BVES

BVES
  • 文章类型: Journal Article
    肢体带型肌营养不良R25(LGMDR25)是由编码cAMP结合蛋白的BVES中的隐性突变引起的,以进行性肌营养不良为特征,患者的肌肉功能恶化和心脏传导受损。目前还没有针对LGMDR25患者的治疗方法。在这里,我们报告了由肌肉特异性启动子MHCK7(AAV9)驱动的重组腺相关病毒9(AAV9)介导的人BVES全身递送的有效性和安全性。BVES)在BVES敲除(BVES-KO)小鼠中。AAV9。当腹膜内注射到新生BVES-KO小鼠中时,BVES有效地转导心脏和骨骼肌组织。AAV9。BVES显著改善了体重增加,肌肉质量,肌肉力量,BVES-KO小鼠的运动表现不分性别。AAV9。BVES还显着改善了肌营养不良的组织病理学特征。在全身性AAV9之后,在运动诱导的应激下,BVES-KO小鼠的心率降低也被正常化。BVES交付。此外,静脉注射AAV9.在疾病发作后对成年BVES-KO小鼠施用BVES也导致体重的显着改善,肌肉质量,肌肉收缩性,和压力引起的心律异常。未检测到明显的毒性。一起来看,这些结果为支持AAV9提供了概念验证证据.LGMDR25的BVES基因治疗。
    Limb-girdle muscular dystrophy type R25 (LGMDR25) is caused by recessive mutations in BVES encoding a cAMP-binding protein, characterized by progressive muscular dystrophy with deteriorating muscle function and impaired cardiac conduction in patients. There is currently no therapeutic treatment for LGMDR25 patients. Here we report the efficacy and safety of recombinant adeno-associated virus 9 (AAV9)-mediated systemic delivery of human BVES driven by a muscle-specific promoter MHCK7 (AAV9.BVES) in BVES-knockout (BVES-KO) mice. AAV9.BVES efficiently transduced the cardiac and skeletal muscle tissues when intraperitoneally injected into neonatal BVES-KO mice. AAV9.BVES dramatically improved body weight gain, muscle mass, muscle strength, and exercise performance in BVES-KO mice regardless of sex. AAV9.BVES also significantly ameliorated the histopathological features of muscular dystrophy. The heart rate reduction was also normalized in BVES-KO mice under exercise-induced stress following systemic AAV9.BVES delivery. Moreover, intravenous AAV9.BVES administration into adult BVES-KO mice after the disease onset also resulted in substantial improvement in body weight, muscle mass, muscle contractility, and stress-induced heart rhythm abnormality. No obvious toxicity was detected. Taken together, these results provide the proof-of-concept evidence to support the AAV9.BVES gene therapy for LGMDR25.
    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
    通过支架蛋白建立大分子复合物是通过锚定的腺苷酸环化酶(AC)局部产生cAMP以及随后的心脏功能所必需的cAMP信号传导的关键。我们确定了一种新型AC支架,含有大力水手结构域(POPDC)的蛋白质。POPDC蛋白家族对心脏起搏和传导非常重要,部分归因于它们的cAMP依赖性结合和TREK-1钾通道的调节。我们显示TREK-1结合AC9:POPDC1复合物,并以POPDC1依赖性方式与心脏中的AC9活性共同纯化。虽然AC9:POPDC1的相互作用与cAMP无关,TREK-1与AC9和POPDC1的结合在β-肾上腺素能受体(βAR)的刺激下减少。AC9活性对于用AC9:POPDC1形成TREK-1复合物的βAR减少和逆转TREK-1电流的POPDC1增强是必需的。最后,编码AC9(Adcy9)的基因缺失会在休息时引起心动过缓和压力引起的心率变异性,表型比Popdc1的丢失温和,但与Kcnk2(TREK-1)的丢失相似。因此,POPDC1代表了AC9与TREK-1相互作用以调节心率控制的新型适配器。
    The establishment of macromolecular complexes by scaffolding proteins is key to the local production of cAMP by anchored adenylyl cyclase (AC) and the subsequent cAMP signaling necessary for cardiac functions. We identify a novel AC scaffold, the Popeye domain-containing (POPDC) protein. The POPDC family of proteins is important for cardiac pacemaking and conduction, due in part to their cAMP-dependent binding and regulation of TREK-1 potassium channels. We show that TREK-1 binds the AC9:POPDC1 complex and copurifies in a POPDC1-dependent manner with AC9 activity in heart. Although the AC9:POPDC1 interaction is cAMP-independent, TREK-1 association with AC9 and POPDC1 is reduced upon stimulation of the β-adrenergic receptor (βAR). AC9 activity is required for βAR reduction of TREK-1 complex formation with AC9:POPDC1 and in reversing POPDC1 enhancement of TREK-1 currents. Finally, deletion of the gene-encoding AC9 (Adcy9) gives rise to bradycardia at rest and stress-induced heart rate variability, a milder phenotype than the loss of Popdc1 but similar to the loss of Kcnk2 (TREK-1). Thus, POPDC1 represents a novel adaptor for AC9 interactions with TREK-1 to regulate heart rate control.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 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
    广泛的研究已经发现了不同形式的突触可塑性和一系列作为正或负调节因子的分子信号机制。具体来说,循环3\',5'-环磷酸腺苷(cAMP)依赖性信号通路与持久的突触可塑性密切相关。在这项研究中,我们检查了含Popeye结构域的蛋白1(POPDC1)(或血管心外膜物质(BVES))的作用,cAMP效应蛋白,调节海马突触可塑性。与其他cAMP效应器不同,如蛋白激酶A(PKA)和cAMP直接激活的交换因子,POPDC1是膜结合的,cAMP结合盒的序列不同于典型的cAMP结合域,这表明POPDC1可能在cAMP介导的信号传导中具有独特的作用。我们的结果表明,Popdc1在包括海马在内的各个大脑区域广泛表达。Popdc1敲除(KO)小鼠的急性海马切片在CA1长时程增强(LTP)中表现出PKA依赖性增强,以响应较弱的刺激范例,其在来自野生型小鼠的切片中仅诱导瞬时LTP。POPDC1的丢失,同时不影响LTP的基础传输或输入特异性,导致高频刺激期间反应改变。Popdc1KO小鼠也显示出毛喉素诱导的增强作用。总的来说,这些发现揭示了POPDC1作为海马突触可塑性的新型负调节因子,连同其与磷酸二酯酶(PDEs)相互作用的最新证据,提示POPDC1参与调节活性依赖性局部cAMP-PKA-PDE信号传导。
    Extensive research has uncovered diverse forms of synaptic plasticity and an array of molecular signaling mechanisms that act as positive or negative regulators. Specifically, cyclic 3\',5\'-cyclic adenosine monophosphate (cAMP)-dependent signaling pathways are crucially implicated in long-lasting synaptic plasticity. In this study, we examine the role of Popeye domain-containing protein 1 (POPDC1) (or blood vessel epicardial substance (BVES)), a cAMP effector protein, in modulating hippocampal synaptic plasticity. Unlike other cAMP effectors, such as protein kinase A (PKA) and exchange factor directly activated by cAMP, POPDC1 is membrane-bound and the sequence of the cAMP-binding cassette differs from canonical cAMP-binding domains, suggesting that POPDC1 may have an unique role in cAMP-mediated signaling. Our results show that Popdc1 is widely expressed in various brain regions including the hippocampus. Acute hippocampal slices from Popdc1 knockout (KO) mice exhibit PKA-dependent enhancement in CA1 long-term potentiation (LTP) in response to weaker stimulation paradigms, which in slices from wild-type mice induce only transient LTP. Loss of POPDC1, while not affecting basal transmission or input-specificity of LTP, results in altered response during high-frequency stimulation. Popdc1 KO mice also show enhanced forskolin-induced potentiation. Overall, these findings reveal POPDC1 as a novel negative regulator of hippocampal synaptic plasticity and, together with recent evidence for its interaction with phosphodiesterases (PDEs), suggest that POPDC1 is involved in modulating activity-dependent local cAMP-PKA-PDE signaling.
    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
    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: Case Reports
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号