Gα13

G α 13
  • 文章类型: Journal Article
    Pharyngeal endoderm cells undergo convergence and extension (C&E), which is essential for endoderm pouch formation and craniofacial development. Our prior work implicates Gα13/RhoA-mediated signaling in regulating this process, but underlying mechanisms remain unclear. Here, we used endoderm-specific transgenic and Gα13 mutant zebrafish to demonstrate that Gα13 plays a crucial role in pharyngeal endoderm C&E by regulating RhoA activation and E-cadherin expression. We showed that during C&E, endodermal cells gradually establish stable cell-cell contacts, acquire apical-basal polarity, and undergo actomyosin-driven apical constriction, processes that require Gα13. Additionally, we found Gα13-deficient embryos exhibit reduced E-cadherin expression, partially contributing to endoderm C&E defects. Notably, interfering with RhoA function disrupts spatial actomyosin activation without affecting E-cadherin expression. Collectively, our findings identify critical cellular processes for pharyngeal endoderm C&E and reveal that Gα13 controls this through two independent pathways: modulating RhoA activation and regulating E-cadherin expression, thus unveiling intricate mechanisms governing pharyngeal endoderm morphogenesis.
    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
    炎症性肠病由炎症和免疫失调定义。这项研究调查了Gα13肝脏特异性敲除(LKO)对葡聚糖硫酸钠(DSS)诱导的小鼠与高脂饮食(HFD)的近端和远端结肠的影响。HFD改善体重增加和疾病活动指数评分。Gα13LKO没有改善。在近端结肠,HFD增强了DSS对Il6的作用,这在Gα13LKO小鼠中未观察到。在远端结肠,HFD加DSS相反地增强了Gα13LKO中Tnfa和Cxcl10mRNA的增加,而不是WT。Il6水平保持不变。使用Gα13LKO肝脏的生物信息学方法显示了胆汁酸和胆固醇代谢相关的基因集。胆酸和鹅去氧胆酸水平在用DSS处理的小鼠的肝脏中增加,这被Gα13LKO逆转了。值得注意的是,用DSS处理的小鼠显示肝脏ABCB11,CYP7B1,CYP7A1和CYP8B1的减少,这被Gα13LKO逆转。总的来说,饲喂HFD可增强DSS对WT近端结肠Il6的影响,但不是Gα13LKO小鼠,并增强DSS对Gα13LKO小鼠远端结肠Tnfa和Cxcl10的作用,提示炎症细胞因子的位点特异性变化,可能是由于BA合成和排泄的变化。
    Inflammatory bowel disease is defined by inflammation and immune dysregulation. This study investigated the effects of Gα13 liver-specific knockout (LKO) on proximal and distal colons of dextran sodium sulfate (DSS)-induced mice in conjunction with a high-fat diet (HFD). HFD improved body weight gain and disease activity index scores. Gα13LKO exerted no improvement. In the proximal colon, HFD augmented the DSS effect on Il6, which was not observed in Gα13LKO mice. In the distal colon, HFD plus DSS oppositely fortified an increase in Tnfa and Cxcl10 mRNA in Gα13LKO but not WT. Il6 levels remained unchanged. Bioinformatic approaches using Gα13LKO livers displayed bile acid and cholesterol metabolism-related gene sets. Cholic acid and chenodeoxycholic acid levels were increased in the liver of mice treated with DSS, which was reversed by Gα13LKO. Notably, mice treated with DSS showed a reduction in hepatic ABCB11, CYP7B1, CYP7A1, and CYP8B1, which was reversed by Gα13LKO. Overall, feeding HFD augments the effect of DSS on Il6 in the proximal colon of WT, but not Gα13LKO mice, and enhances DSS effect on Tnfa and Cxcl10 in the distal colon of Gα13LKO mice, suggesting site-specific changes in the inflammatory cytokines, potentially resulting from changes in BA synthesis and excretion.
    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
    由GNA13和GNA12基因编码的Gα13和Gα12,分别,是Gα蛋白G12家族的成员,以及它们相关的Gβγ亚基,介导来自特定G蛋白偶联受体(GPCRs)的信号传导。晚期前列腺癌的GPCRs表达增加,如CXC基序趋化因子受体4(CXCR4),溶血磷脂酸受体(LPAR),和蛋白酶激活受体1(PAR-1)。这些GPCRs通过G12家族发出信号,或者专门通过Gα13,通常除了其他G蛋白。Gα13的作用可能与Gα12的作用不同,并且Gα13在前列腺癌的发生和发展中的作用在很大程度上尚未被研究。Gα13对前列腺癌细胞迁移和侵袭的致癌作用已被证实。但对线粒体功能和氧化应激等其他生物过程知之甚少。目前关于Gα13和氧化应激之间联系的知识是基于GPCR-Gα13信号传导降低超氧化物水平的动物研究,组成型活性Gα13的过表达促进了抗氧化基因的激活。在人体样本中,线粒体超氧化物歧化酶2(SOD2)与前列腺癌风险和预后Gleason分级相关。然而,SOD2在前列腺癌细胞中的过表达对基础和氧化应激条件下的细胞生长和存活产生了矛盾的结果。因此,有必要探讨Gα13在前列腺癌发生发展中的作用,以及在氧化应激条件下Gα13对前列腺癌细胞生长中SOD2的影响。
    Gα13 and Gα12, encoded by the GNA13 and GNA12 genes, respectively, are members of the G12 family of Gα proteins that, along with their associated Gβγ subunits, mediate signaling from specific G protein-coupled receptors (GPCRs). Advanced prostate cancers have increased expression of GPCRs such as CXC Motif Chemokine Receptor 4 (CXCR4), lysophosphatidic acid receptor (LPAR), and protease activated receptor 1 (PAR-1). These GPCRs signal through either the G12 family, or through Gα13 exclusively, often in addition to other G proteins. The effect of Gα13 can be distinct from that of Gα12, and the role of Gα13 in prostate cancer initiation and progression is largely unexplored. The oncogenic effect of Gα13 on cell migration and invasion in prostate cancer has been characterized, but little is known about other biological processes such as mitochondrial function and oxidative stress. Current knowledge on the link between Gα13 and oxidative stress is based on animal studies in which GPCR-Gα13 signaling decreased superoxide levels, and the overexpression of constitutively active Gα13 promoted antioxidant gene activation. In human samples, mitochondrial superoxide dismutase 2 (SOD2) correlates with prostate cancer risk and prognostic Gleason grade. However, overexpression of SOD2 in prostate cancer cells yielded conflicting results on cell growth and survival under basal versus oxidative stress conditions. Hence, it is necessary to explore the effect of Gα13 on prostate cancer tumorigenesis, as well as the effect of Gα13 on SOD2 in prostate cancer cell growth under oxidative stress conditions.
    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:我们以前的研究表明,麻醉和手术可以加重5XFAD转基因(Tg)小鼠的认知障碍,这种恶化与tau蛋白过度磷酸化有关。我们先前鉴定GNA13(编码Gα13的基因)是具有tau过度磷酸化的hub基因。
    目的:本研究旨在进一步探讨Gα13介导的信号通路是否在该过程中作为促进因子调控cofilin激活和自噬损伤的机制。
    方法:将5XFADTg小鼠及其同窝(LM)小鼠随机分为四组:LM对照组,LM麻醉/手术组,AD控制组,和AD麻醉/手术组。对于麻醉/手术组的小鼠,腹部手术在1.4%异氟烷麻醉下进行,然后持续吸入麻醉长达2小时。
    结果:与AD对照组相比,Gα13,ROCK2,LPAR5和p-tau/tau46比值的蛋白质水平增加,而在AD麻醉/手术组中,p-cofilin/cofilin蛋白表达比率降低。然而,LM组之间这些蛋白质水平的差异不显著.
    结论:这项研究表明,麻醉和手术可能会加剧5XFADTg小鼠的p-tau积累,但不会加剧LM小鼠的p-tau积累。这可能与通过Gα13介导的信号级联激活cofilin密切相关。
    Our previous studies indicated that anesthesia and surgery could aggravate cognitive impairment of 5XFAD transgenic (Tg) mice, and this aggravation was associated with tau hyperphosphorylation. We previously identified that GNA13 (the gene encoding Gα13) was a hub gene with tau hyperphosphorylation.
    This study aims to further investigate the mechanism that whether the Gα13-mediated signaling pathway acts as an instigator to regulate cofilin activation and autophagy impairment in this process.
    5XFAD Tg mice and their littermate (LM) mice were randomly allocated into four groups: LM Control group, LM Anesthesia/Surgery group, AD Control group, and AD Anesthesia/Surgery group. For mice in the Anesthesia/Surgery groups, abdominal surgery was performed under 1.4% isoflurane anesthesia followed by sustaining anesthetic inhalation for up to 2 h.
    Compared with the AD Control group, protein levels of Gα13, ROCK2, LPAR5, and p-tau/tau46 ratio were increased, while p-cofilin/cofilin protein expression ratio was decreased in the AD Anesthesia/Surgery group. However, the differences in these protein levels were not significant among LM groups.
    This study demonstrated that anesthesia and surgery might exacerbate p-tau accumulation in 5XFAD Tg mice but not in LM mice. And this might be closely related to cofilin activation via Gα13-mediated signaling cascade.
    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:突触后密度95/disk-large/ZO-1Rho鸟嘌呤核苷酸交换因子(PDZ-RhoGEF,PRG)充当激活的Gα13的RhoGEF,并在各种病理过程中向下游信号通路传递激活信号。尽管激活的Gα13(鸟嘌呤核苷酸结合蛋白α13;异源三聚体G蛋白)的促肥大作用已得到证实,PDZ-RhoGEF在病理性心肌肥厚中的作用尚不清楚.
    方法:制备基因工程小鼠和新生大鼠心室肌细胞,研究PRG在病理性心肌肥厚中的作用。在心肌和新生大鼠心室肌细胞中测量了促肥大刺激诱导的形态和细胞内信号传导的变化。此外,使用多种分子方法学来鉴定PDZ-RhoGEF功能背后的精确分子机制.
    结果:PDZ-RhoGEF在肥大的心脏和新生大鼠心室肌细胞中的表达增加。在促肥大刺激下,PDZ-RhoGEF缺乏的心脏显示出减轻的心肌细胞增大和减轻的胶原沉积和改善的心脏功能,而心脏和新生大鼠心室肌细胞的不良肥大反应因PDZ-RhoGEF的过表达而明显加剧。机械上,RhoA(ras同系物家族成员A)依赖性信号通路可能是PDZ-RhoGEF在肥厚性重塑中的下游效应子,如使用RhoA抑制剂和显性阴性RhoA的救援实验所证实的。此外,PDZ-RhoGEF与激活的Gα13相关,并有助于Gα13介导的RhoA依赖性信号的激活。
    结论:我们的数据提供了第一个证据,表明PDZ-RhoGEF通过将激活的Gα13与RhoA依赖性信号通路联系起来促进病理性心脏肥大。因此,PDZ-RhoGEF有可能成为病理性心脏肥大的诊断标志物或治疗靶标。
    Postsynaptic density 95/disk-large/ZO-1 Rho guanine nucleotide exchange factor (PDZ-RhoGEF, PRG) functions as a RhoGEF for activated Gα13 and transmits activation signals to downstream signaling pathways in various pathological processes. Although the prohypertrophic effect of activated Gα13 (guanine nucleotide binding protein alpha 13; a heterotrimeric G protein) is well-established, the role of PDZ-RhoGEF in pathological cardiac hypertrophy is still obscure.
    Genetically engineered mice and neonatal rat ventricular myocytes were generated to investigate the function of PRG in pathological myocardial hypertrophy. The prohypertrophic stimuli-induced alternations in the morphology and intracellular signaling were measured in myocardium and neonatal rat ventricular myocytes. Furthermore, multiple molecular methodologies were used to identify the precise molecular mechanisms underlying PDZ-RhoGEF function.
    Increased PDZ-RhoGEF expression was documented in both hypertrophied hearts and neonatal rat ventricular myocytes. Upon prohypertrophic stimuli, the PDZ-RhoGEF-deficient hearts displayed alleviated cardiomyocyte enlargement and attenuated collagen deposition with improved cardiac function, whereas the adverse hypertrophic responses in hearts and neonatal rat ventricular myocytes were markedly exaggerated by PDZ-RhoGEF overexpression. Mechanistically, RhoA (ras homolog family member A)-dependent signaling pathways may function as the downstream effectors of PDZ-RhoGEF in hypertrophic remodeling, as confirmed by rescue experiments using a RhoA inhibitor and dominant-negative RhoA. Furthermore, PDZ-RhoGEF is associated with activated Gα13 and contributes to Gα13-mediated activation of RhoA-dependent signaling.
    Our data provide the first evidence that PDZ-RhoGEF promotes pathological cardiac hypertrophy by linking activated Gα13 to RhoA-dependent signaling pathways. Therefore, PDZ-RhoGEF has the potential to be a diagnostic marker or therapeutic target for pathological cardiac hypertrophy.
    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
    Gα13,G12/13亚家族的异源三聚体G蛋白α亚基,是多种癌症类型的致癌驱动因素。与其他G蛋白亚家族不同,它们通过氨基酸取代消除它们的失活而促进癌症进展,固有的GTP酶活性,Gα13在肿瘤中很少携带这样的突变,而是似乎通过作为野生型形式的过表达来刺激异常细胞生长。目前还不知道为什么这种效应是G12/13亚家族独有的,也没有阐明过表达的Gα13促进肿瘤进展的机制。使用报道基因测定HEK293细胞中血清反应因子(SRF)介导的转录,我们发现短暂表达,野生型Gα13产生稳健的SRF信号,大约是GTP酶缺陷型Gα13振幅的一半。当表位标记时,野生型Gα13在细胞中向上滴定,观察到SRF刺激的急剧增加与Gα13从膜相关部分向可溶性部分的“溢出”同时发生。过表达G蛋白β和γ亚基导致该信号降低和野生型Gα13转移回膜级分,表明αβγ异源三聚体的化学计量失衡导致过表达的Gα13的异常亚细胞定位和信号传导。我们还检查了野生型Gα13对SRF的信号传导的酰化要求。与GTP酶缺陷型Gα13相似,野生型α亚基的S-棕榈酰化是SRF激活所必需的,但可以在功能上被N末端肉豆蔻酰化的工程化位点取代。然而,在野生型和GTPase缺陷型Gα13之间观察到关键差异:而缺乏棕榈酰化位点的后一种蛋白质在其SRF信号传导中被工程化的多碱基序列或C端异戊二烯化位点拯救,这些基序无法通过野生型恢复信令,非棕榈酰化的Gα13。这些发现阐明了过度表达机制的几个组成部分,野生型Gα13有助于生长和致瘤信号,并且与GTP酶缺陷型Gα13相比,其对翻译后修饰的要求更严格。
    Gα13, a heterotrimeric G protein α subunit of the G12/13 subfamily, is an oncogenic driver in multiple cancer types. Unlike other G protein subfamilies that contribute to cancer progression via amino acid substitutions that abolish their deactivating, intrinsic GTPase activity, Gα13 rarely harbors such mutations in tumors and instead appears to stimulate aberrant cell growth via overexpression as a wildtype form. It is not known why this effect is exclusive to the G12/13 subfamily, nor has a mechanism been elucidated for overexpressed Gα13 promoting tumor progression. Using a reporter gene assay for serum response factor (SRF)-mediated transcription in HEK293 cells, we found that transiently expressed, wildtype Gα13 generates a robust SRF signal, approximately half the amplitude observed for GTPase-defective Gα13. When epitope-tagged, wildtype Gα13 was titrated upward in cells, a sharp increase in SRF stimulation was observed coincident with a \"spillover\" of Gα13 from membrane-associated to a soluble fraction. Overexpressing G protein β and γ subunits caused both a decrease in this signal and a shift of wildtype Gα13 back to the membranous fraction, suggesting that stoichiometric imbalance in the αβγ heterotrimer results in aberrant subcellular localization and signalling by overexpressed Gα13. We also examined the acylation requirements of wildtype Gα13 for signalling to SRF. Similar to GTPase-defective Gα13, S-palmitoylation of the wildtype α subunit was necessary for SRF activation but could be replaced functionally by an engineered site for N-terminal myristoylation. However, a key difference was observed between wildtype and GTPase-defective Gα13: whereas the latter protein lacking palmitoylation sites was rescued in its SRF signalling by either an engineered polybasic sequence or a C-terminal isoprenylation site, these motifs failed to restore signalling by wildtype, non-palmitoylated Gα13. These findings illuminate several components of the mechanism in which overexpressed, wildtype Gα13 contributes to growth and tumorigenic signalling, and reveal greater stringency in its requirements for post-translational modification in comparison to GTPase-defective Gα13.
    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
    G蛋白偶联受体(GPCRs),作为人体中最大的受体家族,涉及许多疾病的病理机制。异三聚体G蛋白代表主要的分子开关并从活化的GPCRs接收细胞表面信号。越来越多的证据表明,Gα12亚家族(Gα12/13)介导的信号传导在细胞功能和各种病理过程中起着至关重要的作用。目前对Gα12/13生理病理功能的研究在不断扩大,已经在多种人类疾病中发现了Gα12/13表达水平的变化。然而,对Gα12/13的机理研究比较分散。这篇综述简要描述了Gα12/13亚型的结构序列,并介绍了GPCRs和非GPCRs与Gα12/13的偶联。Gα12/13对RhoA等信号通路的影响及其在细胞增殖中的作用,迁移,和免疫细胞功能,正在讨论。最后,我们关注Gα12/13在癌症中的病理影响,炎症,代谢性疾病,纤维化疾病,和循环障碍被带到焦点。
    G protein-coupled receptors (GPCRs), as the largest family of receptors in the human body, are involved in the pathological mechanisms of many diseases. Heterotrimeric G proteins represent the main molecular switch and receive cell surface signals from activated GPCRs. Growing evidence suggests that Gα12 subfamily (Gα12/13)-mediated signaling plays a crucial role in cellular function and various pathological processes. The current research on the physiological and pathological function of Gα12/13 is constantly expanding, Changes in the expression levels of Gα12/13 have been found in a wide range of human diseases. However, the mechanistic research on Gα12/13 is scattered. This review briefly describes the structural sequences of the Gα12/13 isoforms and introduces the coupling of GPCRs and non-GPCRs to Gα12/13. The effects of Gα12/13 on RhoA and other signaling pathways and their roles in cell proliferation, migration, and immune cell function, are discussed. Finally, we focus on the pathological impacts of Gα12/13 in cancer, inflammation, metabolic diseases, fibrotic diseases, and circulatory disorders are brought to focus.
    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
    Gα13从G蛋白偶联受体转导信号。虽然Gα13在淋巴瘤中起肿瘤抑制因子的作用,目前尚不清楚Gα13在基因工程小鼠(GEM)上皮癌模型中是否具有致瘤性或抑瘤性.这里,我们显示Kras/Tp53(KPC)GEM模型中Gα13的缺失促进了高分化肿瘤并降低了生存率.机械上,在具有Gα13缺失的KPC小鼠中发展的肿瘤表现出增加的E-cadherin表达和mTOR信号传导。重要的是,具有低Gα13表达的人胰腺导管腺癌(PDAC)肿瘤也表现出增加的E-cadherin表达和mTOR信号传导。用mTOR抑制剂雷帕霉素治疗通过促进细胞死亡来降低具有Gα13损失的同基因KPC肿瘤的生长。这项工作在KPCGEM模型中确立了Gα13在胰腺肿瘤发生中的肿瘤抑制作用,并建议在Gα13丢失的人PDAC肿瘤中靶向mTOR。
    Gα13 transduces signals from G-protein-coupled receptors. While Gα13 functions as a tumor suppressor in lymphomas, it is not known whether Gα13 is pro-tumorigenic or tumor suppressive in genetically engineered mouse (GEM) models of epithelial cancers. Here, we show that loss of Gα13 in the Kras/Tp53 (KPC) GEM model promotes well-differentiated tumors and reduces survival. Mechanistically, tumors developing in KPC mice with Gα13 loss exhibit increased E-cadherin expression and mTOR signaling. Importantly, human pancreatic ductal adenocarcinoma (PDAC) tumors with low Gα13 expression also exhibit increased E-cadherin expression and mTOR signaling. Treatment with the mTOR inhibitor rapamycin decreases the growth of syngeneic KPC tumors with Gα13 loss by promoting cell death. This work establishes a tumor-suppressive role of Gα13 in pancreatic tumorigenesis in the KPC GEM model and suggests targeting mTOR in human PDAC tumors with Gα13 loss.
    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
    小胶质细胞是CNS的固有免疫细胞,其响应于多种刺激而被激活。这种表型变化旨在维持局部稳态,还通过遏制侮辱和修复损害。所有这些过程都受到严格调节和协调,小胶质细胞恢复稳态的失败可能导致神经炎症的发展,从而促进病理状况的进展。的确,慢性小胶质细胞激活通常被认为是许多神经系统疾病的标志,尤其是在早期阶段。许多复杂的途径,包括细胞骨架重塑,参与激活过程中发生的小胶质细胞表型和形态变化的控制。在这项工作中,我们关注小胶质细胞暴露于促炎性刺激时,小GTP酶Gα13及其在RhoA和Rac1信号交叉通路中的作用.我们建议Gα13直接参与FAK介导的细胞骨架重排,LIMK/cofilin,和Rac1在小胶质细胞激活期间。事实上,我们显示Gα13敲低显著抑制LPS诱导的小胶质细胞活化,就形态和迁移的变化而言,通过调节FAK及其下游效应器之一,Rac1.总之,我们认为Gα13是小胶质细胞激活过程中形态和功能特性调节的关键因素,这可能成为控制小胶质细胞炎症的干预目标。
    Microglia are the resident immune cells of the CNS that are activated in response to a variety of stimuli. This phenotypical change is aimed to maintain the local homeostasis, also by containing the insults and repair the damages. All these processes are tightly regulated and coordinated and a failure in restoring homeostasis by microglia can result in the development of neuroinflammation that can facilitate the progression of pathological conditions. Indeed, chronic microglia activation is commonly recognized as a hallmark of many neurological disorders, especially at an early stage. Many complex pathways, including cytoskeletal remodeling, are involved in the control of the microglial phenotypical and morphological changes that occur during activation. In this work, we focused on the small GTPase Gα13 and its role at the crossroad between RhoA and Rac1 signaling when microglia is exposed to pro-inflammatory stimulation. We propose the direct involvement of Gα13 in the cytoskeletal rearrangements mediated by FAK, LIMK/cofilin, and Rac1 during microglia activation. In fact, we show that Gα13 knockdown significantly inhibited LPS-induced microglial cell activation, in terms of both changes in morphology and migration, through the modulation of FAK and one of its downstream effectors, Rac1. In conclusion, we propose Gα13 as a critical factor in the regulation of morphological and functional properties of microglia during activation, which might become a target of intervention for the control of microglia inflammation.
    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
    The G12/13 subfamily of heterotrimeric guanine nucleotide binding proteins comprises the α subunits Gα12 and Gα13, which transduce signals for cell growth, cytoskeletal rearrangements, and oncogenic transformation. In an increasing range of cancers, overexpressed Gα12 or Gα13 are implicated in aberrant cell proliferation and/or metastatic invasion. Although Gα12 and Gα13 bind non-redundant sets of effector proteins and participate in unique signalling pathways, the structural features responsible for functional differences between these α subunits are largely unknown. Invertebrates encode a single G12/13 homolog that participates in cytoskeletal changes yet appears to lack signalling to SRF (serum response factor), a transcriptional activator stimulated by mammalian Gα12 and Gα13 to promote growth and tumorigenesis. Our previous studies identified an evolutionarily divergent region in Gα12 for which replacement by homologous sequence from Drosophila melanogaster abolished SRF signalling, whereas the same invertebrate substitution was fully tolerated in Gα13 [Montgomery et al. (2014) Mol. Pharmacol. 85: 586]. These findings prompted our current approach of evolution-guided mutagenesis to identify fine structural features of Gα12 and Gα13 that underlie their respective SRF activation mechanisms. Our results identified two motifs flanking the α4 helix that play a key role in Gα12 signalling to SRF. We found the region encompassing these motifs to provide an interacting surface for multiple Gα12-specific target proteins that fail to bind Gα13. Adjacent to this divergent region, a highly-conserved domain was vital for SRF activation by both Gα12 and Gα13. However, dissection of this domain using invertebrate substitutions revealed different signalling mechanisms in these α subunits and identified Gα13-specific determinants of binding Rho-specific guanine nucleotide exchange factors. Furthermore, invertebrate substitutions in the C-terminal, α5 helical region were selectively disruptive to Gα12 signalling. Taken together, our results identify key structural features near the C-terminus that evolved after the divergence of Gα12 and Gα13, and should aid the development of agents to selectively manipulate signalling by individual α subunits of the G12/13 subfamily.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号