HMGB1

HMGB1
  • 文章类型: Journal Article
    甘草(甘草)是豆科/豆科中甘草属的一种植物,是一种著名的天然草本植物,具有悠久的药用历史。甘草酸(GLY),甘草的主要活性成分,作为临床实践中广泛使用的治疗剂。GLY表现出不同的药用特性,包括消炎药,抗菌,抗病毒,抗肿瘤,免疫调节,肠道环境维护,和肝脏保护作用。然而,目前的研究主要强调GLY的抗病毒活性,同时对其抗菌性能提供有限的见解。GLY通过靶向细菌酶抑制细菌的生长,证明了广谱的抗菌活性,影响细胞膜的形成,改变膜的通透性。此外,GLY还可以通过激活相关的免疫途径来增强宿主免疫力,从而增强病原体清除。本文综述了GLY对各种病原菌引起的病理变化的抑制机制。它作为高迁移率族蛋白1抑制剂在免疫调节中的作用,及其在对抗病原菌引起的疾病方面的功效。此外,GLY与其他抗生素联合使用可降低最小抑制浓度,可能有助于临床开发针对耐药细菌的联合疗法。使用PubMed搜索信息来源,WebofScience,科学直接,和GreenMedical的关键词“甘草”,\"甘草酸\",“抗菌”,“抗炎”,\"HMGB1\",及其组合,主要来自1979年至2024年发表的文章,没有语言限制。筛选由一位作者进行,并由其他人补充。在实验设计中存在实验缺陷的论文和未达到预期的论文(抗真菌论文,等。)被排除在外。
    Licorice (Glycyrrhiza glabra) is a plant of the genus Glycyrrhiza in the family Fabaceae/Leguminosae and is a renowned natural herb with a long history of medicinal use dating back to ancient times. Glycyrrhizin (GLY), the main active component of licorice, serves as a widely utilized therapeutic agent in clinical practice. GLY exhibits diverse medicinal properties, including anti-inflammatory, antibacterial, antiviral, antitumor, immunomodulatory, intestinal environment maintenance, and liver protection effects. However, current research primarily emphasizes GLY\'s antiviral activity, while providing limited insight into its antibacterial properties. GLY demonstrates a broad spectrum of antibacterial activity via inhibiting the growth of bacteria by targeting bacterial enzymes, impacting cell membrane formation, and altering membrane permeability. Moreover, GLY can also bolster host immunity by activating pertinent immune pathways, thereby enhancing pathogen clearance. This paper reviews GLY\'s inhibitory mechanisms against various pathogenic bacteria-induced pathological changes, its role as a high-mobility group box 1 inhibitor in immune regulation, and its efficacy in combating diseases caused by pathogenic bacteria. Furthermore, combining GLY with other antibiotics reduces the minimum inhibitory concentration, potentially aiding in the clinical development of combination therapies against drug-resistant bacteria. Sources of information were searched using PubMed, Web of Science, Science Direct, and GreenMedical for the keywords \"licorice\", \"Glycyrrhizin\", \"antibacterial\", \"anti-inflammatory\", \"HMGB1\", and combinations thereof, mainly from articles published from 1979 to 2024, with no language restrictions. Screening was carried out by one author and supplemented by others. Papers with experimental flaws in their experimental design and papers that did not meet expectations (antifungal papers, etc.) were excluded.
    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
    硬膜外和硬膜下血肿通常与创伤性脑损伤有关。虽然手术切除是这些血肿的主要干预措施,预防和减少创伤后癫痫等并发症也至关重要,这可能是由受损大脑区域的炎症反应引起的。在本研究中,我们观察到大鼠硬膜外血肿(EDH)下的受伤脑区高迁移率组box-1(HMGB1)降低,同时血浆HMGB1水平升高。抗HMGB1单克隆抗体疗法强烈抑制HMGB1释放和随后的血浆水平增加。此外,这种治疗抑制了炎症细胞因子和相关分子如白细胞介素-1β(IL-1β)的上调,肿瘤坏死因子-α(TNF-α),和诱导型一氧化氮合酶(iNOS)在受伤区域。我们使用SH-SY5Y进行的体外实验表明,血肿成分-凝血酶,血红素,亚铁离子促使HMGB1从细胞核转位到细胞质,通过添加抗HMGB1mAb抑制的过程。这些发现表明,抗HMGB1mAb治疗不仅抑制HMGB1易位,而且抑制损伤区域的炎症。从而保护神经组织。因此,抗HMGB1mAb治疗可作为EDH手术前后的补充治疗.
    Epidural and subdural hematomas are commonly associated with traumatic brain injury. While surgical removal is the primary intervention for these hematomas, it is also critical to prevent and reduce complications such as post-traumatic epilepsy, which may result from inflammatory responses in the injured brain areas. In the present study, we observed that high mobility group box-1 (HMGB1) decreased in the injured brain area beneath the epidural hematoma (EDH) in rats, concurrent with elevated plasma levels of HMGB1. Anti-HMGB1 monoclonal antibody therapy strongly inhibited both HMGB1 release and the subsequent increase in plasma levels. Moreover, this treatment suppressed the up-regulation of inflammatory cytokines and related molecules such as interleukin-1-beta (IL-1β), tumor necrosis factor-alpha (TNF-α), and inducible nitric oxide synthase (iNOS) in the injured areas. Our in vitro experiments using SH-SY5Y demonstrated that hematoma components-thrombin, heme, and ferrous ion- prompted HMGB1 translocation from the nuclei to the cytoplasm, a process inhibited by the addition of the anti-HMGB1 mAb. These findings suggest that anti-HMGB1 mAb treatment not only inhibits HMGB1 translocation but also curtails inflammation in injured areas, thereby protecting the neural tissue. Thus, anti-HMGB1 mAb therapy could serve as a complementary therapy for an EDH before/after surgery.
    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:新发现的CircUBE2D2已被证明在多种癌症中异常上调并促进癌症进展。本研究探讨了cirUBE2D2(hsa_circ_0005728)在卵巢癌(OC)进展中的作用。
    方法:CircUBE2D2,miR-885-5p,通过RT-qPCR或WB检测HMGB1。SKOV-3细胞功能(包括细胞活力,凋亡,迁移,和侵袭)使用CCK-8、流式细胞术、划痕试验,和transwell分析,分别。miR-885-5p与circUBE2D2或HMGB1之间的直接关系通过双荧光素酶报告基因和RNA下拉分析得到证实。进一步探讨了cirUBE2D2在体内肿瘤异种移植实验中的作用。
    结果:OC组织和细胞系具有较高的circUBE2D2和HMGB1和较低的miR-885-5p。机械上,CircUBE2D2与miR-885-5p共享结合关系,而miR-885-5p可以直接靶向HMGB1。消除circUBE2D2或miR-885-5p诱导抑制OC细胞活性。然而,通过下调miR-885-5p或HMGB1诱导,这些功能得以缓解.此外,cirUBE2D2敲除降低肿瘤生长。
    结论:CircUBE2D2通过作为miR-885-5p的ceRNA海绵来调节HMGB1的表达,从而促进控制OC细胞的增殖和迁移,抑制细胞凋亡。靶向CircUBE2D2可以作为OC的新的潜在治疗策略。
    The newly discovered CircUBE2D2 has been shown to abnormally upregulate and promote cancer progression in a variety of cancers. The present study explored circUBE2D2 (hsa_circ_0005728) in Ovarian Cancer (OC) progression.
    CircUBE2D2, miR-885-5p, and HMGB1 were examined by RT-qPCR or WB. SKOV-3 cell functions (including cell viability, apoptosis, migration, and invasion) were validated using the CCK-8, flow cytometry, scratch assay, and transwell assay, respectively. The direct relationship between miR-885-5p and circUBE2D2 or HMGB1 was confirmed by a dual-luciferase reporter and RNA pull-down analysis. circUBE2D2\'s role in vivo tumor xenograft experiment was further probed.
    OC tissue and cell lines had higher circUBE2D2 and HMGB1 and lower miR-885-5p. Mechanically, CircUBE2D2 shared a binding relation with miR-885-5p, while miR-885-5p can directly target HMGB1. Eliminating circUBE2D2 or miR-885-5p induction inhibited OC cell activities. However, these functions were relieved by down-regulating miR-885-5p or HMGB1 induction. Furthermore, circUBE2D2 knockout reduced tumor growth.
    CircUBE2D2 regulates the expression of HMGB1 by acting as a sponge of ceRNA as miR-885-5p, thereby promoting the control of OC cell proliferation and migration and inhibiting cell apoptosis. Targeting CircUBE2D2 could serve as a new potential treatment strategy for OC.
    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
    高迁移率族蛋白1(HMGB1)在脓毒症中的促炎特性已被广泛研究。本研究旨在探讨HMGB1对脓毒症诱导的急性肺损伤(ALI)铁凋亡的影响及其分子机制。采用盲肠结扎穿孔法建立脓毒症小鼠模型。阻断HMGB1可提高生存率,减少肺损伤,铁凋亡标志物水平降低(活性氧,丙二醛,和Fe2+),并增强了败血症小鼠的抗氧化酶活性(超氧化物歧化酶和过氧化氢酶)。此外,HMGB1的敲除降低了细胞通透性,铁性凋亡标记,并提高了脂多糖(LPS)刺激的MLE-12细胞中的抗氧化酶水平。HMGB1的沉默导致LPS处理的MLE-12细胞中铁凋亡核心调节因子的表达升高,如溶质载体家族7成员11(SLC7A11),溶质载体家族3成员A2(SLC3A2),和谷胱甘肽过氧化物酶4.此外,阻断HMGB1不会改变铁凋亡,氧化应激相关的变化,和在LPS处理的MLE-12细胞中的通透性,这些细胞用铁抑素-1(铁凋亡抑制剂)预处理。抑制HMGB1还导致核因子红系2相关因子2(Nrf2)及其下游靶标的表达升高,血红素加氧酶-1(HO-1)和NAD(P)H:醌氧化还原酶1(NQO1)在LPS处理的MLE-12细胞和脓毒症小鼠的肺组织中。Nrf2特异性抑制剂ML385逆转了HMGB1沉默对LPS处理的MLE-12细胞中的铁凋亡和细胞通透性的影响。我们的发现表明,抑制HMGB1抑制铁凋亡和氧化应激,从而通过激活Nrf2信号来减轻脓毒症诱导的ALI。
    The proinflammatory properties of high-mobility group box protein 1 (HMGB1) in sepsis have been extensively studied. This study aimed to investigate the impact of HMGB1 on ferroptosis and its molecular mechanism in sepsis-induced acute lung injury (ALI). A septic mouse model was established using the cecal ligation and puncture method. Blocking HMGB1 resulted in improved survival rates, reduced lung injury, decreased levels of ferroptosis markers (reactive oxygen species, malondialdehyde, and Fe2+), and enhanced antioxidant enzyme activities (superoxide dismutase and catalase) in septic mice. In addition, knockdown of HMGB1 reduced cellular permeability, ferroptosis markers, and raised antioxidant enzyme levels in lipopolysaccharide (LPS)-stimulated MLE-12 cells. Silencing of HMGB1 led to elevations in the expressions of ferroptosis core-regulators in LPS-treated MLE-12 cells, such as solute carrier family 7 member 11 (SLC7A11), solute carrier family 3 member A2 (SLC3A2), and glutathione peroxidase 4. Furthermore, blocking HMGB1 did not alter ferroptosis, oxidative stress-related changes, and permeability in LPS-treated MLE-12 cells that were pretreated with ferrostatin-1 (a ferroptosis inhibitor). HMGB1 inhibition also led to elevated expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream targets, heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) in LPS-treated MLE-12 cells and lung tissues from septic mice. The Nrf2-specific inhibitor ML385 reversed the effects of HMGB1 silencing on ferroptosis and cell permeability in LPS-treated MLE-12 cells. Our findings indicated that the inhibition of HMGB1 restrains ferroptosis and oxidative stress, thereby alleviating sepsis-induced ALI through the activation of Nrf2 signaling.
    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
    七氟醚暴露可导致神经毒性,尤其是在儿童中,这仍然是手术后的重要并发症。然而,其相关机制尚不清楚。这里,我们研究了SHARPIN在七氟醚诱导的神经毒性中的生物学作用.通过qPCR检测,蛋白质印迹和免疫组织化学染色,与对照小鼠相比,七氟醚刺激的小鼠SHARPIN和HMGB1表达升高。SHARPIN耗竭减轻海马损伤,抑制HMGB1和M1样巨噬细胞标志物的表达(iNOS,TNF-α,IL-1β,IL-6),但增强了M2样巨噬细胞标志物(ARG-1,IL-10)的表达。GST下拉和Co-IP测定表明SHARPIN与HMGB1直接相互作用以增强SH-SY5Y细胞中的HMGB1表达。SHARPIN沉默对炎症反应和M1样巨噬细胞的抑制作用被HMGB1过表达所抵消。最后,SHARPIN-HMGB1通路通过调节巨噬细胞极化影响七氟醚引发的神经炎症。总的来说,我们的数据表明,SHARPIN通过增强HMGB1表达,将M2样巨噬细胞转化为M1样巨噬细胞,从而刺激七氟醚诱导的神经毒性.SHARPIN干预可能是减轻七氟醚诱导的神经毒性的有希望的治疗方法。
    Sevoflurane exposure can result in neurotoxicity especially among children, which remains an important complication after surgery. However, its related mechanisms remain unclear. Here, we investigated the biological roles of SHARPIN in sevoflurane-induced neurotoxicity. As detected by qPCR, Western blotting and immunohistochemical staining, SHARPIN and HMGB1 expression was elevated in sevoflurane-stimulated mice as compared with the control mice. SHARPIN depletion attenuated hippocampus injury, repressed the expression of HMGB1 and M1-like macrophage markers (iNOS, TNF-α, IL-1β, IL-6), but enhanced the expression of M2-like macrophage markers (ARG-1, IL-10). GST pull-down and Co-IP assays demonstrated that SHARPIN directly interacted with HMGB1 to enhance HMGB1 expression in SH-SY5Y cells. The inhibitory effects of SHARPIN silencing on inflammatory reaction and M1-like macrophages were counteracted by HMGB1 overexpression. Finally, SHARPIN-HMGB1 pathway affected neuroinflammation triggered by sevoflurane via modulating macrophage polarization. Collectively, our data suggested that SHARPIN stimulated sevoflurane-induced neurotoxicity via converting M2-like macrophages to M1-like macrophages by enhancing HMGB1 expression. SHARPIN intervention may be a promising therapeutic method to relieve sevoflurane-induced neurotoxicity.
    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
    最近在一些临床病例中报道了SemenStrychni的神经毒性。因此,本研究旨在探讨HMGB1在马钱子神经毒性模型中的作用,并评估甘草酸(GA)的潜在缓解作用。这与HMGB1释放的调节有关。48只SD大鼠腹腔注射马钱子提取物(175mg/kg),然后口服GA(50mg/kg)4天。SS和GA治疗后,神经元变性,凋亡,通过组织病理学检查观察到坏死。炎性细胞因子(TNF-α和IL-1β),神经递质相关酶(MAO和AChE),血清HMGB1、核和胞浆HMGB1/ph-HMGB1与PP2A、PKC,和HMGB1进行评价。还检查了MAPK途径的影响。因此,这种神经毒性的特征是神经元变性和凋亡,促炎细胞因子的诱导,和神经递质代谢酶的减少。相比之下,GA治疗显著改善了上述效果并减轻了神经损伤。此外,Strychni促进HMGB1磷酸化及其在细胞核和细胞质之间的易位,从而激活NF-κB和MAPK通路,启动各种炎症反应。我们的实验表明,GA可以部分逆转这些影响。总之,GA酸缓解马钱子引起的神经毒性,可能通过抑制HMGB1磷酸化并阻止其从细胞中释放。
    The neurotoxicity of Semen Strychni has been reported recently in several clinical cases. Therefore, this study was conducted to investigate the role of HMGB1 in a model of neurotoxicity induced by Semen Strychni and to assess the potential alleviating effects of glycyrrhizic acid (GA), which is associated with the regulation of HMGB1 release. Forty-eight SD rats were intraperitoneally injected with Semen Strychni extract (175 mg/kg), followed by oral administration of GA (50 mg/kg) for four days. After treatment of SS and GA, neuronal degeneration, apoptosis, and necrosis were observed via histopathological examination. Inflammatory cytokines (TNF-α and IL-1β), neurotransmitter associated enzymes (MAO and AChE), serum HMGB1, nuclear and cytoplasmic HMGB1/ph-HMGB1, and the interaction between PP2A, PKC, and HMGB1 were evaluated. The influence of the MAPK pathway was also examined. As a result, this neurotoxicity was characterized by neuronal degeneration and apoptosis, the induction of pro-inflammatory cytokines, and a reduction in neurotransmitter-metabolizing enzymes. In contrast, GA treatment significantly ameliorated the abovementioned effects and alleviated nerve injury. Furthermore, Semen Strychni promoted HMGB1 phosphorylation and its translocation between the nucleus and cytoplasm, thereby activating the NF-κB and MAPK pathways, initiating various inflammatory responses. Our experiments demonstrated that GA could partially reverse these effects. In summary, GA acid alleviated Semen Strychni-induced neurotoxicity, possibly by inhibiting HMGB1 phosphorylation and preventing its release from the cell.
    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
    高迁移率族蛋白1(HMGB1)受输出蛋白1(XPO1)依赖的核输出,它参与了与免疫疗法抗性有关的功能。我们研究了HMGB1mRNA表达是否与非小细胞肺癌(NSCLC)对免疫检查点抑制剂(ICI)的反应相关。
    从用ICI治疗的晚期NSCLC患者的预处理活检中分离RNA。几个基因的基因表达分析,包括HMGB1,使用NanoString计数器分析系统(PanCancer免疫分析小组)进行。在EGFR和KRAS突变细胞系中进行蛋白质印迹分析和细胞活力测定。在鼠Lewis肺癌模型中确定ICI与XPO1阻断剂(selinexor)和曲美替尼组合的抗肿瘤作用的评估。
    接受ICI治疗的非小细胞肺癌患者的HMGB1mRNA水平与无进展生存期(PFS)相关(中位PFS9.0与18.0个月,P=0.008,高与低HMGB1的风险比=0.30)。TNF-α刺激后,HMGB1在PC9细胞的细胞质中积累,但是这种积累可以通过使用selinexor或抗逆转录病毒药物来预防。EGFR突变细胞中的厄洛替尼或奥希替尼联合selinexor和KRAS突变细胞中的曲美替尼联合selinexor消除肿瘤细胞增殖。Selinexor与PD-1抑制剂一起使用或不使用曲美替尼可以消除小鼠Lewis肺癌模型中的肿瘤生长。
    对HMGB1mRNA和蛋白功能的深入探索有望发现新的潜在靶点,为联合ICI治疗转移性NSCLC提供依据。
    UNASSIGNED: High-mobility group box 1 protein (HMGB1) is subject to exportin 1 (XPO1)-dependent nuclear export, and it is involved in functions implicated in resistance to immunotherapy. We investigated whether HMGB1 mRNA expression was associated with response to immune checkpoint inhibitors (ICI) in non-small cell lung cancer (NSCLC).
    UNASSIGNED: RNA was isolated from pretreatment biopsies of patients with advanced NSCLC treated with ICI. Gene expression analysis of several genes, including HMGB1, was conducted using the NanoString Counter analysis system (PanCancer Immune Profiling Panel). Western blotting analysis and cell viability assays in EGFR and KRAS mutant cell lines were carried out. Evaluation of the antitumoral effect of ICI in combination with XPO1 blocker (selinexor) and trametinib was determined in a murine Lewis lung carcinoma model.
    UNASSIGNED: HMGB1 mRNA levels in NSCLC patients treated with ICI correlated with progression-free survival (PFS) (median PFS 9.0 versus 18.0 months, P=0.008, hazard ratio=0.30 in high versus low HMGB1). After TNF-α stimulation, HMGB1 accumulates in the cytoplasm of PC9 cells, but this accumulation can be prevented by using selinexor or antiretroviral drugs. Erlotinib or osimertinib with selinexor in EGFR-mutant cells and trametinib plus selinexor in KRAS mutant abolish tumor cell proliferation. Selinexor with a PD-1 inhibitor with or without trametinib abrogates the tumor growth in the murine Lewis lung cancer model.
    UNASSIGNED: An in-depth exploration of the functions of HMGB1 mRNA and protein is expected to uncover new potential targets and provide a basis for treating metastatic NSCLC in combination with ICI.
    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
    在中国,中风的特点是发病率高,复发,残疾,和死亡率。rtPA的超早期给药对于恢复急性缺血性卒中的灌注至关重要,尽管它同时增加了出血性转化的风险。高迁移率族蛋白1(HMGB1)在脑缺血和缺血再灌注后的神经炎症中发挥关键作用。坏死细胞被动释放并主动分泌,免疫细胞将HMGB1直接分泌到细胞外空间,将HMGB1包装到细胞内囊泡中,胶质细胞,血小板,和内皮细胞,HMGB1代表典型的损伤相关分子模式(DAMP)。它与动脉粥样硬化的发病机理密切相关,血栓栓塞,和缺血性中风早期的有害炎症。此外,HMGB1对后期神经血管重构和功能恢复有重要作用。重要的是,HMGB1通过促进神经炎症介导出血性转化,直接损害血脑屏障的完整性,并通过与rtPA的相互作用增强MMP9的分泌。作为全身性炎症因子,HMGB1还与卒中后抑郁和卒中相关肺炎的风险升高有关。HMGB1的作用延伸到通过使免疫和神经胶质细胞的各种亚型极化来影响缺血的发病机理。这包括由于兴奋性氨基酸介导的兴奋性毒性,自噬,MMP9版本,NET形成,和自分泌营养途径。鉴于其多方面的作用,HMGB1是缺血性卒中和出血性转化的重要治疗靶点和预后标志物。在这次审查中,我们总结了结构和氧化还原性质,分泌和途径,免疫细胞活性的调节,病理生理机制在中风中的作用,和HMGB1的出血转化,这将为开发新的神经保护药物铺平道路,减少中风后神经炎症,扩大溶栓时间窗。
    Stroke in China is distinguished by its high rates of morbidity, recurrence, disability, and mortality. The ultra-early administration of rtPA is essential for restoring perfusion in acute ischemic stroke, though it concurrently elevates the risk of hemorrhagic transformation. High-mobility group box 1 (HMGB1) emerges as a pivotal player in neuroinflammation after brain ischemia and ischemia-reperfusion. Released passively by necrotic cells and actively secreted, including direct secretion of HMGB1 into the extracellular space and packaging of HMGB1 into intracellular vesicles by immune cells, glial cells, platelets, and endothelial cells, HMGB1 represents a prototypical damage-associated molecular pattern (DAMP). It is intricately involved in the pathogenesis of atherosclerosis, thromboembolism, and detrimental inflammation during the early phases of ischemic stroke. Moreover, HMGB1 significantly contributes to neurovascular remodeling and functional recovery in later stages. Significantly, HMGB1 mediates hemorrhagic transformation by facilitating neuroinflammation, directly compromising the integrity of the blood-brain barrier, and enhancing MMP9 secretion through its interaction with rtPA. As a systemic inflammatory factor, HMGB1 is also implicated in post-stroke depression and an elevated risk of stroke-associated pneumonia. The role of HMGB1 extends to influencing the pathogenesis of ischemia by polarizing various subtypes of immune and glial cells. This includes mediating excitotoxicity due to excitatory amino acids, autophagy, MMP9 release, NET formation, and autocrine trophic pathways. Given its multifaceted role, HMGB1 is recognized as a crucial therapeutic target and prognostic marker for ischemic stroke and hemorrhagic transformation. In this review, we summarize the structure and redox properties, secretion and pathways, regulation of immune cell activity, the role of pathophysiological mechanisms in stroke, and hemorrhage transformation for HMGB1, which will pave the way for developing new neuroprotective drugs, reduction of post-stroke neuroinflammation, and expansion of thrombolysis time window.
    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
    高迁移率族蛋白1(HMGB1)是一种核DNA结合蛋白,在癌症中具有双重作用,作为癌基因和肿瘤抑制因子。这种蛋白质调节核小体结构,DNA损伤修复,和细胞内的基因组稳定性,同时也在免疫细胞功能中发挥作用。本文综述了HMGB1在肿瘤中的生物学和临床意义。包括它参与细胞死亡和存活,它作为治疗靶点和癌症生物标志物的潜力,并作为暴露于细胞毒性抗癌治疗后剩余细胞的前生存信号。我们强调需要更好地了解HMGB1参与癌症的细胞标志物和机制,并旨在更深入地了解其在癌症进展中的作用。
    High mobility group box 1 (HMGB1) is a nuclear DNA-binding protein with a dual role in cancer, acting as an oncogene and a tumor suppressor. This protein regulates nucleosomal structure, DNA damage repair, and genomic stability within the cell, while also playing a role in immune cell functions. This review comprehensively evaluates the biological and clinical significance of HMGB1 in cancer, including its involvement in cell death and survival, its potential as a therapeutic target and cancer biomarker, and as a prosurvival signal for the remaining cells after exposure to cytotoxic anticancer treatments. We highlight the need for a better understanding of the cellular markers and mechanisms involved in the involvement of HMGB1in cancer, and aim to provide a deeper understanding of its role in cancer progression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    急性排斥反应是影响肝移植术后受者存活的重要因素。Salidroside有各种属性,包括消炎药,抗氧化剂,和保肝性能。本研究旨在探讨红景天苷是否可以预防肝移植后的急性排斥反应,并探讨其相关机制。在肝移植后用他克莫司(1mg/kg/d)或红景天苷(10或20mg/kg/d)预处理7天的大鼠中建立了体内急性排斥模型。此外,使用与红景天苷(1、10、50或100μM)孵育的嗜中性粒细胞进行体外实验。苏木精-伊红染色,末端脱氧核苷酸转移酶dUTP缺口末端标记染色,免疫吸附测定,免疫荧光分析,伊文思蓝染色,进行和western印迹分析以检查红景天苷在体外和体内对NET形成和急性排斥的影响。我们发现,红景天苷治疗减少病理性肝损伤,血清转氨酶水平,和血清IL-1β水平,体内IL-6和TNF-α。HMGB1/TLR-4/MAPK信号通路相关蛋白(HMGB1、TLR-4、p-ERK1/2、p-JNK、p-P38,cleavedcaspase-3,cleavedcaspase-9,Bcl-2,Bax,IL-1β,TNF-α,红景天苷治疗后,IL-6)也降低。体外实验表明,红景天苷降低了脂多糖处理的中性粒细胞释放HMGB1/TLR-4/MAPK信号通路相关蛋白的作用。此外,红景天苷通过调节HMGB1/TLR-4/MAPK信号通路抑制NETosis并保护急性排斥反应。此外,红景天苷联合他克莫司的治疗效果优于其他任何一种单独的治疗方法。总之,红景天苷可通过HMGB1/TLR-4/MAPK信号通路减少中性粒细胞胞外阱的发育,从而预防肝移植后急性肝排斥反应。
    Acute rejection is an important factor affecting the survival of recipients after liver transplantation. Salidroside has various properties, including anti-inflammatory, antioxidant, and hepatoprotective properties. This study aims to investigate whether salidroside can prevent acute rejection after liver transplantation and to examine the underlying mechanisms involved. An in vivo acute rejection model is established in rats that are pretreated with tacrolimus (1 mg/kg/d) or salidroside (10 or 20 mg/kg/d) for seven days after liver transplantation. In addition, an in vitro experiment is performed using neutrophils incubated with salidroside (1, 10, 50 or 100 μM). Hematoxylin-eosin staining, terminal deoxynucleotidyl transferase dUTP nick-end labeling staining, immunosorbent assays, immunofluorescence analysis, Evans blue staining, and western blot analysis are performed to examine the impact of salidroside on NET formation and acute rejection in vitro and in vivo. We find that Salidroside treatment reduces pathological liver damage, serum aminotransferase level, and serum levels of IL-1β, IL-6, and TNF-α in vivo. The expressions of proteins associated with the HMGB1/TLR-4/MAPK signaling pathway (HMGB1, TLR-4, p-ERK1/2, p-JNK, p-P38, cleaved caspase-3, cleaved caspase-9, Bcl-2, Bax, IL-1β, TNF-α, and IL-6) are also decreased after salidroside treatment. In vitro experiments show that the release of HMGB1/TLR-4/MAPK signaling pathway-associated proteins from neutrophils treated with lipopolysaccharide is decreased by salidroside. Moreover, salidroside inhibits NETosis and protects against acute rejection by regulating the HMGB1/TLR-4/MAPK signaling pathway. Furthermore, salidroside combined with tacrolimus has a better effect than either of the other treatments alone. In summary, salidroside can prevent acute liver rejection after liver transplantation by reducing neutrophil extracellular trap development through the HMGB1/TLR-4/MAPK signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号