Myosin light chain kinase

肌球蛋白轻链激酶
  • 文章类型: Journal Article
    内皮细胞中细胞质Ca2+的严格控制对于内皮屏障功能的调节是必不可少的。这里,我们研究了电压门控Ca2+(Cav)通道亚基Cavβ3的作用,在调节脑微血管内皮细胞(BMECs)中的Ca2信号传导以及这如何有助于血脑屏障的完整性。
    我们通过Ca2+成像和蛋白质印迹研究了Cavβ3在BMEC中的功能,检查了体外内皮屏障功能和体内血脑屏障的完整性,并使用Cavβ3-/-(Cavβ3缺陷型)小鼠作为对照,评估了小鼠诱导实验性自身免疫性脑脊髓炎后的病程。
    我们鉴定了BMEC中的Cavβ3蛋白,但是电生理记录没有显示出明显的Cav通道活性。在体内,在不存在Cavβ3的情况下,血脑屏障完整性降低.诱导实验性自身免疫性脑脊髓炎后,Cavβ3-/-小鼠表现出更早的疾病发作,加剧了临床残疾和增加的T细胞浸润。体外,Cavβ3-/-BMEC单层的跨内皮阻力低于野生型BMEC单层,并且连接蛋白ZO-1(闭塞带-1)的组织受损。凝血酶刺激肌醇1,4,5-三磷酸依赖性Ca2+释放,其通过MLC(肌球蛋白轻链)的Ca2+依赖性磷酸化促进细胞收缩并增强内皮屏障通透性。这些影响在Cavβ3-/-中比在野生型BMECs中更明显,而在MLCK(MLC激酶)抑制剂ML-7的存在下,差异被消除。Cacnb3cDNA在Cavβ3-/-BMECs中的表达恢复了野生型表型。共免疫沉淀和质谱表明Cavβ3与肌醇1,4,5-三磷酸受体蛋白的关联。
    独立于其作为Cav通道亚基的功能,Cavβ3与肌醇1,4,5-三磷酸受体相互作用,并参与BMEC中细胞质Ca2和Ca2依赖性MLC磷酸化的严格控制,Cavβ3在BMEC中的这种作用有助于血脑屏障的完整性,并减轻实验性自身免疫性脑脊髓炎疾病的严重程度。
    UNASSIGNED: Tight control of cytoplasmic Ca2+ in endothelial cells is essential for the regulation of endothelial barrier function. Here, we investigated the role of Cavβ3, a subunit of voltage-gated Ca2+ (Cav) channels, in modulating Ca2+ signaling in brain microvascular endothelial cells (BMECs) and how this contributes to the integrity of the blood-brain barrier.
    UNASSIGNED: We investigated the function of Cavβ3 in BMECs by Ca2+ imaging and Western blot, examined the endothelial barrier function in vitro and the integrity of the blood-brain barrier in vivo, and evaluated disease course after induction of experimental autoimmune encephalomyelitis in mice using Cavβ3-/- (Cav β3-deficient) mice as controls.
    UNASSIGNED: We identified Cavβ3 protein in BMECs, but electrophysiological recordings did not reveal significant Cav channel activity. In vivo, blood-brain barrier integrity was reduced in the absence of Cavβ3. After induction of experimental autoimmune encephalomyelitis, Cavβ3-/- mice showed earlier disease onset with exacerbated clinical disability and increased T-cell infiltration. In vitro, the transendothelial resistance of Cavβ3-/- BMEC monolayers was lower than that of wild-type BMEC monolayers, and the organization of the junctional protein ZO-1 (zona occludens-1) was impaired. Thrombin stimulates inositol 1,4,5-trisphosphate-dependent Ca2+ release, which facilitates cell contraction and enhances endothelial barrier permeability via Ca2+-dependent phosphorylation of MLC (myosin light chain). These effects were more pronounced in Cavβ3-/- than in wild-type BMECs, whereas the differences were abolished in the presence of the MLCK (MLC kinase) inhibitor ML-7. Expression of Cacnb3 cDNA in Cavβ3-/- BMECs restored the wild-type phenotype. Coimmunoprecipitation and mass spectrometry demonstrated the association of Cavβ3 with inositol 1,4,5-trisphosphate receptor proteins.
    UNASSIGNED: Independent of its function as a subunit of Cav channels, Cavβ3 interacts with the inositol 1,4,5-trisphosphate receptor and is involved in the tight control of cytoplasmic Ca2+ and Ca2+-dependent MLC phosphorylation in BMECs, and this role of Cavβ3 in BMECs contributes to blood-brain barrier integrity and attenuates the severity of experimental autoimmune encephalomyelitis disease.
    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
    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
    炎症性肠病(IBD)的主要原因是肠道通透性异常,这是由于通过病原体介导的炎症机制破坏了肠道屏障的紧密连接以及肠道微生物群的失衡。本研究旨在评估2-酮戊二酸是否通过紧密连接定位缓解了渗透性功能障碍,激活转化生长因子β激活激酶1(TAK1)炎症途径,并在体外和体内IBD模型中调节肠道微生物组的稳态。我们的发现表明,2-酮戊二酸显着抑制异常肠通透性,肠细胞紧密连接蛋白的离域,炎性细胞因子的表达,如TNF-α,在体外和体内。发现2-酮戊二酸直接与TAK1结合并抑制TNF受体相关因子6(TRAF6)-TAK1相互作用,与核因子κB(NF-κB)途径的激活有关,从而调节丝裂原活化蛋白激酶的表达。膳食2-酮戊二酸还可以缓解肠道微生物群失调和IBD症状,如肠道长度的改善和双鞭毛杆菌的丰度所证明的那样,在结肠炎小鼠中未分类的Coriobacteriaceae_UCG_002和Ruminococaceae_。这项研究表明,2-酮戊二酸与TAK1结合以抑制与NF-κB途径相关的活性,并通过调节紧密连接定位和肠道微生物组稳态来减轻异常通透性。因此,2-酮戊二酸是用于治疗IBD的有效营养制剂和益生元。
    The main cause of inflammatory bowel disease (IBD) is abnormal intestinal permeability due to the disruption of the tight junction of the intestinal barrier through a pathogen-mediated inflammatory mechanism and an imbalance of the gut microbiota. This study aimed to evaluate whether 2-ketoglutaric acid alleviated permeability dysfunction with tight junction localization, activated the transforming growth factor beta-activated kinase 1 (TAK1) inflammation pathway, and regulated the homeostasis of the intestinal microbiome in vitro and in vivo IBD model. Our findings revealed that 2-ketoglutaric acid significantly suppressed abnormal intestinal permeability, delocalization of tight junction proteins from the intestinal cell, expression of inflammatory cytokines, such as TNF-α, both in vitro and in vivo. 2-Ketoglutaric acid was found to directly bind to TAK1 and inhibit the TNF receptor-associated factor 6 (TRAF6)-TAK1 interaction, which is related to the activation of nuclear factor kappa B (NF-κB) pathways, thereby regulating the expression of mitogen-activated protein kinase. Dietary 2-ketoglutaric acid also alleviated gut microbiota dysbiosis and IBD symptoms, as demonstrated by improvements in the intestine length and the abundance of Ligilactobacillus, Coriobacteriaceae_UCG_002, and Ruminococcaceae_unclassified in mice with colitis. This study indicated that 2-ketoglutaric acid binds to TAK1 for activity inhibition which is related to the NF-κB pathway and alleviates abnormal permeability by regulating tight junction localization and gut microbiome homeostasis. Therefore, 2-ketoglutaric acid is an effective nutraceutical agent and prebiotic for the treatment of IBD.
    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:心脏特异性肌球蛋白轻链激酶(cMLCK),由MYLK3编码,通过心室肌球蛋白调节轻链的磷酸化调节心脏收缩力。然而,cMLCK在人类心力衰竭中的病理生理和治疗意义尚不清楚.我们旨在研究cMLCK失调是否会导致心脏功能障碍,以及cMLCK的恢复是否可能是收缩性心力衰竭的新型肌力疗法。
    方法:我们产生了具有家族性扩张型心肌病相关MYLK3移码突变(MYLK3+/fs)的敲入小鼠(Mylk3+/fs和Mylk3fs/fs),该突变已被我们先前鉴定(c.1951-1G>T;p.P639Vfs*15)和人类诱导的多能干细胞衍生突变。我们还开发了一种新的cMLCK小分子激活剂(LEUO-1154)。
    结果:两只小鼠(Mylk3+/fs和Mylk3fs/fs)由于无义介导的信使RNA衰变而显示cMLCK表达降低,减少心肌中的MLC2v(心室肌球蛋白调节轻链)磷酸化,和cMLCK剂量依赖性方式的收缩功能障碍。与这个结果一致,来自突变小鼠的心肌显示心脏超松弛/无序松弛状态的比率增加,这可能导致心脏收缩力受损。通过通过AAV9_MYLK3载体补充cMLCK来挽救在敲入小鼠中观察到的表型。MYLK3+/fs诱导的多能干细胞衍生的心肌细胞cMLCK表达减少50%,收缩功能障碍,伴随着超松弛/无序松弛比的增加。CRISPR介导的基因校正,或通过AAV9_MYLK3载体补充cMLCK,成功恢复cMLCK表达,超弛豫/无序弛豫比,和收缩功能障碍。LEUO-1154在心室肌球蛋白调节轻链磷酸化的Vmax中增加人cMLCK活性约2倍,而不影响Km。LEUO-1154治疗人MYLK3+/fs诱导的多能干细胞衍生的心肌细胞恢复了心室肌球蛋白调节轻链磷酸化水平和超松弛/无序松弛率,并改善了心脏收缩力,而不影响钙瞬变,表明cMLCK激活剂充当肌胶。最后,多种原因导致的晚期心力衰竭的人心肌MYLK3/PPP1R12B信使RNA表达率明显低于对照心脏,提示肌球蛋白调节轻链激酶和磷酸酶之间的平衡在衰竭的心肌,不管原因。
    结论:cMLCK失调有助于人类心脏收缩功能障碍的发展。我们恢复cMLCK活性的策略可以为晚期收缩性心力衰竭的新型肌力疗法奠定基础。
    Cardiac-specific myosin light chain kinase (cMLCK), encoded by MYLK3, regulates cardiac contractility through phosphorylation of ventricular myosin regulatory light chain. However, the pathophysiological and therapeutic implications of cMLCK in human heart failure remain unclear. We aimed to investigate whether cMLCK dysregulation causes cardiac dysfunction and whether the restoration of cMLCK could be a novel myotropic therapy for systolic heart failure.
    We generated the knock-in mice (Mylk3+/fs and Mylk3fs/fs) with a familial dilated cardiomyopathy-associated MYLK3 frameshift mutation (MYLK3+/fs) that had been identified previously by us (c.1951-1G>T; p.P639Vfs*15) and the human induced pluripotent stem cell-derived cardiomyocytes from the carrier of the mutation. We also developed a new small-molecule activator of cMLCK (LEUO-1154).
    Both mice (Mylk3+/fs and Mylk3fs/fs) showed reduced cMLCK expression due to nonsense-mediated messenger RNA decay, reduced MLC2v (ventricular myosin regulatory light chain) phosphorylation in the myocardium, and systolic dysfunction in a cMLCK dose-dependent manner. Consistent with this result, myocardium from the mutant mice showed an increased ratio of cardiac superrelaxation/disordered relaxation states that may contribute to impaired cardiac contractility. The phenotypes observed in the knock-in mice were rescued by cMLCK replenishment through the AAV9_MYLK3 vector. Human induced pluripotent stem cell-derived cardiomyocytes with MYLK3+/fs mutation reduced cMLCK expression by 50% and contractile dysfunction, accompanied by an increased superrelaxation/disordered relaxation ratio. CRISPR-mediated gene correction, or cMLCK replenishment by AAV9_MYLK3 vector, successfully recovered cMLCK expression, the superrelaxation/disordered relaxation ratio, and contractile dysfunction. LEUO-1154 increased human cMLCK activity ≈2-fold in the Vmax for ventricular myosin regulatory light chain phosphorylation without affecting the Km. LEUO-1154 treatment of human induced pluripotent stem cell-derived cardiomyocytes with MYLK3+/fs mutation restored the ventricular myosin regulatory light chain phosphorylation level and superrelaxation/disordered relaxation ratio and improved cardiac contractility without affecting calcium transients, indicating that the cMLCK activator acts as a myotrope. Finally, human myocardium from advanced heart failure with a wide variety of causes had a significantly lower MYLK3/PPP1R12B messenger RNA expression ratio than control hearts, suggesting an altered balance between myosin regulatory light chain kinase and phosphatase in the failing myocardium, irrespective of the causes.
    cMLCK dysregulation contributes to the development of cardiac systolic dysfunction in humans. Our strategy to restore cMLCK activity could form the basis of a novel myotropic therapy for advanced systolic heart failure.
    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
    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
    Hughes-Stovin综合征是一种罕见的疾病,其特征是血栓性静脉炎和多发性肺和/或支气管动脉瘤。HSS的病因和发病机制尚不完全清楚。目前的共识是血管炎是致病过程的基础,动脉壁炎症后出现肺血栓。因此,Hughes-Stovin综合征可能属于Behçet综合征肺受累的血管簇,虽然口述阿普泰,关节炎,很少发现葡萄膜炎。Behçet综合征是一种多因素多基因疾病,表观遗传,环境,主要是免疫学贡献者。不同的Behçet综合征表型可能是基于涉及一种以上致病途径的不同遗传决定因素。Hughes-Stovin综合征可能与纤维肌肉发育不良和其他随血管动脉瘤发展的疾病有共同的途径。我们描述了符合Behçet综合征标准的Hughes-Stovin综合征病例。检测到未知意义的MYLK变异体,以及其他可能影响血管生成途径的基因杂合突变。我们讨论了这些基因发现的可能参与,以及Behçet/Hughes-Stovin综合征和血管Behçet综合征中动脉瘤的其他潜在共同决定因素。诊断技术的最新进展,包括基因检测,可以帮助诊断特定的Behçet综合征亚型和其他相关疾病,以个性化疾病管理。
    Hughes-Stovin syndrome is a rare disease characterized by thrombophlebitis and multiple pulmonary and/or bronchial aneurysms. The etiology and pathogenesis of HSS are incompletely known. The current consensus is that vasculitis underlies the pathogenic process, and pulmonary thrombosis follows arterial wall inflammation. As such, Hughes-Stovin syndrome may belong to the vascular cluster with lung involvement of Behçet syndrome, although oral aphtae, arthritis, and uveitis are rarely found. Behçet syndrome is a multifactorial polygenic disease with genetic, epigenetic, environmental, and mostly immunological contributors. The different Behçet syndrome phenotypes are presumably based upon different genetic determinants involving more than one pathogenic pathway. Hughes-Stovin syndrome may have common pathways with fibromuscular dysplasias and other diseases evolving with vascular aneurysms. We describe a Hughes-Stovin syndrome case fulfilling the Behçet syndrome criteria. A MYLK variant of unknown significance was detected, along with other heterozygous mutations in genes that may impact angiogenesis pathways. We discuss the possible involvement of these genetic findings, as well as other potential common determinants of Behçet/Hughes-Stovin syndrome and aneurysms in vascular Behçet syndrome. Recent advances in diagnostic techniques, including genetic testing, could help diagnose a specific Behçet syndrome subtype and other associated conditions to personalize the disease management.
    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

公众号