. However, whether its suppression is involved in HCC cell extrusion, especially in HCC metastasis, remains unknown. This study aims to investigate the role of BVES in tumor cells extrusion in HCC metastasis, as well as the underlying mechanisms.
    Cells extrusion was observed by silicone chamber, petri dish inversion, and three-dimensional cell culture model. Polymerase chain reaction, western blotting, immunohistochemistry, immunofluorescence, co-immunoprecipitation, and RhoA activity assays were used to explore the underlying mechanisms of cell extrusion regulated by BVES. An orthotopic xenograft model was established to investigate the effects of BVES and cell extrusion in HCC metastasis in vivo.
    Tumor cell extrusion was observed in HCC cells and tissues. BVES expression was decreased both in HCC and extruded tumor cells. BVES overexpression led to the decrease in HCC cells extrusion in vitro and in vivo. Moreover, our data showed that BVES co-localized with ZO-1 and GEFT, regulating ZO-1 expression and localization, and GEFT distribution, thus modulating RhoA activity.
    The present study revealed that BVES downregulation in HCC enhanced tumor cells extrusion, thus promoting HCC metastasis, which contributed to a more comprehensive understanding of tumor metastasis, and provided clues for developing novel HCC therapy strategies. Video abstract.
    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
    目的:血管平滑肌细胞(VSMC)可塑性与血管病变的病理过程密切相关。血管心外膜物质(Bves)已成为心内血管发生和器官稳态的重要调节剂。然而,Bves在VSMC可塑性和新内膜病变发展中的参与和作用尚不清楚。
    方法:我们使用移植物动脉硬化和体外PDGF处理的VSMC的体内大鼠模型,并通过转录组学分析和文献检索鉴定了新的VSMC收缩表型相关基因Bves。使用体外敲低和过表达方法来研究VSMC表型可塑性的潜在机制。在体内,在大鼠主动脉移植物中产生VSMC特异性Bves过表达,以评估Bves在新内膜病变发展中的生理功能。
    结果:这里,我们发现Bves的表达在体内主动脉移植物和体外PDGF处理的VSMC中负向调节。Bves的基因敲除显著抑制,而Bves过表达显着促进,VSMC收缩表型。此外,RNA测序揭示了VSMC中Bves与双特异性蛋白磷酸酶1(Dusp1)表达之间的正相关关系。我们发现Bves敲低抑制了Dusp1的表达,但增强p38MAPK和ERK1/2激活,导致VSMC收缩表型的丧失。在体内,对大鼠移植模型的分析证实,主动脉同种异体移植物中VSMC特异性Bves和Dusp1的过表达显着减轻了新内膜病变的形成。
    结论:Bves通过Dusp1依赖性p38MAPK和ERK1/2信号维持VSMC收缩表型,并防止新内膜形成,强调Bves在预防移植血管病变中的重要作用。
    OBJECTIVE: Vascular smooth muscle cell (VSMC) plasticity is tightly associated with the pathological process of vasculopathy. Blood vessel epicardial substance (Bves) has emerged as an important regulator of intracardiac vasculogenesis and organ homeostasis. However, the involvement and role of Bves in VSMC plasticity and neointimal lesion development remain unclear.
    METHODS: We used an in vivo rat model of graft arteriosclerosis and in vitro PDGF-treated VSMCs and identified the novel VSMC contractile phenotype-related gene Bves using a transcriptomic analysis and literature search. In vitro knockdown and overexpression approaches were used to investigate the mechanisms underlying VSMC phenotypic plasticity. In vivo, VSMC-specific Bves overexpression in rat aortic grafts was generated to assess the physiological function of Bves in neointimal lesion development.
    RESULTS: Here, we found that Bves expression was negatively regulated in aortic allografts in vivo and PDGF-treated VSMCs in vitro. The genetic knockdown of Bves dramatically inhibited, whereas Bves overexpression markedly promoted, the VSMC contractile phenotype. Furthermore, RNA sequencing unraveled a positive correlation between Bves and dual-specificity protein phosphatase 1 (Dusp1) expression in VSMCs. We found that Bves knockdown restrained Dusp1 expression, but enhanced p38MAPK and ERK1/2 activation, resulting in the loss of the VSMC contractile phenotype. In vivo, an analysis of a rat graft model confirmed that VSMC-specific Bves and Dusp1 overexpression in aortic allografts significantly attenuated neointimal lesion formation.
    CONCLUSIONS: Bves maintains the VSMC contractile phenotype through Dusp1-dependent p38MAPK and ERK1/2 signaling, and protects against neointimal formation, underscoring the important role of Bves in preventing transplant vasculopathy.
    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: Case Reports
    由于BVES的功能丧失突变,R25型肢体肌营养不良(LGMDR25)是一种罕见的遗传性疾病,以进行性近端下肢无力和房室传导阻滞为特征。在这里,我们报道了一个患有LGMDR25的年轻中国男子,他表现出下肢不对称无力,肌痛,劳累时心悸和呼吸困难。肌肉成像显示股二头肌长头脂肪浸润,内收肌magnus,腓肠肌和比目鱼肌,以及半腱肌和股四头肌的肌水肿,保留股直肌。心电图仅显示轻度窦性心动过速,但肺功能检查提示明显的呼吸肌无力。我们的报告扩展了表型谱,并指出了监测LGMDR25患者呼吸功能的重要性。
    Limb girdle muscular dystrophy type R25 (LGMDR25) is a rare genetic disorder due to loss-of-function mutations in BVES, characterized by progressive proximal lower limb weakness and atrioventricular block. Here we report a young Chinese man with LGMDR25 who presented with asymmetrical lower limb weakness, myalgia, palpitations and dyspnea on exertion. Muscle imaging demonstrated fatty infiltration of the long head of biceps femoris, adductor magnus, gastrocnemius and soleus, and myoedema of semitendinosus and quadriceps, sparing rectus femoris. ECG showed only mild sinus tachycardia but pulmonary function test suggested prominent respiratory muscle weakness. Our report expands the phenotypical spectrum and indicates the importance of monitoring respiratory function in LGMDR25 patients.
    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:Anoctamin5(ANO5)是属于TMEM16/Anoctamin家族的膜蛋白,其缺乏导致肢带肌营养不良R12(LGMDR12)的发展。然而,关于ANO5的相互作用组及其细胞功能知之甚少。
    结果:在这项研究中,我们利用近端标记方法来鉴定稳定表达用BioID2标记的ANO5的C2C12成肌细胞中ANO5的相互作用蛋白。质谱从ANO5-BioID2样品中鉴定出41种独特的蛋白质,包括BVES和POPDC3,但不是来自与ANO6或MG53融合的BioID2。通过免疫共沉淀(Co-IP)进一步证实了ANO5和BVES之间的相互作用,ANO5的N端介导与BVES的C端相互作用。ANO5和BVES共定位在肌细胞中并富集在内质网(ER)膜。基因组编辑介导的ANO5或BVES破坏显着抑制C2C12成肌细胞分化,对增殖影响很小。
    结论:综合来看,这些数据表明,BVES是ANO5的一种新型相互作用蛋白,参与肌肉分化的调节。
    BACKGROUND: Anoctamin 5 (ANO5) is a membrane protein belonging to the TMEM16/Anoctamin family and its deficiency leads to the development of limb girdle muscular dystrophy R12 (LGMDR12). However, little has been known about the interactome of ANO5 and its cellular functions.
    RESULTS: In this study, we exploited a proximal labeling approach to identify the interacting proteins of ANO5 in C2C12 myoblasts stably expressing ANO5 tagged with BioID2. Mass spectrometry identified 41 unique proteins including BVES and POPDC3 specifically from ANO5-BioID2 samples, but not from BioID2 fused with ANO6 or MG53. The interaction between ANO5 and BVES was further confirmed by co-immunoprecipitation (Co-IP), and the N-terminus of ANO5 mediated the interaction with the C-terminus of BVES. ANO5 and BVES were co-localized in muscle cells and enriched at the endoplasmic reticulum (ER) membrane. Genome editing-mediated ANO5 or BVES disruption significantly suppressed C2C12 myoblast differentiation with little impact on proliferation.
    CONCLUSIONS: Taken together, these data suggest that BVES is a novel interacting protein of ANO5, involved in regulation of muscle differentiation.
    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
    在结肠腺癌(COAD)患者中已显示长链非编码RNA血管心外膜物质反义RNA1(BVES-AS1)的表达不足。然而,它在COAD中的作用还有待探索。本研究旨在探讨BVES-AS1在COAD中的作用及可能的作用机制。殖民地的形成,细胞计数试剂盒-8,JC-1线粒体膜电位测定,伤口愈合,transwell,和蛋白质印迹分析用于测量细胞增殖,凋亡,迁移,入侵,和COAD细胞中的上皮-间质转化(EMT)。RNA下拉,荧光素酶报告基因,和RNA结合蛋白免疫沉淀法检测BVES-AS1与下游基因的相互作用。BVES-AS1在COAD细胞中以低水平表达。过表达BVES-AS1抑制COAD细胞增殖,迁移,入侵,和EMT同时升高细胞凋亡。机械上,BVES-AS1作为形成miR-522-3p的竞争性内源性RNA起作用,以调节附近基因血管心外膜物质(BVES)的表达。除此之外,BVES-AS1招募TATA-box结合蛋白相关因子15(TAF15)以促进BVES信使RNA的稳定性。一起来看,我们的研究证实,BVES-AS1通过与miR-522-3p和TAF15相互作用以调节BVES表达来抑制COAD进展,这可能为COAD治疗提供一个视角。
    The underexpression of the long noncoding RNA blood vessel epicardial substance antisense RNA 1 (BVES-AS1) has been shown in colon adenocarcinoma (COAD) patients. However, its role in COAD remains to be explored. This study aimed to investigate the function and potential mechanism of BVES-AS1 in COAD. Colony formation, Cell Counting Kit-8, JC-1 mitochondrial membrane potential assay, wound healing, transwell, and western blot analyses were used to measure cell proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT) in COAD cells. RNA pull-down, luciferase reporter, and RNA binding protein immunoprecipitation assays were used to detect the interaction of BVES-AS1 and downstream genes. BVES-AS1 was expressed at low levels in COAD cells. Overexpressed BVES-AS1 inhibited COAD cell proliferation, migration, invasion, and EMT while elevating cell apoptosis. Mechanistically, BVES-AS1 functioned as a competing endogenous RNA sponging miR-522-3p to regulate the expression of nearby gene blood vessel epicardial substance (BVES). Besides this, BVES-AS1 recruited TATA-box binding protein associated factor 15 (TAF15) to promote BVES messenger RNA stability. Taken together, our study confirmed that BVES-AS1 inhibited COAD progression via interacting with miR-522-3p and TAF15 to regulate BVES expression, which might offer a perspective for COAD treatment.
    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
    Background: Tetralogy of Fallot (TOF) accounts for ∼10% of congenital heart disease cases. The blood vessel epicardial substance (BVES) gene has been reported to play a role in the function of adult hearts. However, whether allelic variants in BVES contribute to the risk of TOF and its possible mechanism remains unknown. Methods: The open reading frame of the BVES gene was sequenced using samples from 146 TOF patients and 100 unrelated healthy controls. qRT-PCR and western blot assays were used to confirm the expression of mutated BVES variants in the TOF samples. The online software Polyphen2 and SIFT were used to predict the deleterious effects of the observed allelic variants. The effects of these allelic variants on the transcriptional activities of genes were examined using dual-fluorescence reporter assays. Results: We genotyped four single nucleotide polymorphisms (SNPs) in the BVES gene from each of the 146 TOF patients. Among them, the minor allelic frequencies of c.385C>T (p.R129W) were 0.035% in TOF, but ∼0.025% in 100 controls and the Chinese Millionome Database. This allelic variant was predicted to be a potentially harmful alteration by the Polyphen2 and SIFT softwares. qRT-PCR and western blot analyses indicated that the expression of BVES in the six right ventricular outflow tract samples with the c.385C>T allelic variant was significantly downregulated. A dual-fluorescence reporter system showed that the c.385C>T allelic variant significantly decreased the transcriptional activity of the BVES gene and also decreased transcription from the GATA4 and NKX2.5 promoters. Conclusions: c.385C>T (p.R129W) is a functional SNP of the BVES gene that reduces the transcriptional activity of BVES in vitro and in vivo in TOF tissues. This subsequently affects the transcriptional activities of GATA4 and NKX2.5 related to TOF. These findings suggest that c.385C>T may be associated with the risk of TOF in the Han Chinese population.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    The axon guidance cues netrin-1 has been reported to be associated with cancer progression in various types of human cancers. However, the underlying molecular mechanism of netrin-1-mediated metastasis remains obscure. In this study, we found that overexpression of netrin-1 promoted HCC cell migration and invasion as determined by transwell assay and 3D cell culture assay. However, netrin-1 knockdown inhibited these processes. Further investigation indicated that netrin-1 decreased the expression of Blood Vessel Epicardial Substance (BVES), which was down-regulated in HCC. Interestingly, LY294002, a special inhibitor to PI3K/AKT signaling which was determined as a downstream pathway of netrin-1, restored the reduction in BVES caused by netrin-1. In addition, BVES exhibited an opposite effect on HCC cell metastasis to that of netrin-1. Importantly, up-regulating BVES expression significantly attenuated netrin-1-enhanced migration and invasion, whereas silencing BVES expression rescued the metastatic phenotype in netrin-1 knockdown cells. Moreover, netrin-1 expression was negatively correlated with BVES in HCC tissues and cell lines with different metastatic potential. Taken together, these results reveal that netrin-1 promotes HCC cell metastasis by regulating BVES expression via AKT activation.
    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