Myosin light chain kinase

肌球蛋白轻链激酶
  • 文章类型: Journal Article
    心脏病的全球发病率正在增加,给医疗保健系统带来巨大的社会经济负担。心血管疾病的发病机制复杂,尚未完全了解,心脏的生理功能与调节良好的心肌运动密不可分。肌球蛋白轻链激酶(MLCK)是心肌收缩和舒张所必需的,心脏电生理稳态,血管神经收缩和血压调节。在这个意义上,MLCK似乎是心脏疾病的有吸引力的治疗靶标。MLCK通过多种途径参与心肌细胞运动和迁移,包括钙稳态的调节,肌球蛋白轻链磷酸化的激活,和刺激血管平滑肌细胞收缩或松弛。最近,肌球蛋白轻链的磷酸化已被证明与心肌运动信号的激活密切相关,MLCK通过肌球蛋白粗丝和肌动蛋白细丝的相互作用介导心脏的收缩和舒张功能。它通过维护细胞骨架的完整性起作用,改变肌球蛋白头部的构象,调节神经支配。MLCK控制血管收缩和舒张功能,并与肾上腺素能和交感神经系统的激活有关。细胞外运输,内皮通透性,以及一氧化氮和血管紧张素II的调节。此外,MLCK在心脏衰老过程中起着至关重要的作用。多种天然产物/植物化学物质和化合物,比如槲皮素,环孢菌素,和盐酸ML-7,已被证明可以调节心肌细胞MLCK。在设计新的治疗剂时应考虑这些化合物的MLCK修饰能力。这篇综述总结了MLCK在心血管系统中的作用机制,以及报道的化合物通过改变MLCK过程在心脏病中的治疗潜力。
    The global incidence of cardiac diseases is increasing, imposing a substantial socioeconomic burden on healthcare systems. The pathogenesis of cardiovascular disease is complex and not fully understood, and the physiological function of the heart is inextricably linked to well-regulated cardiac muscle movement. Myosin light chain kinase (MLCK) is essential for myocardial contraction and diastole, cardiac electrophysiological homeostasis, vasoconstriction of vascular nerves and blood pressure regulation. In this sense, MLCK appears to be an attractive therapeutic target for cardiac diseases. MLCK participates in myocardial cell movement and migration through diverse pathways, including regulation of calcium homeostasis, activation of myosin light chain phosphorylation, and stimulation of vascular smooth muscle cell contraction or relaxation. Recently, phosphorylation of myosin light chains has been shown to be closely associated with the activation of myocardial exercise signaling, and MLCK mediates systolic and diastolic functions of the heart through the interaction of myosin thick filaments and actin thin filaments. It works by upholding the integrity of the cytoskeleton, modifying the conformation of the myosin head, and modulating innervation. MLCK governs vasoconstriction and diastolic function and is associated with the activation of adrenergic and sympathetic nervous systems, extracellular transport, endothelial permeability, and the regulation of nitric oxide and angiotensin II. Additionally, MLCK plays a crucial role in the process of cardiac aging. Multiple natural products/phytochemicals and chemical compounds, such as quercetin, cyclosporin, and ML-7 hydrochloride, have been shown to regulate cardiomyocyte MLCK. The MLCK-modifying capacity of these compounds should be considered in designing novel therapeutic agents. This review summarizes the mechanism of action of MLCK in the cardiovascular system and the therapeutic potential of reported chemical compounds in cardiac diseases by modifying MLCK processes.
    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
    肺动脉高压(PH)是一种难治性疾病,其特征是肺动脉压力和阻力升高。减阻聚合物(DRP)是通过改变血液动力学和流变学来降低血管阻力的血液可溶性大分子。我们以前的工作表明,聚环氧乙烷(PEO)可以显着降低肺动脉的内壁厚度和血管阻力,但具体机制尚不清楚。
    本研究旨在研究PEO对PH中低剪切应力(LSS)诱导的内皮细胞(ECs)细胞内钙[Ca2]i和细胞骨架蛋白的作用和机制。在BioFlux200流动系统中,使原代肺动脉内皮细胞(PAEC)经受稳定的LSS(1dyn/cm2)或生理剪切应力(SS)(10dyn/cm2)20小时。进行了钙内流测定以评估PEO对[Ca2]i的机制。随后,服用诱导细胞骨架重塑的关键蛋白,调节轻链(RLC)磷酸化,作为突破口,这项研究集中于PEO调节RLC磷酸化的两个关键途径:肌球蛋白轻链激酶(MLCK)和Rho相关激酶(ROCK)途径。
    我们目前的研究表明,在LSS(1dyn/cm2)的PEO显着抑制了LSS诱导的[Ca2]i和瞬时受体电位通道1(TRPC1)的表达水平。此外,ECs将LSS刺激转化为细胞骨架蛋白的上调,包括丝状肌动蛋白(F-肌动蛋白),MLCK,ROCK,p-RLC,和pp-RLC。使用药理学抑制剂的进一步实验表明,LSS上的PEO主要通过ROCK和MLCK途径下调细胞骨架相关蛋白。
    本研究以细胞内钙和细胞骨架重排为切入点,研究PEO在生物医学领域的应用,对PH的治疗具有重要的理论意义和实际应用价值。
    UNASSIGNED: Pulmonary hypertension (PH) is a refractory disease characterized by elevated pulmonary artery pressure and resistance. Drag-reducing polymers (DRPs) are blood-soluble macromolecules that reduce vascular resistance by altering the blood dynamics and rheology. Our previous work indicated that polyethylene oxide (PEO) can significantly reduce the medial wall thickness and vascular resistance of the pulmonary arteries, but the specific mechanism is still unclear.
    UNASSIGNED: This study was designed to investigate the role and mechanism of PEO on intracellular calcium [Ca2 +] i and cytoskeletal proteins of endothelial cells (ECs) induced by low shear stress (LSS) in PH. Primary Pulmonary Artery Endothelial Cells (PAECs) were subjected to steady LSS (1 dyn/cm2) or physiological shear stress (SS) (10 dyn/cm2) for 20 h in a BioFlux 200 flow system. Calcium influx assays were conducted to evaluate the mechanisms of PEO on [Ca2 +] i. Subsequently, taking the key protein that induces cytoskeletal remodeling, the regulatory light chain (RLC) phosphorylation, as the breakthrough point, this study focused on the two key pathways of PEO that regulate phosphorylation of RLC: Myosin light chain kinase (MLCK) and Rho-associated kinase (ROCK) pathways.
    UNASSIGNED: Our current research revealed that PEO at LSS (1 dyn/cm2) significantly suppressed LSS-induced [Ca2 +] i and the expression level of transient receptor potential channel 1(TRPC1). In addition, ECs convert LSS stimuli into the upregulation of cytoskeletal proteins, including filamentous actin (F-actin), MLCK, ROCK, p-RLC, and pp-RLC. Further experiments using pharmacological inhibitors demonstrated that PEO at the LSS downregulated cytoskeleton-related proteins mainly through the ROCK and MLCK pathways.
    UNASSIGNED: This study considered intracellular calcium and cytoskeleton rearrangement as entry points to study the application of PEO in the biomedical field, which has important theoretical significance and practical application value for the treatment of PH.
    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
    目的:我们先前的研究发现,与对照组相比,口腔扁平苔藓(OLP)患者颊粘膜表面的黑素普氏菌(Pm)的组成比例显着增加。此外,Pm可侵入OLP患者的上皮。本研究旨在进一步探讨Pm对口腔角质形成细胞的影响。
    方法:建立Pm-人口腔角质形成细胞(HOK)共培养模型,检测单层通透性,闭塞带-1(ZO-1)表达,和Pm的细胞内存活。我们进行RNA-seq,然后鉴定差异表达基因(DEG)和基因本体论(GO)分析,特别关注肌球蛋白轻链激酶(MLCK)。在Pm-HOK共培养模型中使用MLCK抑制剂ML-7来评估其对单层通透性和ZO-1表达的影响。
    结果:HOK单层渗透率增加,共培养后ZO-1表达降低(p<0.05)。Pm可以在HOK细胞中存活。RNA-seq显示MLCK是一种上调的普通DEG。MLCK在Pm-HOK共培养模型中的表达上调。抑制MLCK挽救了上皮通透性的增加,ZO-1表达上调(p<0.05)。
    结论:MLCK可能参与Pm破坏上皮屏障功能。
    OBJECTIVE: Our previous studies have found that the composition ratio of Prevotella melaninogenica (Pm) on buccal mucosa surface of oral lichen planus (OLP) patients increased significantly compared with control. Furthermore, Pm could invade the epithelium of OLP patients. This study aimed to further explore the impact of Pm on oral keratinocytes.
    METHODS: The Pm-human oral keratinocyte (HOK) co-culture model was established to detect monolayer permeability, zona occludens-1 (ZO-1) expression, and intracellular survival of Pm. We performed RNA-seq followed by identification of differentially expressed genes (DEGs) and Gene Ontology (GO) analysis, with a particular focus on myosin light chain kinase (MLCK). An MLCK inhibitor ML-7 was utilized in Pm-HOK co-culture model to assess its effects on monolayer permeability and ZO-1 expression.
    RESULTS: HOK monolayer permeability was increased, and ZO-1 expression was decreased after co-culture (p < 0.05). Pm could survive in HOK cells. RNA-seq revealed MLCK was an upregulated common DEG. The expression of MLCK in the Pm-HOK co-culture model was upregulated. Inhibition of MLCK rescued the increased epithelial permeability, and ZO-1 expression was upregulated (p < 0.05).
    CONCLUSIONS: MLCK may be involved in disrupting epithelial barrier function by Pm.
    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
    氟化物是地下水和农产品的常见污染物,这对动物和人类健康构成了挑战。大量的研究已经证明了其对肠粘膜完整性的有害影响;然而,潜在的机制仍然模糊。本研究旨在探讨细胞骨架在氟化物诱导的屏障功能障碍中的作用。氟化钠(NaF)处理培养的Caco-2细胞后,观察到细胞毒性和细胞形态学变化(内部空泡或大量消融).NaF降低了跨上皮电阻(TEER)并增强了异硫氰酸荧光素葡聚糖4(FD-4)的细胞旁渗透,表明Caco-2单层高渗透性。同时,NaF处理改变了紧密连接蛋白ZO-1的表达和分布。氟化物暴露会增加肌球蛋白轻链II(MLC2)的磷酸化并触发肌动蛋白丝(F-肌动蛋白)的重塑。当Blebistatin对肌球蛋白II的抑制作用阻止NaF诱导的屏障破坏和ZO-1不连续性时,相应的激动剂离子霉素具有与氟化物相当的作用,表明MLC2是效应子。鉴于p-MLC2调节的上游机制,进一步研究表明,NaF激活了RhoA/ROCK信号通路和肌球蛋白轻链激酶(MLCK),显著增加两者的表达。药理学抑制剂(Rhosin,Y-27632和ML-7)逆转了NaF诱导的屏障击穿和应力纤维形成。研究了细胞内钙离子([Ca2]i)在NaF对Rho/ROCK途径和MLCK的影响中的作用。我们发现NaF升高[Ca2+]i,而螯合剂BAPTA-AM减弱增加RhoA和MLCK表达以及ZO-1破裂,因此,恢复屏障功能。总的来说,上述结果表明,NaF通过Ca2+依赖的RhoA/ROCK通路和MLCK诱导屏障损伤,进而触发MLC2磷酸化和ZO-1和F-肌动蛋白的重排。这些结果为氟化物诱导的肠损伤提供了潜在的治疗靶点。
    Fluoride is a common contaminant of groundwater and agricultural commodity, which poses challenges to animal and human health. A wealth of research has demonstrated its detrimental effects on intestinal mucosal integrity; however, the underlying mechanisms remain obscure. This study aimed to investigate the role of the cytoskeleton in fluoride-induced barrier dysfunction. After sodium fluoride (NaF) treatment of the cultured Caco-2 cells, both cytotoxicity and cytomorphological changes (internal vacuoles or massive ablation) were observed. NaF lowered transepithelial electrical resistance (TEER) and enhanced paracellular permeation of fluorescein isothiocyanate dextran 4 (FD-4), indicating Caco-2 monolayers hyperpermeability. In the meantime, NaF treatment altered both the expression and distribution of the tight junction protein ZO-1. Fluoride exposure increased myosin light chain II (MLC2) phosphorylation and triggered actin filament (F-actin) remodeling. While inhibition of myosin II by Blebbistatin blocked NaF-induced barrier failure and ZO-1 discontinuity, the corresponding agonist Ionomycin had effects comparable to those of fluoride, suggesting that MLC2 serves as an effector. Given the mechanisms upstream of p-MLC2 regulation, further studies demonstrated that NaF activated RhoA/ROCK signaling pathway and myosin light chain kinase (MLCK), strikingly increasing the expression of both. Pharmacological inhibitors (Rhosin, Y-27632 and ML-7) reversed NaF-induced barrier breakdown and stress fiber formation. The role of intracellular calcium ions ([Ca2+]i) in NaF effects on Rho/ROCK pathway and MLCK was investigated. We found that NaF elevated [Ca2+]i, whereas chelator BAPTA-AM attenuated increased RhoA and MLCK expression as well as ZO-1 rupture, thus, restoring barrier function. Collectively, abovementioned results suggest that NaF induces barrier impairment via Ca2+-dependent RhoA/ROCK pathway and MLCK, which in turn triggers MLC2 phosphorylation and rearrangement of ZO-1 and F-actin. These results provide potential therapeutic targets for fluoride-induced intestinal 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
    在重症急性胰腺炎(SAP)中,肠黏膜屏障损伤可引起肠道细菌移位,诱发或加重全身感染。血红素加氧酶-1(HO-1)是一种有效的抗氧化剂和细胞保护剂。本研究旨在探讨HO-1对SAP诱导的SAP大鼠肠屏障损伤的作用及机制。健康成年雄性SD大鼠随机分为假手术组,SAP组,SAP+Hemin集团,和SAP+Znpp组。采用胰胆管逆行注射牛磺胆酸钠(5%)建立大鼠SAP模型。在SAP之前24小时,在选定的组中腹膜内注射Hemin(有效的HO-1激活剂)和Znpp(HO-1的竞争性抑制剂)。每组24小时后收集血清和肠组织样品进行分析。Hemin预处理显著减少全身炎症,肠道氧化应激,并通过增加HO-1的表达而使SAP中的肠上皮凋亡。同时,Hemin预处理消除了对紧密连接蛋白表达的抑制作用,并显着抑制了MLCK/P-MLC信号通路的激活。相反,ZnPP完全逆转了这些作用。我们的研究表明,HO-1表达的上调可减轻SAP的肠粘膜屏障损伤。HO-1对肠的保护作用归因于MLCK/p-MLC信号通路抑制。
    In severe acute pancreatitis (SAP), intestinal mucosal barrier damage can cause intestinal bacterial translocation and induce or aggravate systemic infections. Heme oxygenase-1 (HO-1) is a validated antioxidant and cytoprotective agent. This research aimed to investigate the effect and mechanism of HO-1 on SAP-induced intestinal barrier damage in SAP rats. Healthy adult male Sprague-Dawley rats were randomly separated into the sham-operated group, SAP group, SAP + Hemin group, and SAP + Znpp group. The rat model of SAP was established by retrograde injection of sodium taurocholate (5%) into the biliopancreatic duct. Hemin (a potent HO-1 activator) and Znpp (a competitive inhibitor of HO-1) were injected intraperitoneally in the selected groups 24 h before SAP. Serum and intestinal tissue samples were collected for analysis after 24 h in each group. Hemin pretreatment significantly reduced systemic inflammation, intestinal oxidative stress, and intestinal epithelial apoptosis in SAP by increasing HO-1 expression. Meanwhile, pretreatment with Hemin abolished the inhibitory effect on the expression of the tight junction proteins and significantly inhibited the activation of the MLCK/P-MLC signaling pathway. Conversely, ZnPP completely reversed these effects. Our study indicates that upregulation of HO-1 expression attenuates the intestinal mucosal barrier damage in SAP. The protective effect of HO-1 on the intestine is attributed to MLCK/p-MLC signaling pathway inhibition.
    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
    黄曲霉毒素B1(AFB1)是一种广泛存在于食品和饲料中的霉菌毒素。尽管肝脏是AFB1的主要靶器官,但肠是AFB1的第一个暴露器官。然而,AFB1通过调节法尼醇X受体(FXR)介导的肌球蛋白轻链激酶(MLCK)信号通路而引起肠屏障功能障碍的机制研究甚少。在体内,AFB1暴露显着降低了小肠长度并增加了肠通透性。同时,AFB1暴露显著抑制FXR的蛋白表达,ZO-1,闭塞蛋白,和claudin-1并增强MLCK的蛋白表达。体外,AFB1暴露通过FITC-Dextran4kDa通量的升高和以剂量依赖性方式抑制跨上皮电阻来诱导肠屏障功能障碍。此外,AFB1暴露下调FXR的mRNA和蛋白表达,ZO-1,闭塞蛋白,和claudin-1,重新分配ZO-1蛋白,并增强MLCK和p-MLC的蛋白表达。然而,fexaramine(Fex,FXR激动剂)预处理显著逆转AFB1诱导的FXR活性降低,MLCK蛋白激活,和体内外肠道屏障损伤。此外,用ML-7抑制MLCK预处理可显着减轻AFB1诱导的肠屏障功能障碍和紧密连接破坏。总之,AFB1通过在体外和体内调节FXR介导的MLCK信号通路诱导肠屏障损伤,为预防肠道真菌毒素中毒提供了新的见解。
    Aflatoxin B1 (AFB1) is a widespread mycotoxin in food and feed. Although the liver is the main target organ of AFB1, the intestine is the first exposure organ to AFB1. However, the mechanism by which AFB1 induced intestinal barrier dysfunction via regulating the farnesoid X receptor (FXR)-mediated myosin light chain kinase (MLCK) signaling pathway has rarely been studied. In vivo, AFB1 exposure significantly decreased the small intestine length and increased the intestinal permeability. Meanwhile, AFB1 exposure markedly suppressed the protein expressions of FXR, ZO-1, occludin, and claudin-1 and enhanced the protein expression of MLCK. In vitro, AFB1 exposure induced intestinal barrier dysfunction by the elevation in the FITC-Dextran 4 kDa flux and inhibition in the transepithelial electrical resistance in a dose-dependent manner. In addition, AFB1 exposure downregulated the mRNA and protein expressions of FXR, ZO-1, occludin, and claudin-1, redistributed the ZO-1 protein, and enhanced the protein expressions of MLCK and p-MLC. However, fexaramine (Fex, FXR agonist) pretreatment markedly reversed the AFB1-induced FXR activity reduction, MLCK protein activation, and intestinal barrier impairment in vitro and in vivo. Moreover, pretreatment with the inhibition of MLCK with ML-7 significantly alleviated the AFB1-induced intestinal barrier dysfunction and tight junction disruption in vitro. In conclusion, AFB1 induced intestinal barrier impairment via regulating the FXR-mediated MLCK signaling pathway in vitro and in vivo and provided novel insights to prevent mycotoxin poisoning in the intestine.
    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:肠屏障破坏,肠缺血再灌注(I/R)的常见并发症,包括功能障碍和肠道结构改变,其特征在于紧密连接的丧失和肠屏障的增强的渗透性以及增加的死亡率。开发有效和新颖的治疗方法对于改善肠屏障恶化患者的预后至关重要。据报道,重组人血管生成素样蛋白4(rhANGPTL4)在外源给药时可保护血脑屏障,内源性ANGPTL4缺乏可使放射性肠损伤恶化。
    目的:确定rhANGPTL4是否可以保护I/R引起的肠屏障破坏。
    方法:夹住肠系膜上动脉60分钟,再灌注240分钟,引起肠I/R损伤。通过缺氧/复氧攻击肠上皮细胞(Caco-2)和人脐静脉内皮细胞以体外模拟I/R。
    结果:指标包括异硫氰酸荧光素结合的葡聚糖(4千道尔顿;FD-4)清除率,磷酸化肌球蛋白轻链/总肌球蛋白轻链的比例,在肠道I/R或细胞缺氧/复氧后,肌球蛋白轻链激酶和闭合带1,claudin-2和VE-cadherin的丢失显着增加。rhANGPTL4治疗显著逆转了这些指标,与抑制炎症和氧化级联反应有关,过度激活细胞自噬和凋亡,提高生存率。当细胞受到缺氧/复氧时,在体外观察到类似的结果,而rhANGPTL4显著逆转了Caco-2细胞和人脐静脉内皮细胞中接近正常水平的指标。
    结论:rhANGPTL4可作为肠道I/R所致肠道损伤的保护剂,通过维持肠道屏障的结构和功能来改善存活。
    BACKGROUND: Intestinal barrier breakdown, a frequent complication of intestinal ischemia-reperfusion (I/R) including dysfunction and the structure changes of the intestine, is characterized by a loss of tight junction and enhanced permeability of the intestinal barrier and increased mortality. To develop effective and novel therapeutics is important for the improvement of outcome of patients with intestinal barrier deterioration. Recombinant human angiopoietin-like protein 4 (rhANGPTL4) is reported to protect the blood-brain barrier when administered exogenously, and endogenous ANGPTL4 deficiency deteriorates radiation-induced intestinal injury.
    OBJECTIVE: To identify whether rhANGPTL4 may protect intestinal barrier breakdown induced by I/R.
    METHODS: Intestinal I/R injury was elicited through clamping the superior mesenteric artery for 60 min followed by 240 min reperfusion. Intestinal epithelial (Caco-2) cells and human umbilical vein endothelial cells were challenged by hypoxia/ reoxygenation to mimic I/R in vitro.
    RESULTS: Indicators including fluorescein isothiocyanate-conjugated dextran (4 kilodaltons; FD-4) clearance, ratio of phosphorylated myosin light chain/total myosin light chain, myosin light chain kinase and loss of zonula occludens-1, claudin-2 and VE-cadherin were significantly increased after intestinal I/R or cell hypoxia/reoxygenation. rhANGPTL4 treatment significantly reversed these indicators, which were associated with inhibiting the inflammatory and oxidative cascade, excessive activation of cellular autophagy and apoptosis and improvement of survival rate. Similar results were observed in vitro when cells were challenged by hypoxia/reoxygenation, whereas rhANGPTL4 reversed the indicators close to normal level in Caco-2 cells and human umbilical vein endothelial cells significantly.
    CONCLUSIONS: rhANGPTL4 can function as a protective agent against intestinal injury induced by intestinal I/R and improve survival via maintenance of intestinal barrier structure and functions.
    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
    The neonicotinoid pesticide, imidacloprid (IMI), is frequently detected in the environment and in foods. It is absorbed and metabolized by the intestine; however, its effects on intestinal barrier integrity are not well studied. We investigated whether IMI disrupts the permeability of the intestinal epithelial barrier via in vivo tests on male Wistar rats, in vitro assays using the human intestinal epithelial cell line, Caco-2, and in silico analyses. A repeated oral dose 90-day toxicity study was performed (0.06 mg/kg body weight/day). IMI exposure significantly increased intestinal permeability, which led to significantly elevated serum levels of endotoxin and inflammatory biomarkers (tumor necrosis factor-alpha and interleukin-1 beta) without any variation in body weight. Decreased transepithelial electrical resistance with increased permeability was also observed in 100 nM and 100 μM IMI-treated Caco-2 cell monolayers. Amounts of tight junction proteins in IMI-treated colon tissues and between IMI-treated Caco-2 cells were significantly lower than those of controls. Increased levels of myosin light chain phosphorylation, myosin light chain kinase (MLCK), and p65 subunit of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB p65) phosphorylation were found in IMI-exposed cells compared with control cells. Furthermore, the barrier loss caused by IMI was rescued by the MLCK inhibitor, ML-7, and cycloheximide. Pregnane X receptor (PXR, NR1I2) was inhibited by low-dose IMI treatment. In silico analysis indicated potent binding sites between PXR and IMI. Together, these data illustrate that IMI induces intestinal epithelial barrier disruption and produces an inflammatory response, involving the down-regulation of tight junctions and disturbance of the PXR-NF-κB p65-MLCK signaling pathway. The intestinal barrier disruption caused by IMI deserves attention in assessing the safety of this neonicotinoid pesticide.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    证据表明,镇静右美托咪定可以预防肠功能障碍。然而,其对烧伤引起的肠屏障损伤的保护作用的具体机制尚不清楚.我们旨在探讨右美托咪定对烧伤诱导的肠屏障损伤的可能积极作用以及在烧伤实验模型中对肌球蛋白轻链激酶(MLCK)/磷酸化肌球蛋白轻链(p-MLC)信号通路的影响。
    在这项研究中,测量异硫氰酸荧光素标记的葡聚糖(FITC-葡聚糖)的血浆浓度。使用苏木精和伊红(HE)染色评价组织学变化。通过蛋白质印迹和免疫荧光分析评估紧密连接蛋白,以评估肠紧密连接的结构完整性。采用酶联免疫吸附试验(ELISA)检测炎症水平。
    结果表明,烧伤引起的肠道通透性增加伴随着对肠道的组织学损伤,紧密连接蛋白ZonulaOccludens-1(ZO-1)和Occludin的表达降低,炎性细胞因子水平的增加以及MLCK蛋白表达和MLC磷酸化的升高。右美托咪定治疗后,烧伤引起的变化得到改善。
    总之,右美托咪定发挥抗炎作用,并通过抑制MLCK/p-MLC信号通路保护紧密连接复合物免受烧伤诱导的肠屏障损伤.
    Evidence suggests that sedative dexmedetomidine can prevent intestinal dysfunction. However, the specific mechanisms of its protective effects against burn-induced intestinal barrier injury remain unclear. We aimed to explore the possible positive effects of dexmedetomidine on burn-induced intestinal barrier injury and the effects the myosin light chain kinase (MLCK)/phosphorylated myosin light chain (p-MLC) signalling pathway in an experimental model of burn injury.
    In this study, the plasma concentration of fluorescein isothiocyanate-labelled dextran (FITC-dextran) was measured. Histological changes were evaluated using haematoxylin and eosin (HE) staining. Tight junction proteins were evaluated by western blot and immunofluorescence analyses to assess the structural integrity of intestinal tight junctions. The level of inflammation was detected by enzyme-linked immunosorbent assay (ELISA).
    The results shows that the increase in intestinal permeability caused by burn injury is accompanied by histological damage to the intestine, decreases in the expression of the tight junction proteins Zonula Occludens-1 (ZO-1) and Occludin, increases in inflammatory cytokine levels and elevation of both MLCK protein expression and MLC phosphorylation. After dexmedetomidine treatment, the burn-induced changes were ameliorated.
    In conclusion, dexmedetomidine exerted an anti-inflammatory effect and protected tight junction complexes against burn‑induced intestinal barrier damage by inhibiting the MLCK/p-MLC signalling pathways.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Tight junction dysregulation and epithelial damage contribute to intestinal barrier loss in patients with acute liver failure (ALF); however, the regulatory mechanisms of these processes remain poorly understood. The aim of the present study was to investigate the changes of intestinal tight junction and intestinal mucosa in mice with ALF and their mechanisms. In the present study, ALF was induced in mice through an intraperitoneal injection of D‑galactosamine and lipopolysaccharide (D‑GalN/LPS), and the morphological changes of the liver or small intestine were analyzed using hematoxylin and eosin staining, scanning electron microscopy (SEM) and transmission electron microscopy (TEM). The intestinal tissues and isolated serum were analyzed using western blotting, immunofluorescence staining and ELISA. D‑GalN/LPS‑induced mice exhibited signs of hepatocyte necrosis, alongside inflammatory cell infiltration into the liver tissue and partial microvilli detachment in the small intestinal mucosa. TEM demonstrated that the intestinal epithelial tight junctions were impaired, whereas SEM micrographs revealed the presence of abnormal microvilli in D‑GalN/LPS‑induced mice. In addition, the expression levels of phosphorylated (p)‑myosin light chain (MLC), MLC kinase (MLCK) and Rho‑associated kinase (ROCK) were significantly increased in the D‑GalN/LPS‑induced mice compared with those in the control mice, whereas the subsequent inhibition of MLCK or ROCK significantly reduced p‑MLC expression levels. Conversely, the expression levels of occludin and zonula occludens‑1 (ZO‑1) were significantly decreased in the D‑GalN/LPS‑induced mice, and the inhibition of MLCK or ROCK significantly increased occludin and ZO‑1 protein expression levels compared with those in the control group. Changes in the serum levels of tumor necrosis factor‑α (TNF‑α) and interleukin (IL)‑6 were similar to the trend observed in p‑MLC expression levels. In conclusion, the findings of the present study suggested that in a D‑GalN/LPS‑induced ALF model, TNF‑α and IL‑6 signaling may increase MLCK and ROCK expression levels, further mediate phosphorylation of MLC, which may result in tight junction dysregulation and intestinal barrier dysfunction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号