endothelial hyperpermeability

  • 文章类型: Journal Article
    内皮高通透性是脓毒症相关多器官功能障碍的关键。增加血管性血友病因子(vWF)血浆水平,源于脓毒症期间活化的血小板和内皮损伤,可与整合素αvβ3结合,加剧内皮通透性。因此,靶向该途径为脓毒症提供了潜在的治疗途径.最近,我们鉴定了isaridinE(ISE),一种海洋真菌环己肽,作为一种有前途的抗血小板和抗血栓药物,出血风险低。ISE对脓毒症小鼠模型脓毒症死亡率和脓毒症肺损伤的影响,盲肠结扎和穿刺诱导,在这项研究中进行了调查。ISE剂量依赖性地提高生存率,减轻肺损伤,血小板减少症,肺内皮通透性,和小鼠模型中的血管炎症。ISE通过抑制囊泡相关膜蛋白8和可溶性N-乙基马来酰亚胺敏感因子附着蛋白23的过度表达,显着减少了脓毒症小鼠活化血小板中vWF的释放。此外,ISE抑制健康人血小板对培养的脂多糖(LPS)刺激的人脐静脉内皮细胞(HUVECs)的粘附,从而显著降低vWF分泌和内皮高通透性。使用cilengitide,选择性整合素αvβ3抑制剂,发现ISE可以通过抑制vWF与αvβ3的结合来改善内皮通透性。整合素αvβ3-FAK/Src途径的激活可能是vWF诱导的脓毒症内皮功能障碍的基础。总之,ISE通过抑制内皮高通透性和血小板-内皮相互作用来预防脓毒症。
    Endothelial hyperpermeability is pivotal in sepsis-associated multi-organ dysfunction. Increased von Willebrand factor (vWF) plasma levels, stemming from activated platelets and endothelium injury during sepsis, can bind to integrin αvβ3, exacerbating endothelial permeability. Hence, targeting this pathway presents a potential therapeutic avenue for sepsis. Recently, we identified isaridin E (ISE), a marine-derived fungal cyclohexadepsipeptide, as a promising antiplatelet and antithrombotic agent with a low bleeding risk. ISE\'s influence on septic mortality and sepsis-induced lung injury in a mouse model of sepsis, induced by caecal ligation and puncture, is investigated in this study. ISE dose-dependently improved survival rates, mitigating lung injury, thrombocytopenia, pulmonary endothelial permeability, and vascular inflammation in the mouse model. ISE markedly curtailed vWF release from activated platelets in septic mice by suppressing vesicle-associated membrane protein 8 and soluble N-ethylmaleide-sensitive factor attachment protein 23 overexpression. Moreover, ISE inhibited healthy human platelet adhesion to cultured lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs), thereby significantly decreasing vWF secretion and endothelial hyperpermeability. Using cilengitide, a selective integrin αvβ3 inhibitor, it was found that ISE can improve endothelial hyperpermeability by inhibiting vWF binding to αvβ3. Activation of the integrin αvβ3-FAK/Src pathway likely underlies vWF-induced endothelial dysfunction in sepsis. In conclusion, ISE protects against sepsis by inhibiting endothelial hyperpermeability and platelet-endothelium interactions.
    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
    黄病毒如登革热,Zika,和西尼罗河病毒是高度关注对公众健康构成重大风险的病原体。NS1蛋白在黄病毒中是保守的,并且作为黄病毒多蛋白的一部分合成。它在病毒复制中起关键作用,疾病进展,和免疫逃避。翻译后修饰影响NS1的稳定性,分泌,抗原性,以及与宿主因素的相互作用。NS1蛋白与宿主细胞蛋白形成广泛的相互作用,使其能够影响RNA加工等重要过程。基因表达调控,和细胞内稳态,进而影响病毒的复制,疾病的发病机理,和免疫反应。NS1通过延迟补体依赖性的受感染细胞的裂解而充当免疫逃避因子,并且通过诱导内皮细胞损伤和血管渗漏以及触发自身免疫应答而有助于疾病的发病机理。抗NS1抗体已被证明与宿主内皮细胞和血小板发生交叉反应,导致自身免疫破坏,假设有助于疾病的发病机理。然而,相比之下,用NS1蛋白免疫动物模型可保护免受黄病毒如登革热和寨卡病毒的致命攻击。了解NS1在黄病毒发病机制中的多方面作用对于有效的疾病管理和控制至关重要。因此,对NS1生物学的进一步研究,包括其宿主蛋白相互作用和在疾病病理中的其他作用,对于开发成功对抗黄病毒感染的策略和疗法至关重要。这篇综述提供了关于NS1蛋白在黄病毒发病机理中的多方面作用的现有知识的深入探索。
    Flaviviruses such as dengue, Zika, and West Nile viruses are highly concerning pathogens that pose significant risks to public health. The NS1 protein is conserved among flaviviruses and is synthesized as a part of the flavivirus polyprotein. It plays a critical role in viral replication, disease progression, and immune evasion. Post-translational modifications influence NS1\'s stability, secretion, antigenicity, and interactions with host factors. NS1 protein forms extensive interactions with host cellular proteins allowing it to affect vital processes such as RNA processing, gene expression regulation, and cellular homeostasis, which in turn influence viral replication, disease pathogenesis, and immune responses. NS1 acts as an immune evasion factor by delaying complement-dependent lysis of infected cells and contributes to disease pathogenesis by inducing endothelial cell damage and vascular leakage and triggering autoimmune responses. Anti-NS1 antibodies have been shown to cross-react with host endothelial cells and platelets, causing autoimmune destruction that is hypothesized to contribute to disease pathogenesis. However, in contrast, immunization of animal models with the NS1 protein confers protection against lethal challenges from flaviviruses such as dengue and Zika viruses. Understanding the multifaceted roles of NS1 in flavivirus pathogenesis is crucial for effective disease management and control. Therefore, further research into NS1 biology, including its host protein interactions and additional roles in disease pathology, is imperative for the development of strategies and therapeutics to combat flavivirus infections successfully. This Review provides an in-depth exploration of the current available knowledge on the multifaceted roles of the NS1 protein in the pathogenesis of flaviviruses.
    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
    众所周知,肺血管渗漏,脓毒症肺损伤的一个关键病理特征,主要受血管周围细胞调节。然而,潜在的机制尚未完全发现。在本研究中,我们的目的是评估Isthmin1的作用,一种源自肺泡上皮的分泌蛋白,在脓毒症期间肺血管渗漏中的作用,并探讨isthmin1基因转录的调控机制。我们观察到由盲肠结扎和穿孔(CLP)诱导的脓毒症小鼠的肺组织中Ithmin1基因表达升高,以及暴露于脂多糖(LPS)的原代小鼠肺泡Ⅱ型上皮细胞(ATⅡ)。Further,我们证实来自ATⅡ的isthmin1有助于脓毒症时的肺血管渗漏。具体来说,腺病毒介导的ATⅡ中的isthmin1破坏导致暴露于LPS时,PMVEC/ATⅡ共培养系统中肺微血管内皮细胞(PMVEC)通透性增加。此外,腺相关病毒9(AAV9)介导的脓毒血症小鼠肺泡上皮中的isthmin1敲除可显着减轻肺血管渗漏。最后,机制研究表明,核转录因子C/EBPβ通过直接与isthmin1基因座的顺式调节元件结合而参与isthmin1基因的激活,并可能有助于脓毒症期间isthmin1的上调。总的来说,本研究强调了来自ATⅡ的旁分泌蛋白isthmin1的影响,研究脓毒症肺血管通透性的加重,揭示了一种新的isthmin1基因转录调控机制。
    It is known that pulmonary vascular leakage, a key pathological feature of sepsis-induced lung injury, is largely regulated by perivascular cells. However, the underlying mechanisms have not been fully uncovered. In the present study, we aimed to evaluate the role of isthmin1, a secretory protein originating from alveolar epithelium, in the pulmonary vascular leakage during sepsis and to investigate the regulatory mechanisms of isthmin1 gene transcription. We observed an elevated isthmin1 gene expression in the pulmonary tissue of septic mice induced by cecal ligation and puncture (CLP), as well as in primary murine alveolar type II epithelial cells (ATII) exposed to lipopolysaccharide (LPS). Furthermore, we confirmed that isthmin1 derived from ATII contributes to pulmonary vascular leakage during sepsis. Specifically, adenovirus-mediated isthmin1 disruption in ATII led to a significant attenuation of the increased pulmonary microvascular endothelial cell (PMVEC) hyperpermeability in a PMVEC/ATII coculture system when exposed to LPS. In addition, adeno-associated virus 9 (AAV9)-mediated knockdown of isthmin1 in the alveolar epithelium of septic mice significantly attenuated pulmonary vascular leakage. Finally, mechanistic studies unveiled that nuclear transcription factor CCAAT/enhancer binding protein (C/EBP)β participates in isthmin1 gene activation by binding directly to the cis-regulatory element of isthmin1 locus and may contribute to isthmin1 upregulation during sepsis. Collectively, the present study highlighted the impact of the paracrine protein isthmin1, derived from ATII, on the exacerbation of pulmonary vascular permeability in sepsis and revealed a new regulatory mechanism for isthmin1 gene transcription.NEW & NOTEWORTHY This article addresses the role of the alveolar epithelial-secreted protein isthmin1 on the exacerbation of pulmonary vascular permeability in sepsis and identified nuclear factor CCAAT/enhancer binding protein (C/EBP)β as a new regulator of isthmin1 gene transcription. Targeting the C/EBPβ-isthmin1 regulatory axis on the alveolar side would be of great value in the treatment of pulmonary vascular leakage and lung injury induced by sepsis.
    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: Randomized Controlled Trial
    背景:血管渗漏是急性呼吸窘迫综合征(ARDS)的主要特征。我们旨在评估FX06的疗效,这是一种正在开发的稳定内皮细胞间连接的药物,在减少SARS-CoV-2诱导的ARDS期间的血管渗漏。
    方法:这个多中心,双盲,随机试验包括患有COVID-19相关ARDS的成年人,他们接受有创机械通气<5天,并随机接受静脉FX06(400mg/d,5天)或其载体作为安慰剂。主要终点是从第1天到第7天降低经肺热稀释衍生的血管外肺水指数(EVLWi)。
    结果:25名患者随机接受FX06和24名安慰剂。尽管EVLWi在基线时升高(中位数[IQR]15.6mL/kg[13.5;18.5]),FX06接受者和对照组从第1天到第7天的下降幅度相当(分别,-1.9[-3.3;-0.5]vs.-0.8[-5.5;-1.1]毫升/千克;估计效果-0.8[-3.1;+2.4],p=0.51)。心脏指数,肺血管通透性指数,和流体平衡也是可比的,PaO2/FiO2比值和机械通气持续时间。两组的不良事件发生率相似,尽管更多的FX06接受者出现了呼吸机相关性肺炎(16/25(64%)vs.6/24(24%),p=0.009)。
    结论:在这项独特的给药方案研究中,FX06不能降低SARS-CoV-2诱导的肺血管渗漏。未来的研究将需要在疾病期间或使用其他方案的早期评估其疗效。试用登记NCT04618042。2020年11月5日注册。
    Vascular leakage is a major feature of acute respiratory distress syndrome (ARDS). We aimed to evaluate the efficacy of FX06, a drug under development that stabilizes interendothelial cell junctions, at reducing vascular leakage during SARS-CoV-2-induced ARDS.
    This multicenter, double-blinded, randomized trial included adults with COVID-19-associated ARDS who had received invasive mechanical ventilation for < 5 days and were randomized to receive either intravenous FX06 (400 mg/d, for 5 days) or its vehicle as placebo. The primary endpoint was the lowering-from day 1 to day 7-of the transpulmonary thermodilution-derived extravascular lung-water index (EVLWi).
    Twenty-five patients were randomized to receive FX06 and 24 the placebo. Although EVLWi was elevated at baseline (median [IQR] 15.6 mL/kg [13.5; 18.5]), its declines from day 1 to day 7 were comparable for FX06 recipients and controls (respectively, - 1.9 [- 3.3; - 0.5] vs. - 0.8 [- 5.5; - 1.1] mL/kg; estimated effect - 0.8 [- 3.1; + 2.4], p = 0.51). Cardiac indexes, pulmonary vascular permeability indexes, and fluid balances were also comparable, as were PaO2/FiO2 ratios and durations of mechanical ventilation. Adverse event rates were similar for the 2 groups, although more FX06 recipients developed ventilator-associated pneumonia (16/25 (64%) vs. 6/24 (24%), p = 0.009).
    In this unique-dosing-regimen study, FX06 did not lower SARS-CoV-2-induced pulmonary vascular leakage. Future investigations will need to evaluate its efficacy at earlier times during the disease or using other regimens. Trial registration NCT04618042. Registered 5 November 2020.
    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
    目的:血脑屏障(BBB)的损伤是缺血性卒中早期脑损伤的标志。随后的内皮高通透性驱动最初的病理变化并加重神经元死亡。瞬时受体电位美司他丁2(TRPM2)是由氧化应激激活的Ca2通透性非选择性阳离子通道。然而,目前尚不清楚TRPM2是否参与缺血性卒中期间的BBB降解.我们旨在研究TRPM2在缺血性卒中BBB降解中的作用及其潜在的分子机制。
    方法:和结果:使用Cdh5Cre在内皮细胞中特异性缺失Trpm2对大脑中动脉闭塞(MCAO)小鼠的脑损伤产生有效的保护作用,其特征是梗死面积减小,缓解血浆外渗,抑制免疫细胞侵袭和抑制氧化应激。使用培养的脑内皮细胞(CEC)的体外实验表明,Trpm2缺失或TRPM2激活的抑制可减弱氧化应激,Ca2+过载,和氧-葡萄糖剥夺(OGD)和CD36配体血小板反应蛋白1(TSP1)诱导的内皮高通透性。在转染的HEK293T细胞中,OGD和TSP1以CD36依赖性方式激活TRPM2。值得注意的是,在培养的CECs中,删除Trpm2或抑制TRPM2激活也抑制了CD36的激活和OGD或TSP1诱导的细胞功能障碍。
    结论:结论:我们的数据揭示了一种新的分子机制,其中TRPM2和CD36促进彼此的激活,这加剧了缺血性中风期间的内皮功能障碍。我们的研究表明,内皮细胞中的TRPM2是开发更有效,更安全的缺血性中风疗法的有希望的靶标。
    Damage of the blood-brain barrier (BBB) is a hallmark of brain injury during the early stages of ischemic stroke. The subsequent endothelial hyperpermeability drives the initial pathological changes and aggravates neuronal death. Transient receptor potential melastatin 2 (TRPM2) is a Ca2+-permeable nonselective cation channel activated by oxidative stress. However, whether TRPM2 is involved in BBB degradation during ischemic stroke remains unknown. We aimed to investigate the role of TRPM2 in BBB degradation during ischemic stroke and the underlying molecular mechanisms.
    Specific deletion of Trpm2 in endothelial cells using Cdh5 Cre produces a potent protective effect against brain injury in mice subjected to middle cerebral artery occlusion (MCAO), which is characterized by reduced infarction size, mitigated plasma extravasation, suppressed immune cell invasion, and inhibited oxidative stress. In vitro experiments using cultured cerebral endothelial cells (CECs) demonstrated that either Trpm2 deletion or inhibition of TRPM2 activation attenuates oxidative stress, Ca2+ overload, and endothelial hyperpermeability induced by oxygen-glucose deprivation (OGD) and CD36 ligand thrombospondin-1 (TSP1). In transfected HEK293T cells, OGD and TSP1 activate TRPM2 in a CD36-dependent manner. Noticeably, in cultured CECs, deleting Trpm2 or inhibiting TRPM2 activation also suppresses the activation of CD36 and cellular dysfunction induced by OGD or TSP1.
    In conclusion, our data reveal a novel molecular mechanism in which TRPM2 and CD36 promote the activation of each other, which exacerbates endothelial dysfunction during ischemic stroke. Our study suggests that TRPM2 in endothelial cells is a promising target for developing more effective and safer therapies for ischemic stroke.
    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
    内皮屏障功能的破坏和细胞迁移的减少导致内皮功能障碍。最丰富的人乳寡糖之一,6'-唾液酸乳糖(6'-SL),据报道发挥与炎症反应相关的各种生物学功能。在这项研究中,我们评估了6'-SL对脂多糖(LPS)诱导的内皮屏障损伤引起的炎症的影响。我们的结果表明,500ng/mL的LPS不仅强烈地消除了细胞迁移,而且还过度激活了MAPK和NF-κB通路。6'-SL通过ERK1/2、p38和JNKMAPK途径抑制LPS诱导的内皮炎症。6'-SL通过上调PECAM-1表达和紧密连接的mRNA水平支持内皮连接,如ZO-1和闭塞蛋白,通过LPS刺激下调。显著抑制NF-κB的核转位,随着炎症细胞因子的下调,包括TNF-α,IL-1β,MCP-1、VCAM-1和ICAM-1。此外,6\'-SL通过下调STAT3激活和核易位来消除NF-κB介导的STAT3,从而控制内皮迁移和高通透性。最后,LPS诱导小鼠主动脉动脉粥样硬化和动脉粥样硬化保护区域中VCAM-1和ZO-1的过度表达,通过6'-SL治疗逆转。总之,我们的研究结果表明,6'-SL是调节炎症反应和内皮通透性增高的有效治疗剂.
    Disruption of the endothelial barrier function and reduction in cell migration leads to endothelial dysfunction. One of the most abundant human milk oligosaccharides, 6\'-sialylactose (6\'-SL), is reported to exert various biological functions related to inflammatory responses. In this study, we evaluated the effects of 6\'-SL on lipopolysaccharide (LPS)-induced inflammation caused by endothelial barrier damage. Our results showed that LPS at 500 ng/mL strongly not only abolished cell migration but also hyperactivated MAPK and NF-κB pathways. 6\'-SL suppressed LPS-induced endothelial inflammation via ERK1/2, p38, and JNK MAPK pathways. 6\'-SL supported endothelial junctions by upregulating PECAM-1 expression and mRNA levels of tight junctions, such as ZO-1 and occludin, which were downregulated by LPS stimulation. It significantly inhibited the nuclear translocation of NF-κB, along with the downregulation of inflammatory cytokines, including TNF-α, IL-1β, MCP-1, VCAM-1, and ICAM-1. Furthermore, 6\'-SL abolished NF-κB-mediated STAT3 in controlling endothelial migration and hyperpermeability via downregulating STAT3 activation and nuclear translocation. Finally, LPS induced over-expression of VCAM-1 and ZO-1 disassembly in both atheroprone and atheroprotective areas of mouse aorta, which were reversed by 6\'-SL treatment. Altogether, our findings suggest that 6\'-SL is a potent therapeutic agent for modulating inflammatory responses and endothelial hyperpermeability.
    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
    黄病毒非结构蛋白1(NS1)由感染细胞分泌,并以组织依赖性方式导致内皮屏障功能障碍和血管渗漏。这种现象部分地通过破坏内衬内皮的内皮糖萼层(EGL)而发生。此外,我们和其他人已经表明,可溶性DENVNS1诱导细胞间连接(IJCs)的分解,一组对维持内皮稳态和调节血管通透性至关重要的细胞蛋白;然而,NS1介导IJC破坏的具体机制尚不清楚.这里,我们调查了五种黄病毒NS1蛋白的相对贡献,来自登革热(DENV),Zika(ZIKV),西尼罗河(WNV),日本脑炎(JEV),黄热病(YFV)病毒,细胞间连接蛋白β-catenin和VE-cadherin在人脐静脉和脑组织内皮细胞中的表达和定位。我们发现黄病毒NS1以组织依赖性方式诱导β-catenin和VE-cadherin的错位,反映黄病毒病嗜性。机械上,我们观察到NS1处理细胞触发了VE-cadherin的内化,可能是通过网格蛋白介导的内吞作用,和β-连环蛋白的磷酸化,激活糖原合成酶激酶-3β(GSK-3β)的内皮屏障破坏过程中经典IJC重塑途径的一部分。支持这个模型,我们发现GSK-3β的化学抑制剂在体外降低了NS1诱导的人脐静脉和脑微血管内皮细胞单层的通透性,并在小鼠背侧皮内模型中降低了血管渗漏。这些发现提供了对调节NS1介导的内皮功能障碍的分子机制的见解,并确定了GSK-3β作为治疗严重登革热疾病期间血管渗漏的潜在治疗靶标。
    The flavivirus nonstructural protein 1 (NS1) is secreted from infected cells and contributes to endothelial barrier dysfunction and vascular leak in a tissue-dependent manner. This phenomenon occurs in part via disruption of the endothelial glycocalyx layer (EGL) lining the endothelium. Additionally, we and others have shown that soluble DENV NS1 induces disassembly of intercellular junctions (IJCs), a group of cellular proteins critical for maintaining endothelial homeostasis and regulating vascular permeability; however, the specific mechanisms by which NS1 mediates IJC disruption remain unclear. Here, we investigated the relative contribution of five flavivirus NS1 proteins, from dengue (DENV), Zika (ZIKV), West Nile (WNV), Japanese encephalitis (JEV), and yellow fever (YFV) viruses, to the expression and localization of the intercellular junction proteins β-catenin and VE-cadherin in endothelial cells from human umbilical vein and brain tissues. We found that flavivirus NS1 induced the mislocalization of β-catenin and VE-cadherin in a tissue-dependent manner, reflecting flavivirus disease tropism. Mechanistically, we observed that NS1 treatment of cells triggered internalization of VE-cadherin, likely via clathrin-mediated endocytosis, and phosphorylation of β-catenin, part of a canonical IJC remodeling pathway during breakdown of endothelial barriers that activates glycogen synthase kinase-3β (GSK-3β). Supporting this model, we found that a chemical inhibitor of GSK-3β reduced both NS1-induced permeability of human umbilical vein and brain microvascular endothelial cell monolayers in vitro and vascular leakage in a mouse dorsal intradermal model. These findings provide insight into the molecular mechanisms regulating NS1-mediated endothelial dysfunction and identify GSK-3β as a potential therapeutic target for treatment of vascular leakage during severe dengue disease.
    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
    脂多糖(LPS)加剧全身性炎症反应并导致过多的液体渗漏。已发现2,4,6-三羟基-3-香叶基苯乙酮(tHGA)在体外可防止LPS诱导的血管炎症和内皮通透性过高。
    本研究评估了tHGA对内毒素血症小鼠中LPS诱导的全身性炎症和血管通透性的体内保护作用。
    BALB/c小鼠腹膜内用tHGA预处理1小时,然后进行6小时的LPS诱导。在用2、20和100mg/kgtHGA预处理的小鼠(n=6)中进行伊文思蓝渗透性测定和白细胞迁移测定。tHGA(20、40和80mg/kg)对LPS诱导的血清TNF-α分泌的影响,使用ELISA评估肺功能障碍和致死率(n=6),组织病理学分析(n=6)和生存能力测定(n=10),分别。生理盐水和地塞米松作为阴性对照和药物对照。分别。
    tHGA在2、20和100mg/kg时显着抑制血管通透性,抑制百分比为48%,85%和86%,分别,与LPS对照组相比(IC50=3.964mg/kg)。在20和100mg/kg剂量下抑制白细胞浸润,抑制百分比分别为73%和81%,(IC50=17.56mg/kg)。然而,所有tHGA剂量(20,40和80mg/kg)均不能预防内毒素血症小鼠的致死性,因为tHGA不能抑制TNF-α过度产生和器官功能障碍.
    tHGA可以通过与其他抗炎剂组合而被开发为与不受控制的血管渗漏有关的疾病的潜在治疗剂。
    UNASSIGNED: Lipopolysaccharide (LPS) exacerbates systemic inflammatory responses and causes excessive fluid leakage. 2,4,6-Trihydroxy-3-geranyl acetophenone (tHGA) has been revealed to protect against LPS-induced vascular inflammation and endothelial hyperpermeability in vitro.
    UNASSIGNED: This study assesses the in vivo protective effects of tHGA against LPS-induced systemic inflammation and vascular permeability in endotoxemic mice.
    UNASSIGNED: BALB/c mice were intraperitoneally pre-treated with tHGA for 1 h, followed by 6 h of LPS induction. Evans blue permeability assay and leukocyte transmigration assay were performed in mice (n = 6) pre-treated with 2, 20 and 100 mg/kg tHGA. The effects of tHGA (20, 40 and 80 mg/kg) on LPS-induced serum TNF-α secretion, lung dysfunction and lethality were assessed using ELISA (n = 6), histopathological analysis (n = 6) and survivability assay (n = 10), respectively. Saline and dexamethasone were used as the negative control and drug control, respectively.
    UNASSIGNED: tHGA significantly inhibited vascular permeability at 2, 20 and 100 mg/kg with percentage of inhibition of 48%, 85% and 86%, respectively, in comparison to the LPS control group (IC50=3.964 mg/kg). Leukocyte infiltration was suppressed at 20 and 100 mg/kg doses with percentage of inhibition of 73% and 81%, respectively (IC50=17.56 mg/kg). However, all tHGA doses (20, 40 and 80 mg/kg) failed to prevent endotoxemic mice from lethality because tHGA could not suppress TNF-α overproduction and organ dysfunction.
    UNASSIGNED: tHGA may be developed as a potential therapeutic agent for diseases related to uncontrolled vascular leakage by combining with other anti-inflammatory agents.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    Endothelium protection is critical, because of the impact of vascular leakage and edema on pathological conditions such as brain ischemia. Whereas deficiency of class II phosphoinositide 3-kinase alpha (PI3KC2α) results in an increase in vascular permeability, we uncover a crucial role of the beta isoform (PI3KC2β) in the loss of endothelial barrier integrity following injury. Here, we studied the role of PI3KC2β in endothelial permeability and endosomal trafficking in vitro and in vivo in ischemic stroke. Mice with inactive PI3KC2β showed protection against vascular permeability, edema, cerebral infarction, and deleterious inflammatory response. Loss of PI3KC2β in human cerebral microvascular endothelial cells stabilized homotypic cell-cell junctions by increasing Rab11-dependent VE-cadherin recycling. These results identify PI3KC2β as a potential new therapeutic target to prevent aggravating lesions following ischemic stroke.
    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
    Endothelial cells lining the inner vascular wall form a monolayer that contributes to the selective permeability of endothelial barrier. This selective permeability is mainly regulated by an endothelium-specific adherens junctional protein, known as vascular endothelial-cadherin (VE-cadherin). In endothelial cells, the adherens junction comprises of VE-cadherin and its associated adhesion molecules such as p120, α-catenin, and β-catenin, in which α-catenin links cytoplasmic tails of VE-cadherin to actin cytoskeleton through β-catenin. Proinflammatory stimuli such as lipopolysaccharide (LPS) are capable of attenuating vascular integrity through the disruption of VE-cadherin adhesion in endothelial cells. To date, numerous studies demonstrated the disruption of adherens junction as a result of phosphorylation-mediated VE-cadherin disruption. However, the outcomes from these studies were inconsistent and non-conclusive as different cell fractions were used to examine the effect of LPS on the disruption of VE-cadherin. By using Western Blot, some studies utilized total protein lysate and reported decreased protein expression while some studies reported unchanged expression. Other studies which used membrane and cytosolic fractions of protein extract demonstrated decreased and increased VE-cadherin expression, respectively. Despite the irregularities, the results of immunofluorescence staining are consistent with the formation of intercellular gap. Besides that, the overall underlying disruptive mechanisms of VE-cadherin remain largely unknown. Therefore, this mini review will focus on different experiment approaches in terms of cell fractions used in different human endothelial cell studies, and relate these differences to the results obtained in Western blot and immunofluorescence staining in order to give some insights into the overall differential regulatory mechanisms of LPS-mediated VE-cadherin disruption and address the discrepancy in VE-cadherin expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号