cdc42 GTP-Binding Protein

  • 文章类型: Journal Article
    缺氧缺血性脑损伤提出了重大的神经系统挑战,通常表现在围产期。具体来说,脑室周围白质软化(PVL)正在成为脑瘫和智力障碍的重要原因。它损害了大脑微循环,导致大脑脑室周围的氧气或血流不足。正如广泛记录的那样,这些病理状况可能是由包括早产在内的多种因素引起的(占总病例的4-5%),以及单胎流产和遗传变异,例如与GTP酶途径相关的变异。全外显子组测序(WES)分析在患有PVL的患者中鉴定出含有pleckstrin同源结构域的家族G成员1(PLEKHG1)基因内的从头致病变异。PLEKHG1基因普遍表达,在脑组织中显示高表达模式。PLEKHG1是Rho鸟嘌呤核苷酸交换因子家族的一部分,并且该蛋白对于GTP酶途径中的细胞分裂控制蛋白42(CDC42)活化是必需的。CDC42是Rho亚家族的关键小GTP酶,调节各种细胞功能,如细胞形态,迁移,内吞作用,和细胞周期进程。涉及PLEKHG1和CDC42的分子机制在血管内皮细胞的重新定向中具有有趣的作用,因此提示内皮细胞对机械应力的破坏反应可能与白质病变的形成有关。重要的是,CDC42与白质异常的关联由其MIM表型编号强调。相比之下,尽管PLEKHG1最近与显示白质高信号的患者有关,它目前缺乏MIM表型数。此外,在计算机分析分类鉴定的变异为致病性。尽管患者早产并随后进行了二胎妊娠,在此期间,它的库温去世了,我们建议所描述的变体可以强烈促进PVL。本研究的目的是建立PLEKHG1基因与PVL之间的合理关联。
    Hypoxic-ischemic brain damage presents a significant neurological challenge, often manifesting during the perinatal period. Specifically, periventricular leukomalacia (PVL) is emerging as a notable contributor to cerebral palsy and intellectual disabilities. It compromises cerebral microcirculation, resulting in insufficient oxygen or blood flow to the periventricular region of the brain. As widely documented, these pathological conditions can be caused by several factors encompassing preterm birth (4-5% of the total cases), as well single cotwin abortion and genetic variants such as those associated with GTPase pathways. Whole exome sequencing (WES) analysis identified a de novo causative variant within the pleckstrin homology domain-containing family G member 1 (PLEKHG1) gene in a patient presenting with PVL. The PLEKHG1 gene is ubiquitously expressed, showing high expression patterns in brain tissues. PLEKHG1 is part of a family of Rho guanine nucleotide exchange factors, and the protein is essential for cell division control protein 42 (CDC42) activation in the GTPase pathway. CDC42 is a key small GTPase of the Rho-subfamily, regulating various cellular functions such as cell morphology, migration, endocytosis, and cell cycle progression. The molecular mechanism involving PLEKHG1 and CDC42 has an intriguing role in the reorientation of cells in the vascular endothelium, thus suggesting that disruption responses to mechanical stress in endothelial cells may be involved in the formation of white matter lesions. Significantly, CDC42 association with white matter abnormalities is underscored by its MIM phenotype number. In contrast, although PLEKHG1 has been recently associated with patients showing white matter hyperintensities, it currently lacks a MIM phenotype number. Additionally, in silico analyses classified the identified variant as pathogenic. Although the patient was born prematurely and subsequently to dichorionic gestation, during which its cotwin died, we suggest that the variant described can strongly contribute to PVL. The aim of the current study is to establish a plausible association between the PLEKHG1 gene and PVL.
    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
    为了进入上皮细胞,专性细胞内病原体肺炎衣原体分泌早期效应蛋白,它们结合并调节宿主细胞的质膜,并募集几种关键的内吞宿主蛋白。这里,我们提出了进入相关的衣原体效应蛋白的高分辨率结构,SemD.SemD与其宿主结合伴侣的共结晶表明SemD共选择Cdc42结合位点以激活肌动蛋白细胞骨架调节剂N-WASP,使活跃,GTP结合的Cdc42是多余的。虽然SemD与N-WASP的结合比Cdc42强得多,它不结合Cdc42效应蛋白FMNL2,表明效应蛋白特异性。此外,通过识别灵活和结构化的领域,我们表明SemD可以同时与膜相互作用,内吞蛋白SNX9和N-WASP。这里,我们在结构水平上展示了单个效应蛋白如何劫持宿主内吞系统的中心成分以实现有效的内化。
    To enter epithelial cells, the obligate intracellular pathogen Chlamydia pneumoniae secretes early effector proteins, which bind to and modulate the host-cell\'s plasma membrane and recruit several pivotal endocytic host proteins. Here, we present the high-resolution structure of an entry-related chlamydial effector protein, SemD. Co-crystallisation of SemD with its host binding partners demonstrates that SemD co-opts the Cdc42 binding site to activate the actin cytoskeleton regulator N-WASP, making active, GTP-bound Cdc42 superfluous. While SemD binds N-WASP much more strongly than Cdc42 does, it does not bind the Cdc42 effector protein FMNL2, indicating effector protein specificity. Furthermore, by identifying flexible and structured domains, we show that SemD can simultaneously interact with the membrane, the endocytic protein SNX9, and N-WASP. Here, we show at the structural level how a single effector protein can hijack central components of the host\'s endocytic system for efficient internalization.
    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
    长期突触可塑性通常与突触连接的形态变化有关。然而,突触可塑性的功能和结构方面的分子机制仍然不明确。特定形式的突触可塑性需要I型磷酸肌醇-3-激酶(PI3K)的催化活性,如NMDA受体依赖性长期增强(LTP)和mGluR依赖性长期抑郁(LTD)。另一方面,PI3K信号与神经元生长和突触形成有关。因此,PI3K是协调突触强度变化与突触结构重塑的有希望的候选者。为了调查这个问题,我们针对海马神经元中I型PI3Ks的单个调节亚基,并采用电生理,生化和成像技术,以评估其在突触可塑性中的作用。我们发现一种特殊的调节同工型,p85α,对于LTP是选择性需要的。这种特异性基于其BH结构域,它与小的GTP酶Rac1和Cdc42结合,它们是肌动蛋白细胞骨架的关键调节因子。此外,cofilin,LTP诱导后在树突棘中积累的肌动蛋白动力学的关键调节剂,在不存在p85α或其BH结构域作为显性阴性构建物过表达时未能这样做。最后,与肌动蛋白调节机制的这种趋同一致,PI3K复合物中p85α的存在决定了LTP过程中树突棘中肌动蛋白聚合的程度。因此,这项研究揭示了通过PI3K催化活性和调节亚基的特定同工型的协同作用将结构和功能突触可塑性联系起来的分子机制。
    Long-term synaptic plasticity is typically associated with morphological changes in synaptic connections. However, the molecular mechanisms coupling functional and structural aspects of synaptic plasticity are still poorly defined. The catalytic activity of type I phosphoinositide-3-kinase (PI3K) is required for specific forms of synaptic plasticity, such as NMDA receptor-dependent long-term potentiation (LTP) and mGluR-dependent long-term depression (LTD). On the other hand, PI3K signaling has been linked to neuronal growth and synapse formation. Consequently, PI3Ks are promising candidates to coordinate changes in synaptic strength with structural remodeling of synapses. To investigate this issue, we targeted individual regulatory subunits of type I PI3Ks in hippocampal neurons and employed a combination of electrophysiological, biochemical and imaging techniques to assess their role in synaptic plasticity. We found that a particular regulatory isoform, p85α, is selectively required for LTP. This specificity is based on its BH domain, which engages the small GTPases Rac1 and Cdc42, critical regulators of the actin cytoskeleton. Moreover, cofilin, a key regulator of actin dynamics that accumulates in dendritic spines after LTP induction, failed to do so in the absence of p85α or when its BH domain was overexpressed as a dominant negative construct. Finally, in agreement with this convergence on actin regulatory mechanisms, the presence of p85α in the PI3K complex determined the extent of actin polymerization in dendritic spines during LTP. Therefore, this study reveals a molecular mechanism linking structural and functional synaptic plasticity through the coordinate action of PI3K catalytic activity and a specific isoform of the regulatory subunits.
    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
    GTP酶Cdc42调节大多数真核生物的极化生长。在双极酵母裂殖酵母中,Cdc42活化在极化生长位点周期性循环。这些周期性循环是由交替的正反馈和时间延迟的负反馈回路引起的。在每个极化端,当活性Cdc42募集Pak1激酶以防止Cdc42进一步活化时,建立负反馈。目前尚不清楚Cdc42激活如何在依赖Pak1的负反馈后返回到每个末端。我们发现,破坏分支肌动蛋白介导的内吞作用会使Cdc42在细胞末端的再激活失效。使用实验和数学方法,我们表明,从细胞末端去除胞吞依赖性Pak1可以使Cdc42激活剂Scd1返回到该末端,从而使Cdc42重新激活。此外,我们表明Pak1通过激活胞吞作用引起其自身的去除。这些发现为Cdc42调节的自组织提供了更深入的见解,并揭示了细胞极性建立中先前未知的内吞反馈。
    The GTPase Cdc42 regulates polarized growth in most eukaryotes. In the bipolar yeast Schizosaccharomyces pombe, Cdc42 activation cycles periodically at sites of polarized growth. These periodic cycles are caused by alternating positive feedback and time-delayed negative feedback loops. At each polarized end, negative feedback is established when active Cdc42 recruits the Pak1 kinase to prevent further Cdc42 activation. It is unclear how Cdc42 activation returns to each end after Pak1-dependent negative feedback. We find that disrupting branched actin-mediated endocytosis disables Cdc42 reactivation at the cell ends. Using experimental and mathematical approaches, we show that endocytosis-dependent Pak1 removal from the cell ends allows the Cdc42 activator Scd1 to return to that end to enable reactivation of Cdc42. Moreover, we show that Pak1 elicits its own removal via activation of endocytosis. These findings provide a deeper insight into the self-organization of Cdc42 regulation and reveal previously unknown feedback with endocytosis in the establishment of cell polarity.
    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
    环状RNA(circularRNA,circRNA)家族是一组内源性非编码RNA(ncRNAs),在多种生理和病理过程中具有关键功能。包括炎症,癌症,和心血管疾病。然而,它们在调节先天免疫反应中的作用尚不清楚.这里,我们定义了细胞分裂周期42(CDC42)-165aa,一种由circRNAcircCDC42编码的蛋白质,在肺炎克雷伯菌(KP)感染的肺泡巨噬细胞中过表达。高水平的CDC42-165aa诱导了Pyrin炎性体的过度激活,并加重了肺泡巨噬细胞的焦亡,而抑制CDC42-165aa通过抑制Pyrin炎性体介导的细胞凋亡来减轻KP感染后小鼠的肺损伤。总的来说,这些结果表明,CDC42-165aa通过抑制CDC42GTP酶的激活而刺激Pyrin炎性体,为临床上的病原菌感染提供了一个潜在的临床靶点.
    The circular RNA (circRNA) family is a group of endogenous non-coding RNAs (ncRNAs) that have critical functions in multiple physiological and pathological processes, including inflammation, cancer, and cardiovascular diseases. However, their roles in regulating innate immune responses remain unclear. Here, we define Cell division cycle 42 (CDC42)-165aa, a protein encoded by circRNA circCDC42, which is overexpressed in Klebsiella pneumoniae (KP)-infected alveolar macrophages. High levels of CDC42-165aa induces the hyperactivation of Pyrin inflammasomes and aggravates alveolar macrophage pyroptosis, while the inhibition of CDC42-165aa reduces lung injury in mice after KP infection by inhibiting Pyrin inflammasome-mediated pyroptosis. Overall, these results demonstrate that CDC42-165aa stimulates Pyrin inflammasome by inhibiting CDC42 GTPase activation and provides a potential clinical target for pathogenic bacterial infection in clinical practice.
    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
    肌动蛋白组装和动力学对于维持细胞结构和改变生理状态至关重要。肌动蛋白对各种细胞过程的广泛影响使得解剖肌动蛋白调节蛋白的特定作用具有挑战性。使用在肥大细胞皮质上传播的肌动蛋白波作为模型,我们发现,在肌动蛋白波中,在Arp2/3复合物之前招募了福尔马林(FMNL1和mDia3)。GTP酶Cdc42相互作用驱动FMNL1振荡,具有活性Cdc42和FMNL1的组成型活性突变体,能够独立于肌动蛋白波在质膜上形成波。此外,Arp2/3的延迟募集拮抗FMNL1和活性Cdc42。这种拮抗作用不是由于单体肌动蛋白的竞争,而是由于它们共同的上游调节剂,活性Cdc42,其水平通过SHIP1募集受Arp2/3负调控。总的来说,我们的研究强调了肌动蛋白细胞骨架网络动态控制中复杂的反馈回路.
    Actin assembly and dynamics are crucial for maintaining cell structure and changing physiological states. The broad impact of actin on various cellular processes makes it challenging to dissect the specific role of actin regulatory proteins. Using actin waves that propagate on the cortex of mast cells as a model, we discovered that formins (FMNL1 and mDia3) are recruited before the Arp2/3 complex in actin waves. GTPase Cdc42 interactions drive FMNL1 oscillations, with active Cdc42 and the constitutively active mutant of FMNL1 capable of forming waves on the plasma membrane independently of actin waves. Additionally, the delayed recruitment of Arp2/3 antagonizes FMNL1 and active Cdc42. This antagonism is not due to competition for monomeric actin but rather for their common upstream regulator, active Cdc42, whose levels are negatively regulated by Arp2/3 via SHIP1 recruitment. Collectively, our study highlights the complex feedback loops in the dynamic control of the actin cytoskeletal network.
    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:勃起功能障碍(ED)是一种常见的男性性功能障碍,随着发病率的增加,目前的治疗往往是无效的。
    方法:用血管内皮生长因子(VEGFA)治疗骨髓间充质干细胞(BM-MSCs),并通过Transwell测定法测定它们的细胞迁移速率。vonWillebrand因子(vWF)VE-cadherin的表达,通过qRT-PCR和Westernblot分析确定内皮型一氧化氮合酶(eNOS)内皮标志物。通过用si-MALAT1转染VEGFA诱导的BM-MSC并过表达CDC42和PAK1,探索了MALAT1诱导的BM-MC通过CDC42/PAK1/桩蛋白途径向EC分化。通过蛋白质免疫沉淀检查了VEGFA处理和非VEGFA处理的BM-MSC中CDC42,PAK1和桩蛋白之间的结合能力。MiR-206在VEGFA诱导的BM-MSC中过表达,MALAT1、miR-206和CDC42的结合位点使用荧光素酶测定进行鉴定。将60只雄性SD大鼠分为6组(n=10/组)。通过APO实验证明了DMED建模,并通过测量血糖水平进行了评估。通过测量海绵体内压(ICP)和平均动脉压(MAP)评估勃起功能。通过qRT-PCR分析阴茎勃起组织,蛋白质印迹分析,和免疫组织化学染色。
    结果:VEGFA处理条件下的MALAT1通过调节CDC42/PAK1/桩蛋白轴来调节BM-MSCs向ECs的分化。体外实验表明,干扰CDC42和MALAT1的表达抑制了BM-MSCs向EC的分化。CDC42与PAK1结合,PAK1与桩蛋白结合。此外,VEGFA组中的CDC42具有更大的与PAK1结合的能力,而VEGFA组中的PAK1具有更大的与桩蛋白结合的能力。miR-206在VEGFA诱导的BM-MSC中的过表达表明MALAT1与CDC423'-UTR竞争结合miR-206,进而参与BM-MSC向EC的分化。与DMED模型组相比,3个BM-MSCs治疗组的ICP/MAP比值显著增高.
    结论:MALAT1通过调节miR-206/CDC42/PAK1/桩蛋白轴来促进BM-MSC分化为ECs,从而改善ED。本发现揭示了MALAT1在修复BM-MSCs勃起功能中的重要作用,并为BM-MSC介导的DMED修复提供了新的机制见解。
    BACKGROUND: Erectile dysfunction (ED) is a common male sexual dysfunction, with an increasing incidence, and the current treatment is often ineffective.
    METHODS: Vascular endothelial growth factor (VEGFA) was used to treat bone marrow-derived mesenchymal stem cells (BM-MSCs), and their cell migration rates were determined by Transwell assays. The expression of the von Willebrand Factor (vWF)VE-cadherin, and endothelial nitric oxide synthase(eNOS) endothelial markers was determined by qRT‒PCR and Western blot analyses. The MALAT1-induced differentiation of BM-MCs to ECs via the CDC42/PAK1/paxillin pathway was explored by transfecting VEGFA-induced BM-MSC with si-MALAT1 and overexpressing CDC42 and PAK1. The binding capacity between CDC42, PAK1, and paxillin in VEGFA-treated and non-VEGFA-treated BM-MSCs was examined by protein immunoprecipitation. MiR-206 was overexpressed in VEGFA-induced BM-MSC, and the binding sites of MALAT1, miR-206, and CDC42 were identified using a luciferase assay. Sixty male Sprague‒Dawley rats were divided into six groups (n = 10/group). DMED modelling was demonstrated by APO experiments and was assessed by measuring blood glucose levels. Erectile function was assessed by measuring the intracavernosa pressure (ICP) and mean arterial pressure (MAP). Penile erectile tissue was analysed by qRT‒PCR, Western blot analysis, and immunohistochemical staining.
    RESULTS: MALAT1 under VEGFA treatment conditions regulates the differentiation of BM-MSCs into ECs by modulating the CDC42/PAK1/paxillin axis. In vitro experiments demonstrated that interference with CDC42 and MALAT1 expression inhibited the differentiation of BM-MSCs to ECs. CDC42 binds to PAK1, and PAK1 binds to paxillin. In addition, CDC42 in the VEGFA group had a greater ability to bind to PAK1, whereas PAK1 in the VEGFA group had a greater ability to bind to paxillin. Overexpression of miR-206 in VEGFA-induced BM-MSCs demonstrated that MALAT1 competes with the CDC42 3\'-UTR for binding to miR-206, which in turn is involved in the differentiation of BM-MSCs to ECs. Compared to the DMED model group, the ICP/MAP ratio was significantly greater in the three BM-MSCs treatment groups.
    CONCLUSIONS: MALAT1 facilitates BM-MSC differentiation into ECs by regulating the miR-206/CDC42/PAK1/paxillin axis to improve ED. The present findings revealed the vital role of MALAT1 in the repair of BM-MSCs for erectile function and provided new mechanistic insights into the BM-MSC-mediated repair of DMED.
    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
    我们以前报道了三取代嘧啶先导化合物,即,ARN22089和ARN25062阻断CDC42与其特定下游效应物之间的相互作用,PAK蛋白。这种相互作用对于多种肿瘤类型的进展至关重要。此类抑制剂在体内显示出抗癌功效。这里,我们描述了具有良好药物样特性的第二类CDC42抑制剂。在这里报道的25种化合物中,化合物15(ARN25499)是最佳的先导化合物,具有改善的药代动力学特征,增加生物利用度,和在体内PDX肿瘤小鼠模型中的功效。我们的结果表明,这些CDC42抑制剂代表了发现抗癌药物的一个有前途的化学类别,ARN25499作为临床前开发的额外先导候选物。
    We previously reported trisubstituted pyrimidine lead compounds, namely, ARN22089 and ARN25062, which block the interaction between CDC42 with its specific downstream effector, a PAK protein. This interaction is crucial for the progression of multiple tumor types. Such inhibitors showed anticancer efficacy in vivo. Here, we describe a second class of CDC42 inhibitors with favorable drug-like properties. Out of the 25 compounds here reported, compound 15 (ARN25499) stands out as the best lead compound with an improved pharmacokinetic profile, increased bioavailability, and efficacy in an in vivo PDX tumor mouse model. Our results indicate that these CDC42 inhibitors represent a promising chemical class toward the discovery of anticancer drugs, with ARN25499 as an additional lead candidate for preclinical development.
    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
    17β-雌二醇(E2)可以抑制女性心力衰竭(HF)患者的心脏纤维化并激活细胞分裂周期42(Cdc42),然而,尚不清楚17β-雌二醇(E2)是否可以通过调节细胞分裂周期42(Cdc42)来改善TGF-β1刺激的小鼠心脏成纤维细胞(MCF)的分化和胶原蛋白合成。本研究旨在探讨雌激素和Cdc42在预防心肌纤维化中的作用及其分子机制。用ELISA法测定心力衰竭(HF)患者血清中E2和Cdc42的水平,蛋白质印迹法用于测量Cdc42在TGF-β1刺激的永生化MCF中的表达水平。用Cdc42过表达(OE)慢病毒或小干扰RNA(siRNA)转染MCF,或用Cdc42抑制剂(MLS-573151)处理,Cdc42的功能是通过蛋白质印迹法评估的,免疫荧光染色,逆转录-定量PCR和双荧光素酶报告基因测定。进行蛋白质印迹和免疫荧光染色以验证E2对TGF-β1刺激的MCF的保护作用。以及保护作用与Cdc42之间的关联。结果表明,心力衰竭患者血清中Cdc42水平升高,并与E2水平呈正相关;然而,TGF-β1刺激的MCF中Cdc42水平降低。Cdc42抑制MCF分化和胶原合成,如α-平滑肌肌动蛋白的蛋白质表达所示,胶原蛋白I和胶原蛋白III。机械上,Cdc42通过促进p21(RAC1)激活的激酶1(Pak1)/JNK/c‑Jun信号通路蛋白的表达和抑制Tgfb1基因启动子的活性来抑制TGF‑β1的转录。此外,E2抑制TGF-β1刺激的MCF的分化和胶原合成,并促进Pak1,JNK和c‑Jun的蛋白表达,与Cdc42的作用一致,而当Cdc42被击倒时,E2的作用被消除。上述发现表明,E2可以通过调节Cdc42和下游Pak1/JNK/c‑Jun信号通路来抑制TGF‑β1刺激的MCF中的分化和胶原蛋白合成。
    17β‑estradiol (E2) can inhibit cardiac fibrosis in female patients with heart failure (HF) and activate cell division cycle 42 (Cdc42), however it is unknown whether 17β‑estradiol (E2) can ameliorate differentiation and collagen synthesis in TGF‑β1‑stimulated mouse cardiac fibroblasts (MCFs) by regulating cell division cycle 42 (Cdc42). The present study aimed to investigate the roles of estrogen and Cdc42 in preventing myocardial fibrosis and the underlying molecular mechanisms. An ELISA was used to measure the levels of E2 and Cdc42 in the serum of patients with heart failure (HF), and western blotting was used to measure the expression levels of Cdc42 in TGF‑β1‑stimulated immortalized MCFs. MCFs were transfected with a Cdc42 overexpression (OE) lentivirus or small interfering RNA (siRNA), or treated with a Cdc42 inhibitor (MLS‑573151), and the function of Cdc42 was assessed by western blotting, immunofluorescence staining, reverse transcription‑quantitative PCR and dual‑luciferase reporter assays. Western blotting and immunofluorescence staining were performed to verify the protective effect of E2 on TGF‑β1‑stimulated MCFs, and the association between the protective effect and Cdc42. The results demonstrated that Cdc42 levels were increased in the serum of patients with HF and were positively correlated with the levels of E2; however, Cdc42 levels were decreased in TGF‑β1‑stimulated MCFs. Cdc42 inhibited MCF differentiation and collagen synthesis, as indicated by the protein expression of α‑smooth muscle actin, collagen I and collagen III. Mechanistically, Cdc42 inhibited the transcription of TGF‑β1 by promoting the expression of p21 (RAC1)‑activated kinase 1 (Pak1)/JNK/c‑Jun signaling pathway proteins and inhibiting the activity of the Tgfb1 gene promoter. In addition, E2 inhibited the differentiation and collagen synthesis of TGF‑β1‑stimulated MCFs, and promoted the protein expression of Pak1, JNK and c‑Jun, consistent with the effects of Cdc42, whereas the effects of E2 were abolished when Cdc42 was knocked down. The aforementioned findings suggested that E2 could inhibit differentiation and collagen synthesis in TGF‑β1‑stimulated MCFs by regulating Cdc42 and the downstream Pak1/JNK/c‑Jun signaling pathway.
    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:核苷酸结合寡聚化结构域的成员,富含亮氨酸的重复序列和含pyrin结构域(NLRP)家族调节各种生理和病理过程。然而,在卵母细胞减数分裂的不对称分裂过程中,没有人显示出调节肌动蛋白帽的形成或纺锤体易位I。NLRP4E已被报道为女性生育力的候选蛋白,但它的功能是未知的。
    方法:免疫荧光,逆转录聚合酶链反应(RT-PCR),采用免疫印迹法检测NLRP4E及其相关蛋白在小鼠卵母细胞中的定位和表达水平。小干扰RNA(siRNA)和抗体转染用于敲除NLRP4E和其他蛋白.免疫沉淀(IP)-质谱用于鉴定与NLRP4E相互作用的潜在蛋白质。使用共免疫沉淀(Co-IP)来验证蛋白质相互作用。将野生型(WT)或突变型NLRP4E信使RNA(mRNA)注射到卵母细胞中用于拯救实验。体外磷酸化用于检查NLRP4E对类固醇受体共激活剂(SRC)的激活。
    结果:与其他NLRP4成员相比,NLRP4E在卵母细胞中更占优势。NLRP4E敲低显著抑制肌动蛋白帽的形成和纺锤体向帽区的易位,导致减数分裂I结束时极体挤压失败,GRIN1和GANO1通过在Ser429和Thr430磷酸化激活NLRP4E;p-NLRP4E易位并在纺锤体易位过程中在肌动蛋白帽区域积累。接下来,我们发现p-NLRP4E在Tyr418处直接磷酸化SRC,而p-SRC负调节p-CDC42-S71,这是一种无活性形式的CDC42,以GTP结合形式促进肌动蛋白帽形成和纺锤体易位.
    结论:GRIN1和GANO1激活的NLRP4E在减数分裂过程中通过上调p-SRC-Tyr418和下调p-CDC42-S71来调节肌动蛋白帽的形成和纺锤体向帽区的易位。
    BACKGROUND: Members of the nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing (NLRP) family regulate various physiological and pathological processes. However, none have been shown to regulate actin cap formation or spindle translocation during the asymmetric division of oocyte meiosis I. NLRP4E has been reported as a candidate protein in female fertility, but its function is unknown.
    METHODS: Immunofluorescence, reverse transcription polymerase chain reaction (RT-PCR), and western blotting were employed to examine the localization and expression levels of NLRP4E and related proteins in mouse oocytes. small interfering RNA (siRNA) and antibody transfection were used to knock down NLRP4E and other proteins. Immunoprecipitation (IP)-mass spectrometry was used to identify the potential proteins interacting with NLRP4E. Coimmunoprecipitation (Co-IP) was used to verify the protein interactions. Wild type (WT) or mutant NLRP4E messenger RNA (mRNA) was injected into oocytes for rescue experiments. In vitro phosphorylation was employed to examine the activation of steroid receptor coactivator (SRC) by NLRP4E.
    RESULTS: NLRP4E was more predominant within oocytes compared with other NLRP4 members. NLRP4E knockdown significantly inhibited actin cap formation and spindle translocation toward the cap region, resulting in the failure of polar body extrusion at the end of meiosis I. Mechanistically, GRIN1, and GANO1 activated NLRP4E by phosphorylation at Ser429 and Thr430; p-NLRP4E is translocated and is accumulated in the actin cap region during spindle translocation. Next, we found that p-NLRP4E directly phosphorylated SRC at Tyr418, while p-SRC negatively regulated p-CDC42-S71, an inactive form of CDC42 that promotes actin cap formation and spindle translocation in the GTP-bound form.
    CONCLUSIONS: NLRP4E activated by GRIN1 and GANO1 regulates actin cap formation and spindle translocation toward the cap region through upregulation of p-SRC-Tyr418 and downregulation of p-CDC42-S71 during meiosis I.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号