c-Src

c - Src
  • 文章类型: Journal Article
    血管壁内的内皮细胞与周围的细胞外基质错综复杂地沟通,将机械线索转化为生化信号。此外,血管需要酶降解周围基质的能力,以促进血管扩张。c-Src在血管生长中起关键作用,其在内皮中的损失减少了血管发芽和局灶性粘附信号。这里,我们显示内皮细胞中c-Src的组成型激活导致血管快速扩张,独立于生长因子刺激或流体剪切应力。这是由局灶性粘附信号和大小的增加驱动的,增强了负责细胞外基质重塑的基质金属蛋白酶的局部分泌。基质金属蛋白酶活性的抑制导致由提高的c-Src活性引起的血管扩张的有力挽救。这支持了一个前提,即缓和局灶性粘连相关事件和基质降解可以抵消异常血管扩张,与异常血管形态驱动的病理有关。
    Endothelial cells lining the blood vessel wall communicate intricately with the surrounding extracellular matrix, translating mechanical cues into biochemical signals. Moreover, vessels require the capability to enzymatically degrade the matrix surrounding them, to facilitate vascular expansion. c-Src plays a key role in blood vessel growth, with its loss in the endothelium reducing vessel sprouting and focal adhesion signalling. Here, we show that constitutive activation of c-Src in endothelial cells results in rapid vascular expansion, operating independently of growth factor stimulation or fluid shear stress forces. This is driven by an increase in focal adhesion signalling and size, with enhancement of localised secretion of matrix metalloproteinases responsible for extracellular matrix remodelling. Inhibition of matrix metalloproteinase activity results in a robust rescue of the vascular expansion elicited by heightened c-Src activity. This supports the premise that moderating focal adhesion-related events and matrix degradation can counteract abnormal vascular expansion, with implications for pathologies driven by unusual vascular morphologies.
    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
    肿瘤坏死因子α(TNFα,TNF)是一种多效性细胞因子,通过1型TNF受体(TNFR1)发挥其大部分作用。TNF结合后,TNFR1招募TRADD(肿瘤坏死因子受体1型相关死亡结构域)。这种相互作用引发信号体复合物的形成,这些复合物被声称诱导细胞凋亡(通过下游胱天蛋白酶激活),炎症(通过NF-κB)和应激途径(JNK&p38)。然而,TNF诱导的ERK和AKT激活的潜在机制尚未完全揭示.已知TNFR1组成型结合c-Src和JAK2,并且这些酶先前被证明调节TNF信号传导。因此,我们假设TNFR1可能被JAK2和/或c-Src酪氨酸磷酸化,而TNF诱导的ERK和Akt激活可能由这种磷酸化介导.
    进行定点诱变(SDM),以丙氨酸(A)或天冬氨酸(D)取代TNFR1上的两个推定酪氨酸磷酸化位点(Y360和Y401),抑制或模拟组成型磷酸化,分别。在用突变或野生型TNFR1转染的293T细胞中,通过蛋白质印迹测定ERK和Akt激活。TNFR1与c-Src相互作用,通过co-IP检查JAK2、p85和Grb2。NF-kB激活通过荧光素酶测定法测量,而增殖通过MTT测量,凋亡通过比色caspase8/3测定法评估。为了确定坏死率,进行细胞DNA片段化ELISA。
    在本报告中,我们显示TNFR1在Y401被JAK2酪氨酸激酶磷酸化,在Y360和Y401被c-Src磷酸化。Y360和Y401的磷酸化增强了Grb2和PI3Kp85与TNFR1的相互作用。我们还证明Y360D和Y401D的磷模拟突变增强ERK和Akt活化。
    TNFR1是由c-Src和JAK2磷酸化的酪氨酸,触发“非规范”途径,激活ERK和Akt.
    UNASSIGNED: Tumor necrosis factor alpha (TNFα, a.k.a. TNF) is a pleiotropic cytokine that exerts most of its effects through type 1 TNF receptor (TNFR1). Following TNF binding, TNFR1 recruits TRADD (tumor necrosis factor receptor type 1-associated DEATH domain). This interaction triggers formation of signalosome complexes which have been claimed to induce apoptosis (via downstream caspase activations), inflammation (via NF-kappaB) and stress pathways (JNK & p38). However, the mechanism underlying TNF-induced ERK and AKT activation is not completely revealed. TNFR1 is known to constitutively bind c-Src and JAK2, and these enzymes were previously demonstrated to modulate TNF signaling. Therefore, we hypothesized that TNFR1 could be tyrosine phosphorylated by JAK2 and/or c-Src and TNF-induced ERK and Akt activation may be mediated by this phosphorylation.
    UNASSIGNED: Site-directed mutagenesis (SDM) was performed to substitute the two putative Tyrosine phosphorylation sites on TNFR1 (Y360 and Y401) with alanine (A) or with aspartic acid (D), to inhibit or mimic constitutive phosphorylation, respectively. In 293T cells transfected with mutated or wild type TNFR1, ERK and Akt activations were determined by western blot. TNFR1 interaction with c-Src, JAK2, p85 and Grb2 was examined by co-IP. NF-kB activation was measured by luciferase assay, while proliferation was measured by MTT and apoptosis was evaluated by colorimetric caspase 8/3 assays. For determination of necrosis rates, cellular DNA fragmentation ELISA was performed.
    UNASSIGNED: In this report, we show that TNFR1 is phosphorylated by JAK2 tyrosine kinase at Y401 and by c-Src at Y360 and Y401. Phosphorylation of Y360 and Y401 augments the interaction of Grb2 and PI3Kp85 with TNFR1. We also demonstrate that phosphomimetic mutations of Y360D and Y401D enhance ERK and Akt activation.
    UNASSIGNED: TNFR1 is tyrosine phosphorylated by both c-Src and JAK2, triggering a \"noncanonical\" pathway, that activates ERK and Akt.
    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
    蛋白酪氨酸激酶(RTK)调节多种非恶性疾病的病理生理事件,包括糖尿病并发症.寻找驱动糖尿病心肌病(DCM)发展的新靶点,我们对糖尿病小鼠心脏中的RTKs磷酸化阵列进行了分析,并确定了心肌细胞中磷酸化成纤维细胞生长因子受体1(p-FGFR1)水平增加,提示FGFR1可能参与了DCM的发病机制。使用原代心肌细胞和H9C2细胞系,我们发现高浓度葡萄糖(HG)通过toll样受体4(TLR4)和c-Src反式激活FGFR1激酶结构域,独立于FGF配体。敲低TLR4或c-Src的水平可防止心肌细胞中HG激活的FGFR1。RNA测序分析表明,在HG攻击的心肌细胞中,升高的FGFR1活性通过MAPKs-NFκB信号通路诱导促炎反应,这进一步导致纤维化和肥大。然后,我们产生了心肌细胞特异性FGFR1敲除小鼠,并表明心肌细胞中缺乏FGFR1可以预防糖尿病诱导的心脏炎症并保留小鼠的心脏功能。选择性抑制剂对FGFR1的药理学抑制作用,AZD4547,还可以预防心脏炎症,纤维化,1型和2型糖尿病小鼠的功能障碍。这些研究已经确定FGFR1是驱动DCM的新参与者,并支持进一步测试FGFR1抑制剂可能的心脏保护益处。
    Protein tyrosine kinases (RTKs) modulate a wide range of pathophysiological events in several non-malignant disorders, including diabetic complications. To find new targets driving the development of diabetic cardiomyopathy (DCM), we profiled an RTKs phosphorylation array in diabetic mouse hearts and identified increased phosphorylated fibroblast growth factor receptor 1 (p-FGFR1) levels in cardiomyocytes, indicating that FGFR1 may contribute to the pathogenesis of DCM. Using primary cardiomyocytes and H9C2 cell lines, we discovered that high-concentration glucose (HG) transactivates FGFR1 kinase domain through toll-like receptor 4 (TLR4) and c-Src, independent of FGF ligands. Knocking down the levels of either TLR4 or c-Src prevents HG-activated FGFR1 in cardiomyocytes. RNA-sequencing analysis indicates that the elevated FGFR1 activity induces pro-inflammatory responses via MAPKs-NFκB signaling pathway in HG-challenged cardiomyocytes, which further results in fibrosis and hypertrophy. We then generated cardiomyocyte-specific FGFR1 knockout mice and showed that a lack of FGFR1 in cardiomyocytes prevents diabetes-induced cardiac inflammation and preserves cardiac function in mice. Pharmacological inhibition of FGFR1 by a selective inhibitor, AZD4547, also prevents cardiac inflammation, fibrosis, and dysfunction in both type 1 and type 2 diabetic mice. These studies have identified FGFR1 as a new player in driving DCM and support further testing of FGFR1 inhibitors for possible cardioprotective benefits.
    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
    正常细胞向癌细胞的异常转化,被称为致癌作用,是一个复杂的过程,涉及许多遗传和分子改变,以响应先天和环境刺激。Src家族激酶(SFK)是与癌发生有关的信号通路的关键组成部分,c-Src及其致癌对应物v-Src通常起着重要作用。c-Src的发现代表了令人信服的叙述,突出了开创性的发现和对致癌作用的分子机制的宝贵见解。在致癌激活后,c-Src激活多个下游信号通路,包括PI3K-AKT通路,Ras-MAPK通路,JAK-STAT3通路,和FAK/Paxillin途径,这对细胞增殖很重要,生存,迁移,入侵,转移,和抗药性。在这次审查中,我们深入研究了c-Src和v-Src的发现,c-Src的结构,以及激活c-Src的分子机制。我们还关注c-Src用于促进肿瘤发生和对化疗药物以及分子靶向药物的耐药性的各种信号通路。
    The aberrant transformation of normal cells into cancer cells, known as carcinogenesis, is a complex process involving numerous genetic and molecular alterations in response to innate and environmental stimuli. The Src family kinases (SFK) are key components of signaling pathways implicated in carcinogenesis, with c-Src and its oncogenic counterpart v-Src often playing a significant role. The discovery of c-Src represents a compelling narrative highlighting groundbreaking discoveries and valuable insights into the molecular mechanisms underlying carcinogenesis. Upon oncogenic activation, c-Src activates multiple downstream signaling pathways, including the PI3K-AKT pathway, the Ras-MAPK pathway, the JAK-STAT3 pathway, and the FAK/Paxillin pathway, which are important for cell proliferation, survival, migration, invasion, metastasis, and drug resistance. In this review, we delve into the discovery of c-Src and v-Src, the structure of c-Src, and the molecular mechanisms that activate c-Src. We also focus on the various signaling pathways that c-Src employs to promote oncogenesis and resistance to chemotherapy drugs as well as molecularly targeted agents.
    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
    c-Met has been an attractive target of prognostic and therapeutic studies in various cancers. TPX-0022 is a macrocyclic inhibitor of c-Met, c-Src and CSF1R kinases and is currently in phase I/II clinical trials in patients with advanced solid tumors harboring MET gene alterations. In this study, we determined the co-crystal structures of the c-Met/TPX-0022 and c-Src/TPX-0022 complexes to help elucidate the binding mechanism. TPX-0022 binds to the ATP pocket of c-Met and c-Src in a local minimum energy conformation and is stabilized by hydrophobic and hydrogen bond interactions. In addition, TPX-0022 exhibited potent activity against the resistance-relevant c-Met L1195F mutant and moderate activity against the c-Met G1163R, F1200I and Y1230H mutants but weak activity against the c-Met D1228N and Y1230C mutants. Overall, our study reveals the structural mechanism underlying the potency and selectivity of TPX-0022 and the ability to overcome acquire resistance mutations and provides insight into the development of selective c-Met macrocyclic inhibitors.
    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
    Lenvatinib,一种多激酶抑制剂,在不可切除的肝细胞癌(HCC)的治疗中起着至关重要的作用。然而,>50%接受乐伐替尼治疗的患者在1年内经历肿瘤生长或转移,强调需要解决获得性耐药作为一个关键的临床挑战。为了阐明与来伐替尼获得性耐药相关的因素,通过暴露对lenvatinib敏感的HCC细胞系建立了对lenvatinib耐药的HCC细胞系(JHH-7_LR),JHH-7,来伐替尼。使用蛋白质组学方法分析了与抗性发展相关的蛋白质表达的变化,检测1,321种蛋白和267种蛋白表达的显著变化。使用创造性途径分析生物信息学软件,研究表明,多个信号通路的活性随着这些蛋白质表达的变化而变化,c-SRC被鉴定为参与许多这些信号通路的蛋白质,其活性随着抗性的获得而显著变化。当共同使用达沙替尼时,c-SRC抑制剂,在JHH-7_LR细胞系中观察到lenvatinib敏感性的部分恢复。本研究表明,c-SRC表达增加与肝癌对乐伐替尼的耐药性部分相关,表明c-SRC抑制可以降低肝癌对乐伐替尼的耐药性。
    Lenvatinib, a multi-kinase inhibitor, serves a crucial role in the treatment of unresectable hepatocellular carcinoma (HCC). However, >50% of patients receiving lenvatinib therapy experience tumor growth or metastasis within 1 year, highlighting the need to address acquired resistance as a critical clinical challenge. To elucidate the factors associated with acquired resistance to lenvatinib, a lenvatinib-resistant HCC cell line (JHH-7_LR) was established by exposing a lenvatinib-sensitive HCC cell line, JHH-7, to lenvatinib. The changes in protein expression associated with the development of resistance were analyzed using a proteomic approach, detecting 1,321 proteins and significant changes in the expression of 267 proteins. Using Ingenuity Pathway Analysis bioinformatics software, it was revealed that the activity of multiple signaling pathways varied alongside the changes in expression of these proteins, and c-SRC was identified as a protein involved in a number of these signaling pathways, with its activity varying markedly upon the acquisition of resistance. When co-administering dasatinib, a c-SRC inhibitor, the partial restoration of lenvatinib sensitivity in the JHH-7_LR cell line was observed. The present study demonstrated that increased c-SRC expression was partially associated with HCC resistance to lenvatinib, suggesting that c-SRC inhibition could reduce the resistance of HCC to lenvatinib.
    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
    肿瘤细胞通常过度表达免疫检查点蛋白,包括CD47,用于免疫逃避。然而,是否或如何致癌激活受体酪氨酸激酶,它们是肿瘤发展的关键驱动因素,调节CD47表达是未知的。这里,已证明表皮生长因子受体(EGFR)激活通过增加c-Src与CD47的结合来诱导CD47表达,从而导致c-Src介导的CD47Y288磷酸化。这种磷酸化抑制了泛素E3连接酶TRIM21和CD47之间的相互作用,从而消除了TRIM21介导的CD47K99/102聚泛素化和CD47降解。CD47Y288F的敲入表达降低了CD47的表达,增加肿瘤细胞的巨噬细胞吞噬,并抑制小鼠脑肿瘤的生长。相比之下,与CD47Y288F表达相比,CD47K99/102R的敲入表达引起相反的作用。重要的是,使用抗CD47抗体治疗的CD47-SIRPα阻断显着增强EGFR靶向癌症治疗。此外,人胶质母细胞瘤(GBM)标本中的CD47表达水平与EGFR和c-Src激活以及人GBM的加重相关。这些发现阐明了EGFR激活的肿瘤细胞中CD47上调的新机制,并强调了EGFR-c-Src-TRIM21-CD47信号轴在肿瘤逃避中的作用,以及结合CD47阻断和EGFR靶向治疗改善当前癌症治疗的潜力。
    Tumor cells often overexpress immune checkpoint proteins, including CD47, for immune evasion. However, whether or how oncogenic activation of receptor tyrosine kinases, which are crucial drivers in tumor development, regulates CD47 expression is unknown. Here, it is demonstrated that epidermal growth factor receptor (EGFR) activation induces CD47 expression by increasing the binding of c-Src to CD47, leading to c-Src-mediated CD47 Y288 phosphorylation. This phosphorylation inhibits the interaction between the ubiquitin E3 ligase TRIM21 and CD47, thereby abrogating TRIM21-mediated CD47 K99/102 polyubiquitylation and CD47 degradation. Knock-in expression of CD47 Y288F reduces CD47 expression, increases macrophage phagocytosis of tumor cells, and inhibits brain tumor growth in mice. In contrast, knock-in expression of CD47 K99/102R elicits the opposite effects compared to CD47 Y288F expression. Importantly, CD47-SIRPα blockade with an anti-CD47 antibody treatment significantly enhances EGFR-targeted cancer therapy. In addition, CD47 expression levels in human glioblastoma (GBM) specimens correlate with EGFR and c-Src activation and aggravation of human GBM. These findings elucidate a novel mechanism underlying CD47 upregulation in EGFR-activated tumor cells and underscore the role of the EGFR-c-Src-TRIM21-CD47 signaling axis in tumor evasion and the potential to improve the current cancer therapy with a combination of CD47 blockade with EGFR-targeted remedy.
    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
    为了启动抗利什曼酶适应性免疫反应,树突状细胞(DC)必须携带利什曼原虫抗原从外周组织到局部引流淋巴结。然而,DC的迁移能力在利什曼原虫donovani感染期间受到很大损害。这种有缺陷的DC迁移的分子机制尚未完全了解。这里,我们证明多诺瓦尼乳杆菌感染通过降低C型凝集素受体2(CLEC-2)的表达而损害了DCs的淋巴结归巢能力。多诺瓦尼乳杆菌通过诱导DC分泌转化生长因子-β(TGF-β)来发挥这种抑制作用。的确,以这种方式产生的TGF-β通过激活c-Src抑制核因子-κB(NF-κB)介导的CLEC-2在DC上的表达。值得注意的是,c-Src表达的抑制通过阻止多诺瓦尼乳杆菌诱导的CLEC-2对DCs的下调而显着改善了DCs在引流淋巴结中的到达。这些发现揭示了一种独特的机制,其中L.donovani抑制DC迁移到淋巴结,并提示TGF-β的关键作用,c-Src,和CLEC-2调节这个过程。重要性树突状细胞(DC)在启动T细胞介导的针对内脏利什曼病(VL)的保护性免疫中起关键作用,世界上第二致命的寄生虫病。然而,DC的T细胞诱导能力关键取决于DC向局部淋巴结迁移的程度。值得注意的是,据报道,在VL期间DC的迁移受损。这种受损的DC迁移的原因,然而,仍然不明确。这里,我们提供了第一个证据L.Donovani,VL的病原体,通过降低DC上C型凝集素受体2(CLEC-2)的表达来减弱DC的淋巴结归巢能力。此外,我们已经证明了L.donovani如何介导这种抑制作用。总的来说,我们的工作揭示了多诺瓦尼乳杆菌诱导的DC迁移受损的独特机制,并提出了一种通过增加DC到达淋巴结来改善抗白质T细胞活性的潜在策略.
    To initiate an antileishmanial adaptive immune response, dendritic cells (DCs) must carry Leishmania antigens from peripheral tissues to local draining lymph nodes. However, the migratory capacity of DCs is largely compromised during Leishmania donovani infection. The molecular mechanism underlying this defective DC migration is not yet fully understood. Here, we demonstrate that L. donovani infection impaired the lymph node homing ability of DCs by decreasing C-type lectin receptor 2 (CLEC-2) expression. L. donovani exerted this inhibitory effect by inducing transforming growth factor-β (TGF-β) secretion from DCs. Indeed, TGF-β produced in this manner inhibited nuclear factor-κB (NF-κB)-mediated CLEC-2 expression on DCs by activating c-Src. Notably, suppression of c-Src expression significantly improved the arrival of DCs in draining lymph nodes by preventing L. donovani-induced CLEC-2 downregulation on DCs. These findings reveal a unique mechanism by which L. donovani inhibits DC migration to lymph nodes and suggest a key role for TGF-β, c-Src, and CLEC-2 in regulating this process. IMPORTANCE Dendritic cells (DCs) play a key role in initiating T cell-mediated protective immunity against visceral leishmaniasis (VL), the second most lethal parasitic disease in the world. However, the T cell-inducing ability of DCs critically depends on the extent of DC migration to regional lymph nodes. Notably, the migration of DCs is reported to be impaired during VL. The cause of this impaired DC migration, however, remains ill-defined. Here, we provide the first evidence that L. donovani, the causative agent of VL, attenuates the lymph node homing capacity of DCs by decreasing C-type lectin receptor 2 (CLEC-2) expression on DCs. Additionally, we have demonstrated how L. donovani mediates this inhibitory effect. Overall, our work has revealed a unique mechanism underlying L. donovani-induced impairment of DC migration and suggests a potential strategy to improve antileishmanial T cell activity by increasing DC arrival in lymph nodes.
    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
    核到细胞质的错误定位和多个RNA结合蛋白(RBPs)的聚集,包括FUS,是大多数肌萎缩侧索硬化症(ALS)和额颞叶变性(FTLD)病例的主要神经病理学特征。在ALS-FUS中,这些聚集体来自FUS的疾病相关突变,而在FTLD-FUS中,细胞质内含物不含突变型FUS,提示FTLD中FUS发病机制的不同分子机制尚待研究。我们先前已经表明,由于与核输入受体TNPO1的结合受损,FUS的C末端Tyr526的磷酸化导致FUS的细胞质保留增加。受到上述概念的启发,在当前的研究中,我们开发了一种针对C端磷酸化Tyr526FUS(FUSp-Y526)的新型抗体,该抗体能够特异性识别磷酸化的细胞质FUS,这是由其他市售FUS抗体很难识别。使用FUSp-Y526抗体,我们证明了FUS磷酸化对各种细胞中可溶性和不溶性FUSp-Y526的细胞质分布的特异性影响,并证实了Src激酶家族参与Tyr526FUS磷酸化.此外,我们发现FUSp-Y526表达模式与小鼠特定脑区的活性pSrc/pAbl激酶相关,表明cAbl优先参与皮质神经元中FUSp-Y526的细胞质错位。最后,活性cAbl激酶和FUSp-Y526的免疫反应性模式显示,与对照组相比,FUSp-Y526在FTLD患者死后额叶皮质组织的皮质神经元中的细胞质分布发生了改变.FUSp-Y526和FUS信号的重叠优先出现在小的弥散性包裹体中,而在成熟的聚集体中不存在。提示FUSp-Y526可能参与细胞质中早期毒性FUS聚集体的形成,这些聚集体大部分未被市售FUS抗体检测到。鉴于cAbl活性和FUSp-Y526分布在皮质神经元中的重叠模式,和cAbl诱导FUSp-Y526在应激细胞中螯合到G3BP1阳性颗粒中,我们认为cAbl激酶积极参与介导FTLD患者大脑中野生型FUS的细胞质错位和毒性聚集,作为FTLD-FUS病理生理和进展的一种新的推定潜在机制。
    Nuclear to cytoplasmic mislocalization and aggregation of multiple RNA-binding proteins (RBPs), including FUS, are the main neuropathological features of the majority of cases of amyotrophic lateral sclerosis (ALS) and frontotemporal lobular degeneration (FTLD). In ALS-FUS, these aggregates arise from disease-associated mutations in FUS, whereas in FTLD-FUS, the cytoplasmic inclusions do not contain mutant FUS, suggesting different molecular mechanisms of FUS pathogenesis in FTLD that remain to be investigated. We have previously shown that phosphorylation of the C-terminal Tyr526 of FUS results in increased cytoplasmic retention of FUS due to impaired binding to the nuclear import receptor TNPO1. Inspired by the above notions, in the current study we developed a novel antibody against the C-terminally phosphorylated Tyr526 FUS (FUSp-Y526) that is specifically capable of recognizing phosphorylated cytoplasmic FUS, which is poorly recognized by other commercially available FUS antibodies. Using this FUSp-Y526 antibody, we demonstrated a FUS phosphorylation-specific effect on the cytoplasmic distribution of soluble and insoluble FUSp-Y526 in various cells and confirmed the involvement of the Src kinase family in Tyr526 FUS phosphorylation. In addition, we found that FUSp-Y526 expression pattern correlates with active pSrc/pAbl kinases in specific brain regions of mice, indicating preferential involvement of cAbl in the cytoplasmic mislocalization of FUSp-Y526 in cortical neurons. Finally, the pattern of immunoreactivity of active cAbl kinase and FUSp-Y526 revealed altered cytoplasmic distribution of FUSp-Y526 in cortical neurons of post-mortem frontal cortex tissue from FTLD patients compared with controls. The overlap of FUSp-Y526 and FUS signals was found preferentially in small diffuse inclusions and was absent in mature aggregates, suggesting possible involvement of FUSp-Y526 in the formation of early toxic FUS aggregates in the cytoplasm that are largely undetected by commercially available FUS antibodies. Given the overlapping patterns of cAbl activity and FUSp-Y526 distribution in cortical neurons, and cAbl induced sequestration of FUSp-Y526 into G3BP1 positive granules in stressed cells, we propose that cAbl kinase is actively involved in mediating cytoplasmic mislocalization and promoting toxic aggregation of wild-type FUS in the brains of FTLD patients, as a novel putative underlying mechanism of FTLD-FUS pathophysiology and progression.
    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
    短链脂肪酸(SCFA)如丁酸(BUT)在很大程度上影响血管的完整性,并与心血管疾病的发生和发展密切相关。然而,它们对血管内皮钙粘蛋白(VEC)的影响,一种主要的血管粘附和信号分子,基本上是未知的。这里,我们探索了SCFABUT对VEC(Y731,Y685和Y658)的特定酪氨酸残基磷酸化的影响,据报道,这对VEC调节和血管完整性至关重要。此外,我们揭示了BUT参与影响VEC磷酸化的信号通路。因此,我们使用磷酸特异性抗体评估了人主动脉内皮细胞(HAOECs)中响应SCFA丁酸钠的VEC磷酸化,并进行了葡聚糖测定以分析EC单层的通透性.分别使用c-Src家族激酶和FFAR2/3的抑制剂和拮抗剂分析了c-Src和SCFA受体FFAR2和FFAR3在诱导VEC磷酸化中的作用,以及通过RNAi介导的敲低。通过荧光显微镜评估响应于BUT的VEC的定位。HAOEC的BUT处理导致Y731在VEC处的特异性磷酸化,而对Y685和Y658的影响较小。因此,BUT参与FFAR3、FFAR2和c-Src激酶诱导VEC磷酸化。VEC磷酸化与内皮通透性增强和交界性VEC的c-Src依赖性重塑相关。我们的数据表明,但是,SCFA和肠道微生物群衍生的代谢物,通过靶向VEC磷酸化影响血管完整性,对血管疾病的病理生理学和治疗有潜在影响。
    Short-chain fatty acids (SCFAs) like butyrate (BUT) largely influence vascular integrity and are closely associated with the onset and progression of cardiovascular diseases. However, their impact on vascular endothelial cadherin (VEC), a major vascular adhesion and signaling molecule, is largely unknown. Here, we explored the effect of the SCFA BUT on the phosphorylation of specific tyrosine residues of VEC (Y731, Y685, and Y658), which are reported to be critical for VEC regulation and vascular integrity. Moreover, we shed light on the signaling pathway engaged by BUT to affect the phosphorylation of VEC. Thereby, we used phospho-specific antibodies to evaluate the phosphorylation of VEC in response to the SCFA sodium butyrate in human aortic endothelial cells (HAOECs) and performed dextran assays to analyze the permeability of the EC monolayer. The role of c-Src and SCFA receptors FFAR2 and FFAR3 in the induction of VEC phosphorylation was analyzed using inhibitors and antagonists for c-Src family kinases and FFAR2/3, respectively, as well as by RNAi-mediated knockdown. Localization of VEC in response to BUT was assessed by fluorescence microscopy. BUT treatment of HAOEC resulted in the specific phosphorylation of Y731 at VEC with minor effects on Y685 and Y658. Thereby, BUT engages FFAR3, FFAR2, and c-Src kinase to induce phosphorylation of VEC. VEC phosphorylation correlated with enhanced endothelial permeability and c-Src-dependent remodeling of junctional VEC. Our data suggest that BUT, an SCFA and gut microbiota-derived metabolite, impacts vascular integrity by targeting VEC phosphorylation with potential impact on the pathophysiology and therapy of vascular diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号