rho GTP-Binding Proteins

rho GTP 结合蛋白
  • 文章类型: Journal Article
    内皮细胞和许多其他细胞的迁移需要驱动局部细胞形状变化的突伸和收缩性细胞骨架重排的时空调节。出乎意料的是,小GTPaseRho,细胞运动的关键调节器,据报道,在局部细胞突起和缩回中都有活性,提出了Rho活性如何协调细胞迁移的问题。这里,我们表明,Rho活动在局部突起中不存在,在缩回过程中活跃。在撤回期间,Rho快速激活ezrin-radixin-moesin蛋白(ERMs)以增加肌动蛋白膜附着,and,延迟,非肌肉肌球蛋白2(NM2)。Rho活动是兴奋的,NM2充当慢负反馈调节器。引人注目的是,抑制SLK/LOK激酶,Rho通过它激活ERM,导致细胞形态拉长,受损的Rho诱导的细胞收缩,并恢复了Rho引起的气泡。一起,我们的研究表明,Rho活性通过依次增强ERM介导的肌动蛋白膜附着以促进力传递和NM2依赖性收缩性来驱动收缩.
    Migration of endothelial and many other cells requires spatiotemporal regulation of protrusive and contractile cytoskeletal rearrangements that drive local cell shape changes. Unexpectedly, the small GTPase Rho, a crucial regulator of cell movement, has been reported to be active in both local cell protrusions and retractions, raising the question of how Rho activity can coordinate cell migration. Here, we show that Rho activity is absent in local protrusions and active during retractions. During retractions, Rho rapidly activated ezrin-radixin-moesin proteins (ERMs) to increase actin-membrane attachment, and, with a delay, nonmuscle myosin 2 (NM2). Rho activity was excitable, with NM2 acting as a slow negative feedback regulator. Strikingly, inhibition of SLK/LOK kinases, through which Rho activates ERMs, caused elongated cell morphologies, impaired Rho-induced cell contractions, and reverted Rho-induced blebbing. Together, our study demonstrates that Rho activity drives retractions by sequentially enhancing ERM-mediated actin-membrane attachment for force transmission and NM2-dependent contractility.
    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
    血小板由巨核细胞(MK)产生,他们的父细胞,在骨髓中。一旦成熟,MK穿过正弦血管,并且初始突起进一步作为前血小板或芽伸长以释放血小板。控制启动前血小板和血小板形成的决定的机制尚不清楚。这里,我们表明,微环境的机械特性阻止骨髓基质中的前血小板和血小板释放,同时允许这一过程在血液中。骨髓抑制后骨髓封闭的丧失导致不适当的前血小板和血小板释放到血管外空间。我们进一步使用惰性粘弹性水凝胶来评估压缩应力的影响。转录分析表明,三维凝胶中的培养物诱导与Rho-GTP酶途径相关的基因上调。我们发现更高的Rho-GTP酶激活,肌球蛋白轻链磷酸化和F-肌动蛋白在机械约束下,而前血小板形成被抑制。使用latrunculin-A减少F-肌动蛋白可促进凝胶内的微管依赖性出芽和前血小板延伸。此外,完整骨髓离体暴露于latrunculin-A触发了间质空间中的前血小板延伸。在体内,这种限制介导的高细胞内张力是形成外围区的原因,独特的富含肌动蛋白的结构。细胞骨架重组在到达液体环境时诱导外围区的消失以促进前血小板和血小板形成。因此,我们的数据为防止骨髓基质中异位血小板释放的机制提供了见解.鉴定此类途径对于理解改变骨髓力学的病理如化疗或骨髓纤维化尤其重要。
    Blood platelets are produced by megakaryocytes (MKs), their parent cells, which are in the bone marrow. Once mature, MK pierces through the sinusoid vessel, and the initial protrusion further elongates as proplatelet or buds to release platelets. The mechanisms controlling the decision to initiate proplatelet and platelet formation are unknown. Here, we show that the mechanical properties of the microenvironment prevent proplatelet and platelet release in the marrow stroma while allowing this process in the bloodstream. Loss of marrow confinement following myelosuppression led to inappropriate proplatelet and platelet release into the extravascular space. We further used an inert viscoelastic hydrogel to evaluate the impact of compressive stress. Transcriptional analysis showed that culture in three-dimensional gel induced upregulation of genes related to the Rho-GTPase pathway. We found higher Rho-GTPase activation, myosin light chain phosphorylation and F-actin under mechanical constraints while proplatelet formation was inhibited. The use of latrunculin-A to decrease F-actin promoted microtubule-dependent budding and proplatelet extension inside the gel. Additionally, ex vivo exposure of intact bone marrow to latrunculin-A triggered proplatelet extensions in the interstitial space. In vivo, this confinement-mediated high intracellular tension is responsible for the formation of the peripheral zone, a unique actin-rich structure. Cytoskeleton reorganization induces the disappearance of the peripheral zone upon reaching a liquid milieu to facilitate proplatelet and platelet formation. Hence, our data provide insight into the mechanisms preventing ectopic platelet release in the marrow stroma. Identifying such pathways is especially important for understanding pathologies altering marrow mechanics such as chemotherapy or myelofibrosis.
    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
    ARHGAP25是免疫过程中的关键分子,作为Rac特异性GTP酶激活蛋白。它在细胞迁移和吞噬细胞功能中的作用,影响复杂的免疫疾病如类风湿性关节炎的结果,使其成为药物研究的有希望的目标。尽管它很重要,我们对其细胞内相互作用的了解仍然有限。这项研究采用了谷胱甘肽S-转移酶(GST)标签下拉的蛋白质组学分析和中性粒细胞细胞裂解物的免疫共沉淀,揭示了76个潜在物理相互作用的候选者,补充了ARHGAP25的已知概况。值得注意的是,四个小GTPases(RAC2,RHOG,ARF4和RAB27A)对ARHGAP25表现出高亲和力。ARHGAP25-RAC2和ARHGAP25-RHOG相互作用似乎受到小GTP酶激活状态的影响,表明GTP-GDP周期依赖性相互作用。在计算机二聚体预测中,将ARHGAP25的GAP域确定为可靠的结合界面,表明其适合GTP水解。此外,Fc受体相关激酶的列表,磷酸酶,14-3-3成员中有三个被确定为潜在合作伙伴,电脑预测突出了八个结合位点,对ARHGAP25的潜在调控机制提出了新的见解。
    ARHGAP25, a crucial molecule in immunological processes, serves as a Rac-specific GTPase-activating protein. Its role in cell migration and phagocyte functions, affecting the outcome of complex immunological diseases such as rheumatoid arthritis, renders it a promising target for drug research. Despite its importance, our knowledge of its intracellular interactions is still limited. This study employed proteomic analysis of glutathione S-transferase (GST)-tag pulldowns and co-immunoprecipitation from neutrophilic granulocyte cell lysate, revealing 76 candidates for potential physical interactions that complement ARHGAP25\'s known profile. Notably, four small GTPases (RAC2, RHOG, ARF4, and RAB27A) exhibited high affinity for ARHGAP25. The ARHGAP25-RAC2 and ARHGAP25-RHOG interactions appeared to be affected by the activation state of the small GTPases, suggesting a GTP-GDP cycle-dependent interaction. In silico dimer prediction pinpointed ARHGAP25\'s GAP domain as a credible binding interface, suggesting its suitability for GTP hydrolysis. Additionally, a list of Fc receptor-related kinases, phosphatases, and three of the 14-3-3 members were identified as potential partners, with in silico predictions highlighting eight binding sites, presenting novel insight on a potential regulatory mechanism for ARHGAP25.
    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
    纤维化后的肺再生需要形成功能性的新血管,这对于气体交换和细胞与其他肺细胞的串扰至关重要。尚不清楚肺脉管系统如何在没有纤维化的情况下再生。这里,我们测试了N6-甲基腺苷(m6A)修饰叉头盒蛋白O1(Foxo1)mRNA在小鼠肺切除术(PNX)后肺再生中的作用,手术切除后肺再生模型。甲基转移酶样3(Mettl3)和Foxo1的内皮细胞(EC)特异性敲除引起非生产性肠套叠性血管生成(IA),这损害了再生和增强纤维化。这种非生产性IA的特征是内皮增殖增强,血管分裂增加,支柱EC数量增加。小鼠Mettl3的内皮选择性敲除刺激非生产性IA和PNX后促纤维化因子的上调,促进再生到纤维化转变。小鼠Foxo1基因中m6A修饰位点的EC特异性突变揭示内皮Mettl3修饰了Foxo1mRNA中的A504和A2035位点以维持促再生内皮糖酵解,确保生产性IA和肺再生而不纤维化。Mettl3-Foxo1信号传导的抑制刺激了糖酵解和过度增殖的6-磷酸果糖-2-激酶/果糖-2,6-双磷酸酶3(Pfkfb3)的子集,Ras同源家族成员J(Rhoj)+,人和小鼠肺纤维化中的血小板衍生生长因子亚基B(Pdgfb)ECs。抑制该Pfkfb3+Rhoj+Pdgfb+EC子集归一化IA,减轻纤维化,并在博来霉素(BLM)损伤的小鼠肺中恢复再生。我们发现小鼠脉管系统中Foxo1的m6A修饰在PNX和BLM损伤后促进肺再生而不是纤维化。
    Lung regeneration after fibrosis requires formation of functional new vasculature, which is essential for gas exchange and cellular cross-talk with other lung cells. It remains unknown how the lung vasculature can be regenerated without fibrosis. Here, we tested the role of N6-methyladenosine (m6A) modification of forkhead box protein O1 (Foxo1) mRNA in lung regeneration after pneumonectomy (PNX) in mice, a model for lung regrowth after surgical resection. Endothelial cell (EC)-specific knockout of methyltransferase-like 3 (Mettl3) and Foxo1 caused nonproductive intussusceptive angiogenesis (IA), which impaired regeneration and enhanced fibrosis. This nonproductive IA was characterized by enhanced endothelial proliferation and increased vascular splitting with increased numbers of pillar ECs. Endothelial-selective knockout of Mettl3 in mice stimulated nonproductive IA and up-regulation of profibrotic factors after PNX, promoting regeneration to fibrotic transition. EC-specific mutation of m6A modification sites in the Foxo1 gene in mice revealed that endothelial Mettl3 modified A504 and A2035 sites in the Foxo1 mRNA to maintain pro-regenerative endothelial glycolysis, ensuring productive IA and lung regeneration without fibrosis. Suppression of Mettl3-Foxo1 signaling stimulated a subset of hyperglycolytic and hyperproliferative 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3)+, Ras homolog family member J (Rhoj)+, and platelet-derived growth factor subunit B (Pdgfb)+ ECs in both human and mouse lungs with fibrosis. Inhibiting this Pfkfb3+Rhoj+Pdgfb+ EC subset normalized IA, alleviated fibrosis, and restored regeneration in bleomycin (BLM)-injured mouse lungs. We found that m6A modification of Foxo1 in the mouse vasculature promoted lung regeneration over fibrosis after PNX and BLM injury.
    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
    据报道,产前暴露于邻苯二甲酸二丁酯(DBP)会导致成年后代大鼠勃起功能障碍(ED)。然而,其潜在机制尚未完全理解。以前,我们发现DBP激活男性生殖系统中的RhoA/ROCK通路.这项研究调查了产前暴露于DBP如何激活RhoA/ROCK信号通路,导致雄性大鼠后代ED。将妊娠大鼠分为DBP暴露组和NC组,暴露组从妊娠第14-18天通过管饲法接受750毫克/千克/天(mg/kg/天)的DBP。DBP暴露激活了后代阴茎海绵体(CC)中的RhoA/ROCK途径,导致平滑肌细胞收缩,纤维化,和细胞凋亡,所有这些都有助于ED。体外实验证实DBP诱导CC平滑肌细胞凋亡和RhoA/ROCK通路激活。用ROCK抑制剂Y-27632治疗DBP暴露后代8周,显著改善平滑肌细胞状况,勃起功能,减少纤维化。因此,产前DBP暴露通过RhoA/ROCK途径激活诱导子代ED,和ROCK抑制剂Y-27632显示出作为DBP诱导的ED的有效治疗的潜力。
    Prenatal exposure to dibutyl phthalate (DBP) has been reported to cause erectile dysfunction (ED) in adult offspring rats. However, its underlying mechanisms are not fully understood. Previously, we found that DBP activates the RhoA/ROCK pathway in the male reproductive system. This study investigated how prenatal exposure to DBP activates the RhoA/ROCK signalling pathway, leading to ED in male rat offspring. Pregnant rats were stratified into DBP-exposed and NC groups, with the exposed group receiving 750 milligrams per kilogram per day (mg/kg/day) of DBP through gavage from days 14-18 of gestation. DBP exposure activated the RhoA/ROCK pathway in the penile corpus cavernosum (CC) of descendants, causing smooth muscle cell contraction, fibrosis, and apoptosis, all of which contribute to ED. In vitro experiments confirmed that DBP induces apoptosis and RhoA/ROCK pathway activation in CC smooth muscle cells. Treatment of DBP-exposed offspring with the ROCK inhibitor Y-27632 for 8 weeks significantly improved smooth muscle cell condition, erectile function, and reduced fibrosis. Thus, prenatal DBP exposure induces ED in offspring through RhoA/ROCK pathway activation, and the ROCK inhibitor Y-27632 shows potential as an effective treatment for DBP-induced ED.
    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:转移性前列腺癌是男性癌症相关发病率和死亡率的主要原因,然而,潜在的分子机制知之甚少。神经丛蛋白是信号素的跨膜受体,在许多形式的癌症中具有不同的作用。我们最近发现,一个单一的临床相关的特异性氨基酸变化(Proline1597亮氨酸,(P1597L)),在前列腺癌患者的转移性沉积物中发现,将PlexinB1从转移抑制基因转化为体内驱动前列腺癌转移的基因。然而,PlexinB1(P1597L)促进转移的机制尚不清楚。
    方法:使用GST-RalGDS或-GSTRAf1-RBD的下拉测定分别揭示突变型或野生型PlexinB1表达对Rap和Ras活性的影响。在GST下拉试验中评估蛋白质-蛋白质相互作用,通过免疫印迹的Akt/ERK磷酸化和通过用环己酰亚胺处理的蛋白质稳定性。通过测量MLC2磷酸化和肌动蛋白应力纤维形成来监测Rho/ROCK活性。使用细胞塌陷测定法测量神经丛蛋白B1功能。
    结果:我们在此显示,单个临床相关的P1597L氨基酸变化将PlexinB1从Ras的阻遏物转化为Ras激活物。PlexinB1(P1597L)突变抑制PlexinB1的RapGAP活性,促进Ras活性的显著增加。P1597L突变还阻断了PlexinB1介导的Rho/ROCK活性降低,抑制野生型PlexinB1过表达诱导的MLC2磷酸化和肌动蛋白应激纤维形成的减少。PlexinB1(P1597L)对PlexinB1与小GTP酶或受体酪氨酸激酶的相互作用几乎没有影响,并且不抑制PlexinB1刺激的Akt或ERK磷酸化。这些结果表明突变通过Rap/Ras途径影响Rho信号传导。PlexinB1(P1597L)突变抑制野生型PlexinB1表达诱导的形态细胞塌陷,表明突变诱导抑制性肿瘤抑制功能的丧失。
    结论:这些结果表明,在前列腺癌小鼠模型中,PlexinB1中的临床相关P1597L突变可能通过降低PlexinB1的RapGAP活性,将PlexinB1从抑制基因转变为转移驱动基因,导致Ras激活。这些发现强调了PlexinB1-Rap-Ras途径在前列腺癌中的治疗性干预。
    BACKGROUND: Metastatic prostate cancer is a leading cause of cancer-related morbidity and mortality in men, yet the underlying molecular mechanisms are poorly understood. Plexins are transmembrane receptors for semaphorins with divergent roles in many forms of cancer. We recently found that a single clinically relevant specific amino acid change (Proline1597Leucine, (P1597L)), found in metastatic deposits of prostate cancer patients, converts PlexinB1 from a metastasis suppressor to a gene that drives prostate cancer metastasis in vivo. However, the mechanism by which PlexinB1(P1597L) promotes metastasis is not known.
    METHODS: Pull down assays using GST-RalGDS or -GSTRaf1-RBD were used to reveal the effect of mutant or wild-type PlexinB1 expression on Rap and Ras activity respectively. Protein-protein interactions were assessed in GST pulldown assays, Akt/ERK phosphorylation by immunoblotting and protein stability by treatment with cycloheximide. Rho/ROCK activity was monitored by measuring MLC2 phosphorylation and actin stress fiber formation. PlexinB1 function was measured using cell-collapse assays.
    RESULTS: We show here that the single clinically relevant P1597L amino acid change converts PlexinB1 from a repressor of Ras to a Ras activator. The PlexinB1(P1597L) mutation inhibits the RapGAP activity of PlexinB1, promoting a significant increase in Ras activity. The P1597L mutation also blocks PlexinB1-mediated reduction in Rho/ROCK activity, restraining the decrease in MLC2 phosphorylation and actin stress fiber formation induced by overexpression of wild-type PlexinB1. PlexinB1(P1597L) has little effect on the interaction of PlexinB1 with small GTPases or receptor tyrosine kinases and does not inhibit PlexinB1-stimulated Akt or ERK phosphorylation. These results indicate that the mutation affects Rho signalling via the Rap/Ras pathway. The PlexinB1(P1597L) mutation inhibits morphological cell collapse induced by wild-type PlexinB1 expression, suggesting that the mutation induces a loss of an inhibitory tumour suppressor function.
    CONCLUSIONS: These results suggest that the clinically relevant P1597L mutation in PlexinB1 may transform PlexinB1 from a suppressor to a driver of metastasis in mouse models of prostate cancer by reducing the RapGAP activity of PlexinB1, leading to Ras activation. These findings highlight the PlexinB1-Rap-Ras pathway for therapeutic intervention in prostate cancer.
    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
    Rho/Racofplant(ROP)GTPases是一种植物特异性蛋白质,可作为分子开关,由鸟嘌呤核苷酸交换因子(GEF)激活,并由GTP酶激活蛋白(GAP)灭活。苔藓植物Marchantiapolymorpha包含ROP(MpROP)的单拷贝,GEF(ROPGEF和SPIKE(SPK)),和差距(ROPGAP和ROP增强器(REN))。MpROP调节各种组织和器官的发育,如根状体,Gemmae,和空气室。而ROPGEF,KARAPPO(MpKAR)对于宝石启动至关重要,对其他ROP调节因子的功能了解较少。本研究集中于两个GAP:MpROPGAP和MpREN。Mpren单突变体在叶状体生长中显示出缺陷,根状茎尖生长,gemma发展,和气室的形成,而Mpropgap突变体没有显示出可见的异常。然而,MpropgapMpren双突变体比Mpren单突变体具有更严重的表型,建议MpROPGAP在涉及MpREN的进程中的备份角色。MpROPGAP的过表达,MpREN导致类似的配子体缺陷,强调MpROP激活/失活循环(或平衡)的重要性。因此,MpREN主要是,和MpROPGAP作为备份,调节配子体发育,最有可能通过控制多形性分枝杆菌中的MpROP激活。
    Rho/Rac of plant (ROP) GTPases are plant-specific proteins that function as molecular switches, activated by guanine nucleotide exchange factors (GEFs) and inactivated by GTPase-activating proteins (GAPs). The bryophyte Marchantia polymorpha contains single copies of ROP (MpROP), GEFs [ROPGEF and SPIKE (SPK)] and GAPs [ROPGAP and ROP ENHANCER (REN)]. MpROP regulates the development of various tissues and organs, such as rhizoids, gemmae and air chambers. The ROPGEF KARAPPO (MpKAR) is essential for gemma initiation, but the functions of other ROP regulatory factors are less understood. This study focused on two GAPs: MpROPGAP and MpREN. Mpren single mutants showed defects in thallus growth, rhizoid tip growth, gemma development, and air-chamber formation, whereas Mpropgap mutants showed no visible abnormalities. However, Mpropgap Mpren double mutants had more severe phenotypes than the Mpren single mutants, suggesting backup roles of MpROPGAP in processes involving MpREN. Overexpression of MpROPGAP and MpREN resulted in similar gametophyte defects, highlighting the importance of MpROP activation/inactivation cycling (or balancing). Thus, MpREN predominantly, and MpROPGAP as a backup, regulate gametophyte development, likely by controlling MpROP activation in M. polymorpha.
    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:胃癌(GC)是世界范围内常见的癌症;然而,其分子机制和致病机制尚不清楚。microRNAs(miRNAs),靶向GC中的关键基因,与肿瘤的促进或抑制有关。因此,识别新的miRNA机制可以改善GC患者的新诊断和治疗策略。
    方法:探讨miR-135b-5p在GC中的生物学功能,生物信息学分析和体外功能测定,包括菌落形成,伤口愈合,Transwell,和EdU化验,用于评估增殖,侵入性,和GC细胞的迁移能力。使用RNA-seq和在线数据库预测靶基因。双荧光素酶报告基因测定,荧光原位杂交和免疫印迹用于确认miR-135b-5p和CLIP4之间的调控关系。使用临床样品以及体外和体内测定来评估CLIP4在肿瘤进展中的作用。使用挽救试验阐明了CLIP4在GC中的肿瘤抑制机制。
    结果:我们的研究确定miR-135b-5p是GC组织中超表达的三大miRNA之一,RT-qPCR证实其上调。功能分析显示上调的miR-135b-5p促进GC细胞的恶性表型。机制研究表明miR-135b-5p通过靶向CLIP4作为癌症启动子。此外,我们的研究提示CLIP4通过抑制JAK2/STAT3信号通路发挥肿瘤抑制功能.
    结论:本研究揭示了miR-135b-5p通过靶向CLIP4发挥其肿瘤促进功能的新机制。还阐明了CLIP4通过灭活JAK2/STAT3途径的肿瘤抑制功能。miR-135b-5p对CLIP4的调控机制为GC患者提供了一种有前景的新型治疗策略。
    BACKGROUND: Gastric cancer (GC) is a common cancer worldwide; however, its molecular and pathogenic mechanisms remain unclear. MicroRNAs (miRNAs), which target key genes in GC, are associated with tumor promotion or suppression. Therefore, identifying new miRNA mechanisms could improve the novel diagnostic and therapeutic strategies for patients with GC.
    METHODS: To explore the biological functions of miR-135b-5p in GC, bioinformatic analysis and in vitro functional assays, including colony formation, wound healing, Transwell, and EdU assays, were used to assess the proliferative, invasive, and migratory capacities of GC cells. Target genes were predicted using RNA-seq and online databases. Dual-luciferase reporter assay, fluorescence in situ hybridization and western blotting were used to confirm the regulatory relationship between miR-135b-5p and CLIP4. The role of CLIP4 in tumor progression was assessed using clinical samples and both in vitro and in vivo assays. The tumor-suppressive mechanism of CLIP4 in GC was elucidated using rescue assays.
    RESULTS: Our study identified that miR-135b-5p as one of the top three over-expressed miRNAs in GC tissues, with RT-qPCR confirming its upregulation. Functional analysis showed that upregulated miR-135b-5p promoted malignant phenotypes in GC cells. Mechanistic research indicated that miR-135b-5p acts as a cancer promoter by targeting CLIP4. Moreover, our study suggested that CLIP4 exerts its tumor-suppressive function by inhibiting the JAK2/STAT3 signaling pathway.
    CONCLUSIONS: This study reveals a novel mechanism by which miR-135b-5p exerts its tumor-promoting functions by targeting CLIP4. The tumor-suppressive function of CLIP4 by inactivating the JAK2/STAT3 pathway is also elucidated. Regulatory mechanism of CLIP4 by miR-135b-5p provides a promising novel therapeutic strategy for GC 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
    GNAO1编码G蛋白亚基αO1(Gαo)。GNAO1的致病变异导致发育迟缓,顽固性癫痫发作,和从婴儿早期开始的渐进性不自主运动。由于GNAO1在发育中的大脑中的功能作用尚不清楚,对于出现GNAO1相关脑病的患者,治疗策略仍未确立.我们在此报道siRNA介导的Gnao1耗竭扰乱了与Neuro2a细胞中RhoGTP酶信号相关的转录本的表达。始终如一,siRNA处理阻碍了神经突生长和延伸。从具有Gαo致病性变体的患者的iPSC分化的单层神经元中,生长锥的形成被明显破坏(p。G203R)。这种变体使神经球形组装失效,获得有组织的结构,来自患者iPSCs的皮质类器官中磷酸化MLC2的极化信号。我们证实Rho激酶抑制剂Y27632恢复了这些形态表型。因此,Gαo通过调节Rho相关途径来决定发育中大脑的自组织过程。这些数据表明,RhoGTP酶途径可能是GNAO1相关性脑病患者治疗的替代靶标。
    GNAO1 encodes G protein subunit alpha O1 (Gαo). Pathogenic variations in GNAO1 cause developmental delay, intractable seizures, and progressive involuntary movements from early infancy. Because the functional role of GNAO1 in the developing brain remains unclear, therapeutic strategies are still unestablished for patients presenting with GNAO1-associated encephalopathy. We herein report that siRNA-mediated depletion of Gnao1 perturbs the expression of transcripts associated with Rho GTPase signaling in Neuro2a cells. Consistently, siRNA treatment hampered neurite outgrowth and extension. Growth cone formation was markedly disrupted in monolayer neurons differentiated from iPSCs from a patient with a pathogenic variant of Gαo (p.G203R). This variant disabled neuro-spherical assembly, acquisition of the organized structure, and polarized signals of phospho-MLC2 in cortical organoids from the patient\'s iPSCs. We confirmed that the Rho kinase inhibitor Y27632 restored these morphological phenotypes. Thus, Gαo determines the self-organizing process of the developing brain by regulating the Rho-associated pathway. These data suggest that Rho GTPase pathway might be an alternative target of therapy for patients with GNAO1-associated encephalopathy.
    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
    神经干细胞(NSC)在静止状态和增殖状态之间的转变是大脑发育和稳态的基础。NSC再激活的缺陷与神经发育障碍有关。果蝇静止的NSC向神经纤维延伸富含肌动蛋白的初级突起。然而,肌动蛋白细胞骨架在NSC再激活过程中的功能尚不清楚。这里,我们通过扩展和超分辨率显微镜揭示了静止神经干细胞突起中的细丝状肌动蛋白(F-actin)结构。我们表明,F-肌动蛋白聚合促进myocardin相关转录因子的核易位,一种小头畸形相关的转录因子,用于NSC再激活和大脑发育。F-肌动蛋白聚合受由G蛋白偶联受体Smog组成的信号级联调节,G蛋白αq亚基,Rho1鸟苷三磷酸酶,以及NSC重新激活期间的透明(Dia)/Formin。Further,星形胶质细胞分泌Smog配体折叠的胃泌以调节Gαq-Rho1-Dia介导的NSC再激活。一起,我们确定Smog-Gαq-Rho1信号轴来自星形胶质细胞,NSC利基市场,调节NSC再激活中Dia介导的F-肌动蛋白动力学。
    The transitioning of neural stem cells (NSCs) between quiescent and proliferative states is fundamental for brain development and homeostasis. Defects in NSC reactivation are associated with neurodevelopmental disorders. Drosophila quiescent NSCs extend an actin-rich primary protrusion toward the neuropil. However, the function of the actin cytoskeleton during NSC reactivation is unknown. Here, we reveal the fine filamentous actin (F-actin) structures in the protrusions of quiescent NSCs by expansion and super-resolution microscopy. We show that F-actin polymerization promotes the nuclear translocation of myocardin-related transcription factor, a microcephaly-associated transcription factor, for NSC reactivation and brain development. F-actin polymerization is regulated by a signaling cascade composed of G protein-coupled receptor Smog, G protein αq subunit, Rho1 guanosine triphosphatase, and Diaphanous (Dia)/Formin during NSC reactivation. Further, astrocytes secrete a Smog ligand folded gastrulation to regulate Gαq-Rho1-Dia-mediated NSC reactivation. Together, we establish that the Smog-Gαq-Rho1 signaling axis derived from astrocytes, an NSC niche, regulates Dia-mediated F-actin dynamics in NSC reactivation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号