Myosin Light Chains

肌球蛋白轻链
  • 文章类型: Journal Article
    肥厚型心肌病(HCM)是一种以左心室(LV)增厚为特征的心脏遗传疾病,过度收缩,和受损的放松。HCM主要是由肌节蛋白的可遗传突变引起的,如β肌球蛋白重链。直到最近,临床上用于HCM的药物并不直接针对肌节潜在的收缩变化.这里,我们研究了一个新的小分子,RLC-1,在牛心脏肌原纤维高通量筛选中鉴定。RLC-1高度依赖于调节轻链的存在来结合心脏肌球蛋白并调节其ATP酶活性。在脱膜的大鼠LV小梁中,RLC-1降低了最大Ca2+激活力和力的Ca2+敏感性,同时增加了张力重建的次最大速率常数。在从大鼠LV分离的肌原纤维中,最大和次最大Ca2激活力都降低了近50%。此外,松弛的快速和慢速阶段大约是DMSO对照的两倍,缓慢阶段的持续时间较短。在结构上,x射线衍射研究表明,RLC-1使肌球蛋白头部远离粗丝骨架,并降低了肌球蛋白头部的顺序,这与其他肌球蛋白抑制剂不同。在完整的小梁和分离的心肌细胞中,RLC-1处理导致峰值抽搐幅度降低以及更快的激活和松弛动力学。总之,RLC-1在脱膜组织中加速动力学和减少的力产生,完整的组织,和完整的整个细胞,导致较小的心脏抽搐,这可以改善与HCM相关的潜在收缩变化。
    Hypertrophic cardiomyopathy (HCM) is a genetic disease of the heart characterized by thickening of the left ventricle (LV), hypercontractility, and impaired relaxation. HCM is caused primarily by heritable mutations in sarcomeric proteins, such as β myosin heavy chain. Until recently, medications in clinical use for HCM did not directly target the underlying contractile changes in the sarcomere. Here, we investigate a novel small molecule, RLC-1, identified in a bovine cardiac myofibril high-throughput screen. RLC-1 is highly dependent on the presence of a regulatory light chain to bind to cardiac myosin and modulate its ATPase activity. In demembranated rat LV trabeculae, RLC-1 decreased maximal Ca2+-activated force and Ca2+ sensitivity of force, while it increased the submaximal rate constant for tension redevelopment. In myofibrils isolated from rat LV, both maximal and submaximal Ca2+-activated force are reduced by nearly 50%. Additionally, the fast and slow phases of relaxation were approximately twice as fast as DMSO controls, and the duration of the slow phase was shorter. Structurally, x-ray diffraction studies showed that RLC-1 moved myosin heads away from the thick filament backbone and decreased the order of myosin heads, which is different from other myosin inhibitors. In intact trabeculae and isolated cardiomyocytes, RLC-1 treatment resulted in decreased peak twitch magnitude and faster activation and relaxation kinetics. In conclusion, RLC-1 accelerated kinetics and decreased force production in the demembranated tissue, intact tissue, and intact whole cells, resulting in a smaller cardiac twitch, which could improve the underlying contractile changes associated with HCM.
    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
    脓毒症诱导的急性肺损伤(SALI)是脓毒症的常见并发症,导致高发病率和死亡率。SALI的主要发病机制是急性炎症和内皮屏障损伤之间的相互作用。研究表明山奈酚(KPF)具有抗脓毒症作用。鞘氨醇激酶1(Sphk1)/鞘氨醇-1-磷酸(S1P)信号通路在急性肺损伤和S1P受体1(S1PR1)激动剂在肌球蛋白轻链2(MLC2)磷酸化中的潜在意义。KPF是否能调节SphK1/S1P/SIPR1/MLC2信号通路保护肺内皮屏障尚不清楚。本研究探讨了KPF对LPS诱导的脓毒症小鼠和人脐静脉内皮细胞(HUVECs)内皮细胞屏障损伤的修复作用及其分子机制。在脓毒症小鼠模型中,KPF通过降低IL-6和TNF-α的合成显著减轻肺组织损伤并显示抗炎作用。Further,KPF可降低LPS诱导的内皮细胞屏障的高通透性,减轻肺内皮细胞屏障损伤。机制研究表明,KPF预处理可以抑制MLC2过度磷酸化,降低SphK1、S1P、和S1PR1级别。SphK1/S1P/S1PR1/MLC2信号通路控制与内皮屏障损伤相关的下游蛋白,蛋白质印迹(WB)显示KPF升高了蛋白质水平。这些蛋白质包括闭塞带(ZO)-1,血管内皮(VE)-钙黏着蛋白和Occludin。目前的工作表明,在表现出由LPS引发的败血症的小鼠中,KPF加强了内皮屏障,减轻了炎症反应。SphK1/S1P/S1PR1/MLC2通路的调节是这种影响的潜在机制。
    Sepsis-induced acute lung injury (SALI) is the common complication of sepsis, resulting in high incidence and mortality rates. The primary pathogenesis of SALI is the interplay between acute inflammation and endothelial barrier damage. Studies have shown that kaempferol (KPF) has anti-sepsis properties. Sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P) signaling pathway\'s significance in acute lung damage and S1P receptor 1 (S1PR1) agonists potential in myosin light chain 2 (MLC2) phosphorylation are documented. Whether KPF can regulate the SphK1/S1P/S1PR1/MLC2 signaling pathway to protect the lung endothelial barrier remains unclear. This study investigates the KPF\'s therapeutic effects and molecular mechanisms in repairing endothelial cell barrier damage in both LPS-induced sepsis mice and human umbilical vein endothelial cells (HUVECs). KPF significantly reduced lung tissue damage and showed anti-inflammatory effects by decreasing IL-6 and TNF-α synthesis in the sepsis mice model. Further, KPF administration can reduce the high permeability of the LPS-induced endothelial cell barrier and alleviate lung endothelial cell barrier injury. Mechanistic studies showed that KPF pretreatment can suppress MLC2 hyperphosphorylation and decrease SphK1, S1P, and S1PR1 levels. The SphK1/S1P/S1PR1/MLC2 signaling pathway controls the downstream proteins linked to endothelial barrier damage, and the Western blot (WB) showed that KPF raised the protein levels. These proteins include zonula occludens (ZO)-1, vascular endothelial (VE)-cadherin and Occludin. The present work revealed that in mice exhibiting sepsis triggered by LPS, KPF strengthened the endothelial barrier and reduced the inflammatory response. The SphK1/S1P/S1PR1/MLC2 pathway\'s modulation is the mechanism underlying this impact.
    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: English Abstract
    研究整合素和N-cadherin介导的机械粘附对间充质干细胞极化的协同调节以及潜在的机械生物学机制。
    双层聚乙烯乙二醇(PEG)水凝胶用RGD和HAVDI肽配制和修饰,分别,实现对整合素和N-钙粘蛋白的机械粘附,并复制细胞与细胞外基质之间的整合素介导的机械相互作用以及N-钙粘蛋白介导的细胞-细胞机械相互作用。极性蛋白质,磷脂酰肌醇3-激酶(PI3K)和磷酸化肌球蛋白轻链(pMLC),在整合素介导的粘附下,通过免疫荧光染色在有或没有与HAVDI肽接触的单个细胞中进行表征,N-钙粘蛋白介导的粘附,和不同的细胞内力。使用ImageJ分析了它们的表达水平和极性分布。
    整合素介导的粘附诱导的PI3K和pMLC的极性强度明显高于无接触组,导致PI3K对β-catenin的极角浓度在135°至180°的范围内,而pMLC对β-catenin的极角浓度在0°至45°的范围内。整合素功能的抑制导致PI3K在接触基团中极性分布的抑制,但没有改变pMLC蛋白的极性分布。N-钙粘蛋白对PI3K和pMLC极性分布的影响与整合素相似。然而,抑制N-cadherin的机械粘附导致接触组中PI3K和pMLC蛋白的极性强度和极性角分布受到抑制。此外,抑制N-cadherin的机械粘附会导致整合素β1的极性强度减弱,从而降低整合素β1和β-catenin之间极性角集中在135°至180°范围内的细胞比例。此外,细胞内力影响PI3K和pMLC蛋白的极性分布。减少细胞内力削弱了PI3K和pMLC蛋白的极性强度及其极性分布,而细胞内力的增加增强了PI3K和pMLC蛋白的极性强度及其极性分布。
    整合素和N-cadherin共同调节细胞蛋白的极性分布,N-cadherin可以通过局部抑制整合素在干细胞的极性调节中起重要作用。
    UNASSIGNED: To investigate the synergistic regulation of the polarization of mesenchymal stem cells by integrin and N-cadherin-mediated mechanical adhesion and the underlying mechanobiological mechanisms.
    UNASSIGNED: Bilayer polyethylene glyeol (PEG) hydrogels were formulated and modified with RGD and HAVDI peptides, respectively, to achieve mechanical adhesion to integrin and N-cadherin and to replicate the integrin-mediated mechanical interaction between cells and the extracellular matrix and the N-cadherin-mediated cell-cell mechanical interaction. The polar proteins, phosphatidylinositol 3-kinase (PI3K) and phosphorylated myosin light chain (pMLC), were characterized through immunofluorescence staining in individual cells with or without contact with HAVDI peptides under integrin-mediated adhesion, N-cadherin-mediated adhesion, and different intracellular forces. Their expression levels and polar distribution were analyzed using Image J.
    UNASSIGNED: Integrin-mediated adhesion induced significantly higher polar strengths of PI3K and pMLC in the contact group than in those in the no contact group, resulting in the concentration of the polar angle of PI3K to β-catenin in the range of 135° to 180° and the concentration of the polar angle of pMLC to β-catenin in the range of 0° to 45° in the contact group. Inhibition of integrin function led to inhibition of the polarity distribution of PI3K in the contact group, but did not change the polarity distribution of pMLC protein. The effect of N-cadherin on the polarity distributions of PI3K and pMLC was similar to that of integrin. However, inhibition of the mechanical adhesion of N-cadherin led to inhibition of the polarity intensity and polarity angle distribution of PI3K and pMLC proteins in the contact group. Furthermore, inhibition of the mechanical adhesion of N-cadherin caused weakened polarity intensity of integrin β1, reducing the proportion of cells with polarity angles between integrin β1 and β-catenin concentrating in the range of 135° to 180°. Additionally, intracellular forces influenced the polar distribution of PI3K and pMLC proteins. Reducing intracellular forces weakened the polarity intensity of PI3K and pMLC proteins and their polarity distribution, while increasing intracellular forces enhanced the polarity intensity of PI3K and pMLC proteins and their polarity distribution.
    UNASSIGNED: Integrin and N-cadherin co-regulate the polarity distribution of cell proteins and N-cadherin can play an important role in the polarity regulation of stem cells through local inhibition of integrin.
    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
    紧密连接蛋白相关的血脑屏障(BBB)损伤在缺血性卒中的发病机制中起重要作用。利贝特,胆碱磷酸转移酶(CPT)的抑制剂,已被用作降血脂剂。然而,利贝特在缺血性卒中中的保护作用及其潜在机制尚未明确阐明.这里,我们采用了MCAO的体内小鼠模型和体外OGD/R模型。在小鼠模型中,评估神经功能缺损评分和梗死体积.使用伊文思蓝溶液检测BBB通透性。检查TEER以确定脑内皮单层通透性。这里,我们发现,利贝特可改善卒中患者的神经功能障碍.此外,利贝特可显著改善卒中期间BBB通透性的增加。相应地,在mRNA和蛋白质水平上,Liberpate恢复了紧密连接蛋白ZO-1的表达降低.使用体外模型,我们发现,利贝特通过增加细胞活力但减少LDH的释放,改善了OGD/R诱导的人bEnd.3脑微血管内皮细胞损伤。重要的是,通过挽救ZO-1表达,利贝特抑制了OGD/R诱导的内皮单层通透性增加和TEER降低。机械上,在OGD/R刺激的bEnd.3细胞中,利贝特阻断MLCK/p-MLC信号通路的激活。相比之下,MLCK的过度表达消除了Liberate在内皮单层通透性中的保护作用,TEER,以及ZO-1的表达。我们的结果为进一步研究利贝特在卒中的神经保护机制提供了基础。
    Dysfunction of tight junction proteins-associated damage to the blood-brain barrier (BBB) plays an important role in the pathogenesis of ischemic stroke. Lifibrate, an inhibitor of cholinephosphotransferase (CPT), has been used as an agent for serum lipid lowering. However, the protective effects of Lifibrate in ischemic stroke and the underlying mechanism have not been clearly elucidated. Here, we employed an in vivo mice model of MCAO and an OGD/R model in vitro. In the mice models, neurological deficit scores and infarct volume were assessed. Evans Blue solution was used to detect the BBB permeability. The TEER was examined to determine brain endothelial monolayer permeability. Here, we found that Lifibrate improved neurological dysfunction in stroke. Additionally, increased BBB permeability during stroke was significantly ameliorated by Lifibrate. Correspondingly, the reduced expression of the tight junction protein ZO-1 was restored by Lifibrate at both the mRNA and protein levels. Using an in vitro model, we found that Lifibrate ameliorated OGD/R-induced injury in human bEnd.3 brain microvascular endothelial cells by increasing cell viability but reducing the release of LDH. Importantly, Lifibrate suppressed the increase in endothelial monolayer permeability and the reduction in TEER induced by OGD/R via the rescue of ZO-1 expression. Mechanistically, Lifibrate blocked activation of the MLCK/ p-MLC signaling pathway in OGD/R-stimulated bEnd.3 cells. In contrast, overexpression of MLCK abolished the protective effects of Lifibrate in endothelial monolayer permeability, TEER, as well as the expression of ZO-1. Our results provide a basis for further investigation into the neuroprotective mechanism of Lifibrate during 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
    肠道致病菌肠道沙门氏菌侵入肠道上皮细胞后迅速进入血液,但沙门氏菌是如何突破肠道血管屏障的,在很大程度上是未知的。这里,我们报道沙门氏菌在感染早期通过肠道CX3CR1+巨噬细胞进入血液。机械上,沙门氏菌以依赖于宿主细胞肌动蛋白和病原体效应物SteC的方式诱导巨噬细胞的迁移/侵袭特性。SteC募集宿主肌球蛋白轻链蛋白Myl12a并磷酸化其Ser19和Thr20残基。Myl12a磷酸化导致肌动蛋白重排,增强巨噬细胞的迁移和侵袭。SteC能够利用除ATP以外的多种NTP来磷酸化Myl12a。我们进一步解决了SteC的晶体结构,这表明了非典型的二聚化介导的催化机理。最后,体内数据表明,SteC介导的细胞骨架操作对于沙门氏菌突破肠道血管屏障并扩散到靶器官至关重要。
    The intestinal pathogen Salmonella enterica rapidly enters the bloodstream after the invasion of intestinal epithelial cells, but how Salmonella breaks through the gut-vascular barrier is largely unknown. Here, we report that Salmonella enters the bloodstream through intestinal CX3CR1+ macrophages during early infection. Mechanistically, Salmonella induces the migration/invasion properties of macrophages in a manner dependent on host cell actin and on the pathogen effector SteC. SteC recruits host myosin light chain protein Myl12a and phosphorylates its Ser19 and Thr20 residues. Myl12a phosphorylation results in actin rearrangement, and enhanced migration and invasion of macrophages. SteC is able to utilize a wide range of NTPs other than ATP to phosphorylate Myl12a. We further solved the crystal structure of SteC, which suggests an atypical dimerization-mediated catalytic mechanism. Finally, in vivo data show that SteC-mediated cytoskeleton manipulation is crucial for Salmonella breaching the gut vascular barrier and spreading to target organs.
    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
    背景:血小板中的Kindlin-3在支持整合素αIIbβ3活化中起重要作用,血小板扩散,聚合,和凝块通过与整联蛋白β3细胞质尾结合而缩回。然而,kindlin-3介导血小板中整合素αIIbβ3与肌球蛋白串扰的机制尚不清楚。
    目的:研究Myl6在支持血小板中整合素αIIbβ3活化中的作用。
    方法:Myl6fl/flPF4-Cre小鼠,巨核细胞谱系缺乏Myl6,产生了,并分析了Myl6缺陷型血小板中整合素αIIbβ3的激活。
    结果:我们鉴定了一种新的kindlin-3结合蛋白,Myl6,血小板中肌球蛋白的必需轻链。Myl6fl/flPF4-Cre小鼠表现出由前血小板形成缺陷引起的显著大血小板减少症。在没有Myl6的情况下,血小板中整合素αIIbβ3的激活被显著抑制,血小板聚集明显受损。有趣的是,与单价配体相比,血小板中Myl6的缺乏优先影响多价配体与整合素αIIbβ3的活化结合,表明Myl6可能通过与kindlin-3结合而有助于整合素αIIbβ3的亲合力调节。此外,Myl6fl/flPF4-Cre小鼠的凝血能力受损,并且始终如一,这些小鼠表现出止血和血栓形成功能的缺陷。
    结论:总之,这些结果表明,Myl6作为一种新型的kindlin-3结合伴侣,需要支持血小板中整合素αIIbβ3的活化,在止血和血栓形成中起重要作用。
    BACKGROUND: Kindlin-3 in platelets plays an essential role in supporting integrin αIIbβ3 activation, platelet spreading, aggregation, and clot retraction by binding to the integrin β3 cytoplasmic tail. However, the mechanism by which kindlin-3 mediates the crosstalk between integrin αIIbβ3 and myosin in platelets remains unknown.
    OBJECTIVE: To examine the role of myosin light chain 6 (Myl6) in supporting integrin αIIbβ3 activation in platelets.
    METHODS: Myl6fl/flPF4-Cre mice with a deficiency of Myl6 in the megakaryocyte lineage were generated, and integrin αIIbβ3 activation in Myl6-deficient platelets was analyzed.
    RESULTS: We identified a novel kindlin-3 binding protein, Myl6, an essential light chain of myosin in platelets. Myl6fl/flPF4-Cre mice exhibited significant macrothrombocytopenia resulting from defective proplatelet formation. In the absence of Myl6, integrin αIIbβ3 activation in platelets was significantly suppressed, and platelet aggregation was substantially impaired. Interestingly, the deficiency of Myl6 in platelets preferentially affected the binding of a multivalent ligand compared to a monovalent ligand to integrin αIIbβ3 upon activation, indicating that Myl6 may contribute to the avidity modulation of integrin αIIbβ3 by binding to kindlin-3. Furthermore, blood coagulation ability was impaired in Myl6fl/flPF4-Cre mice, and consistently, these mice exhibited defects in both hemostatic and thrombotic functions.
    CONCLUSIONS: In summary, these results suggest that Myl6, as a novel kindlin-3 binding partner, is required to support integrin αIIbβ3 activation in platelets, which plays an important role in both hemostasis and thrombosis.
    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
    我们研究了虾肽QMDDQ在小肠中的吸收机制,探索其抑制神经元过度活动的生理功能,并验证其在体内进入大脑以显示功能活动。采用大鼠外翻囊模型和Caco-2细胞旁吸收单层模型,表明QMDDQ具有良好的吸收能力,表观渗透系数(Papp)>1×10-6cm/s,QMDDQ的吸收与浓度有关。当QMDDQ的浓度为1mM,转运时间为180min时,QMDDQ的最高吸收浓度为41.17±3.48μM(P<0.05)。肌球蛋白轻链激酶(MLCK)特异性抑制剂ML-7和激活剂MPA,西方印迹,免疫荧光结果显示QMDDQ吸收是通过介导MLCK-p-MLCK-MLC信号通路发生的,可逆地打开小带闭塞-1(ZO-1),紧密连接(TJ)中的occludin,上调claudin-2表达,并通过血液到达目标以抑制神经元过度活动。体内荧光成像结果证实QMDDQ可在口服给药后4小时进入大脑。研究结果为探讨活性肽的细胞旁吸收机制提供了理论基础,也为开发干预阿尔茨海默病的功能性食品提供了起点。
    We investigated the absorption mechanism of the shrimp peptide QMDDQ in small intestines, explored its physiological function in inhibiting neuronal hyperactivity, and verified its entry into the brain in vivo to display functional activity. The everted rat sac model and a Caco-2 paracellular absorption monolayer model were used, indicating that QMDDQ has a good absorption capacity with an apparent permeability coefficient (Papp) > 1 × 10-6 cm/s and the absorption of QMDDQ was concentration-dependent. When the concentration of QMDDQ was 1 mM and the transport time was 180 min, the highest absorption concentration of QMDDQ was 41.17 ± 3.48 μM (P < 0.05). The myosin light-chain kinase (MLCK)-specific inhibitor ML-7 and activator MPA, Western blotting, and immunofluorescence results showed that QMDDQ absorption takes place by mediating the MLCK-p-MLCK-MLC signaling pathway, reversibly opening the zonula occludens-1 (ZO-1), occludin in tight junctions (TJs), upregulating claudin-2 expression, and reaching targets through blood to inhibit neuronal overactivity. Results of fluorescence imaging in vivo verified that QMDDQ could enter the brain 4 h after oral administration. The results provide a theoretical foundation for the mechanism of paracellular absorption of active peptides and a starting point for the development of functional foods for Alzheimer\'s disease intervention.
    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
    便秘是一种常见的胃肠道疾病,其患病率呈逐年上升趋势。便秘的发生涉及许多因素,如平滑肌收缩异常和胃肠激素分泌紊乱。砂仁(A.villosum)已被证明可有效改善消化系统疾病,但是没有关于改善便秘的报道。因此,采用网络药理学预测与动物实验相结合的方法,探讨了山葵的关键活性成分及其药理机制。网络药理预测结果表明,β-谷甾醇是紫草的关键泻药化合物,可能通过激活肾上腺素受体α1A-肌球蛋白轻链(ADRA1A-MLC)途径发挥泻药作用。进一步的动物实验表明,β-谷甾醇可显著缩短首次黑便时间;增加粪便重量,粪便数量,和粪便水含量;并促进胃肠蠕动。β-谷甾醇可能通过上调ADRA1A和肌球蛋白轻链9(Myl9)mRNA和蛋白在结肠中的表达来促进肠道运动。从而激活ADRA1A-MLC信号通路。此外,有可能通过调节血清神经递质和胃肠动力相关因子来改善便秘症状,如血清中5-羟色胺(5-HT)和乙酰胆碱酯酶(AchE)的含量和5-羟色胺受体4(5-HT4)的mRNA表达,干细胞因子(SCF),结肠干细胞因子受体(c-Kit)和平滑肌肌球蛋白轻链激酶(SMLCK)。本研究结果为山葵和β-谷甾醇在便秘中的应用奠定了基础。
    Constipation is a prevalent gastrointestinal disorder, with its prevalence showing an annual upward trend. There are many factors involved in the occurrence of constipation, such as abnormal smooth muscle contraction and disorders of gastrointestinal hormone secretion. Amomum villosum (A. villosum) has been proven to be effective in improving digestive system diseases, but there is no report on improving constipation. Therefore, we used network pharmacology prediction combined with animal experiments to explore the key active components of A. villosum and their pharmacological mechanisms. The results of network pharmacological prediction showed that β-sitosterol was the key laxative compound of A. villosum, which may play a laxative role by activating the adrenoceptor alpha 1 A-myosin light chain (ADRA1A-MLC) pathway. Further animal experiments showed that β-sitosterol could significantly shorten the time to first black stool; increase faecal weight, faecal number, and faecal water content; and promote gastrointestinal motility. β-sitosterol may promote intestinal motility by upregulating the expression of ADRA1A and myosin light chain 9 (Myl9) mRNA and protein in the colon, thereby activating the ADRA1A-MLC signalling pathway. In addition, it is possible to improve constipation symptoms by regulating serum neurotransmitters and gastrointestinal motility-related factors, such as the serum content of 5-hydroxytryptamine (5-HT) and acetylcholinesterase (AchE) and the mRNA expression of 5-hydroxytryptamine receptor 4 (5-HT4), stem cell factor (SCF), stem cell factor receptor (c-Kit) and smooth muscle myosin light chain kinase (smMLCK) in the colon. These results lay a foundation for the application of A. villosum and β-sitosterol in constipation.
    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
    脊髓损伤(SCI)可以立即破坏血液脊髓屏障(BSCB)。恢复该屏障的完整性对于SCI后神经功能的恢复至关重要。UTX蛋白,组蛋白去甲基酶,在先前的研究中已显示出促进SCI小鼠的血管再生和神经恢复。然而,目前尚不清楚UTX敲除是否可以通过降低BSCB的渗透性来促进BSCB的恢复。在这项研究中,我们系统地研究了SCI后不同时间点的BSCB破坏和通透性,发现内皮细胞(ECs)中条件UTX缺失可以降低BSCB通透性,减少炎症细胞浸润和ROS产生,改善SCI后神经功能恢复。随后,我们使用RNA测序和ChIP-qPCR证实条件UTX敲除ECs可以下调肌球蛋白轻链激酶(MLCK)的表达,特异性介导肌球蛋白轻链(MLC)磷酸化并参与肌动蛋白收缩,细胞回缩,和紧密连接(TJs)蛋白质完整性。此外,我们发现MLCK过表达可以增加p-MLC/MLC的比例,进一步打破TJ,并加剧BSCB恶化。总的来说,我们的发现表明UTX敲除可以抑制MLCK/p-MLC途径,导致BSCB渗透率下降,并最终促进小鼠的神经系统恢复。这些结果表明UTX是治疗SCI的有希望的新靶标。
    Spinal cord injury (SCI) can prompt an immediate disruption to the blood-spinal cord barrier (BSCB). Restoring the integrity of this barrier is vital for the recovery of neurological function post-SCI. The UTX protein, a histone demethylase, has been shown in previous research to promote vascular regeneration and neurological recovery in mice with SCI. However, it is unclear whether UTX knockout could facilitate the recovery of the BSCB by reducing its permeability. In this study, we systematically studied BSCB disruption and permeability at different time points after SCI and found that conditional UTX deletion in endothelial cells (ECs) can reduce BSCB permeability, decrease inflammatory cell infiltration and ROS production, and improve neurological function recovery after SCI. Subsequently, we used RNA sequencing and ChIP-qPCR to confirm that conditional UTX knockout in ECs can down-regulate expression of myosin light chain kinase (MLCK), which specifically mediates myosin light chain (MLC) phosphorylation and is involved in actin contraction, cell retraction, and tight junctions (TJs) protein integrity. Moreover, we found that MLCK overexpression can increase the ratio of p-MLC/MLC, further break TJs, and exacerbate BSCB deterioration. Overall, our findings indicate that UTX knockout could inhibit the MLCK/p-MLC pathway, resulting in decreased BSCB permeability, and ultimately promoting neurological recovery in mice. These results suggest that UTX is a promising new target for treating SCI.
    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:本研究探讨宫颈鳞状细胞癌(SCC)进展的机制。
    方法:采用逆转录-定量聚合酶链反应和免疫印迹法检测肌球蛋白轻链9(MYL9)在SCC组织和细胞系中的表达。此外,Transwell和Boyden测定用于评估MYL9在SCC进展中的功能。此外,通过乳酸水平和有氧糖酵解来探索SCC中MYL9的详细机制。
    结果:SCC组织中MYL9的mRNA和蛋白水平升高,MYL9敲除抑制SCC细胞系的迁移和侵袭。一项机制研究表明,MYL9通过增强有氧糖酵解和增加Janus激酶2(JAK2)/信号转导和转录激活因子3(STAT3)途径的活性来促进SCC的迁移和侵袭。
    结论:MYL9在SCC中上调,它增强了JAK2/STAT3通路的活性,促进了SCC的转移和糖酵解。
    OBJECTIVE: This study explored the mechanism of squamous cervical cancer (SCC) progression.
    METHODS: Reverse transcription-quantitative polymerase chain reaction and western blotting were used to evaluate the expression of myosin light chain 9 (MYL9) in SCC tissues and cell lines. Furthermore, Transwell and Boyden assays were used to assess the function of MYL9 in SCC progression. In addition, the levels of lactate and aerobic glycolysis were used to explore the detailed mechanism of MYL9 in SCC.
    RESULTS: The mRNA and protein levels of MYL9 were elevated in SCC tissues, and MYL9 knockdown inhibited the migration and invasion of SCC cell lines. A mechanistic study demonstrated that MYL9 promotes SCC migration and invasion by enhancing aerobic glycolysis and increasing the activity of the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway.
    CONCLUSIONS: MYL9 was upregulated in SCC, and it enhanced JAK2/STAT3 pathway activity and promoted metastasis and glycolysis in SCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号