Inflammatory damage

  • 文章类型: Journal Article
    背景:加味白虎汤(JWBHD),用七种中药材配制的处方在减轻中暑(HS)患者的脑损伤方面具有临床疗效。
    目的:本研究旨在通过整合网络药理学和药效学方法,评价JWBHD对HS大鼠模型的治疗效果,并探讨其治疗机制。使用UPLC-MS/MS分析了哪些主要成分。
    方法:首先进行网络药理学分析以预测JWBHD的潜在活性成分和治疗靶标。然后在经历HS的大鼠上评估JWBHD的抗HS有效性。收集大鼠脑组织进行一系列全面的实验,包括westernblot,PCR,H&E染色,尼氏染色,ELISA,透射电子显微镜,流式细胞术和免疫荧光法验证JWBHD对HS诱导的脑损伤的保护作用。此外,在HS攻击的F98细胞系上进一步验证了JWBHD对胶质细胞TLR4/NF-κB信号和线粒体自噬的抑制作用。最后,利用UPLC-MS/MS分析了JWBHD水提取物的化学成分。
    结果:网络药理学已经确定了50个核心靶标和许多HS相关信号通路作为JWBHD的潜在治疗靶标。蛋白质-蛋白质相互作用(PPI)和GO的分析表明JWBHD可以抑制HS诱导的炎症信号。在HS大鼠身上进行的实验中,JWBHD显著降低了核心温度,恢复血压,减轻神经功能缺损。此外,JWBHD下调了白细胞和单核细胞的计数,降低炎症细胞因子的水平,如IL-1β,外周血中IL-6和TNF-α,并抑制HS大鼠大脑皮质TLR4和NF-κB的表达。此外,JWBHD抑制大脑皮质细胞凋亡,减轻HS组大脑皮质损伤。相反,在HS大鼠的大脑皮层中观察到过度活跃的线粒体自噬。然而,JWBHD恢复了线粒体膜电位,并下调了包括Pink1,Parkin,LC3B和Tom20。JWBHD减少了Pink1和GFAP的共同定位,HS大鼠大脑皮层星形胶质细胞的特异性标记物。此外,在F98细胞中进一步证实了JWBHD对TLR4/NF-κB信号传导的抑制作用和过度激活的线粒体自噬。最后,UPLC-MS/MS分析表明,JWBHD的主要成分包括异甘草素,甘草苷,甘草酸二钾,人参皂苷Rb1,人参皂苷Re,等。结论:JWBHD通过抑制TLR4/NF-κB信号和胶质细胞的线粒体自噬来保护大鼠免受HS并预防HS诱导的大脑皮质损伤。
    BACKGROUND: Jiawei Bai-Hu-Decoction (JWBHD), a prescription formulated with seven traditional Chinese medicinal material has demonstrated clinical efficacy in mitigating brain injury among heat stroke (HS) patients.
    OBJECTIVE: This study aimed to evaluate the therapeutic efficacy of JWBHD on rat model of HS and to explore its therapeutic mechanisms by integrating network pharmacology and pharmacodynamic methodologies, which major components were analyzed by using UPLC-MS/MS.
    METHODS: The network pharmacology analysis was firstly conducted to predict the potential active ingredients and therapeutic targets of JWBHD. The anti-HS effectiveness of JWBHD was then evaluated on rats experienced HS. Rat brain tissues were harvested for a comprehensive array of experiments, including Western blot, PCR, H&E staining, Nissl staining, ELISA, transmission electron microscope, flow cytometry and immunofluorescence to validate the protective effects of JWBHD against HS-induced brain damage. Furthermore, the inhibitory effects of JWBHD on TLR4/NF-κB signal and mitophagy of glial were further verified on HS-challenged F98 cell line. Finally, the chemical compositions of the water extract of JWBHD were analyzed by using UPLC-MS/MS.
    RESULTS: Network pharmacology has identified fifty core targets and numerous HS-related signaling pathways as potential therapeutic targets of JWBHD. Analysis of protein-protein interaction (PPI) and GO suggests that JWBHD may suppress HS-induced inflammatory signals. In experiments conducted on HS-rats, JWBHD significantly reduced the core temperature, restored blood pressure and alleviated neurological defect. Furthermore, JWBHD downregulated the counts of white blood cells and monocytes, decreased the levels of inflammatory cytokines such as IL-1β, IL-6 and TNF-α in peripheral blood, and suppressed the expression of TLR4 and NF-κB in the cerebral cortex of HS-rats. Besides, JWBHD inhibited the apoptosis of cortical cells and mitigated the damage to the cerebral cortex in HS group. Conversely, overactive mitophagy was observed in the cerebral cortex of HS-rats. However, JWBHD restored the mitochondrial membrane potential and downregulated expressions of mitophagic proteins including Pink1, Parkin, LC3B and Tom20. JWBHD reduced the co-localization of Pink1 and GFAP, a specific marker of astrocytes in the cerebral cortex of HS-rats. In addition, the inhibitory effect of JWBHD on TLR4/NF-κB signaling and overactive mitophagy were further confirmed in F98 cells. Finally, UPLC-MS/MS analysis showed that the main components of JWBHD include isoliquiritigenin, liquiritin, dipotassium glycyrrhizinate, ginsenoside Rb1, ginsenoside Re, etc. CONCLUSIONS: JWBHD protected rats from HS and prevented HS-induced damage in the cerebral cortex by suppressing TLR4/NF-κB signaling and mitophagy of glial.
    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:肠道免疫失调与肿瘤的发生和形成密切相关。环指蛋白128(RNF128)已被鉴定为在先天和适应性系统中发挥不同的免疫调节功能。然而,RNF128在结肠炎和结直肠癌(CRC)等肠道炎症中的生理作用仍存在争议.
    目的:阐明RNF128在结肠炎和CRC中的作用和机制。
    方法:在WT和Rnf128缺陷小鼠中建立葡聚糖硫酸钠(DSS)诱导的结肠炎和氧化甲烷(AOM)/DSS诱导的CRC动物模型,并通过组织病理学进行评估。采用免疫共沉淀和泛素化分析来研究RNF128在IL-6-STAT3信号传导中的作用。
    结果:与配对的肿瘤周围组织相比,RNF128在临床CRC组织中显著下调。Rnf128缺陷型小鼠对DSS诱导的结肠炎和AOM/DSS或APC突变诱导的CRC均高度敏感。当IL-6刺激时,RNF128的缺失在体内和体外癌变的早期转化阶段促进了CRC细胞的增殖和STAT3的激活。机械上,RNF128与IL-6受体α亚基(IL-6Rα)和膜糖蛋白gp130相互作用,并以连接酶活性依赖性方式介导其溶酶体降解。通过一系列IL-6受体的点突变,我们发现RNF128在K398/K401促进IL-6Rα的K48连接多泛素化,在K718/K816/K866促进gp130。此外,阻断STAT3激活可有效根除Rnf128缺陷小鼠在癌变转化阶段的炎症损伤。
    结论:RNF128通过抑制IL-6-STAT3信号传导减弱结肠炎和结直肠肿瘤发生,这为IL-6受体的调节和炎症到癌症的转变提供了新的见解。
    BACKGROUND: Intestinal immune dysregulation is strongly linked to the occurrence and formation of tumors. RING finger protein 128 (RNF128) has been identified to play distinct immunoregulatory functions in innate and adaptive systems. However, the physiological roles of RNF128 in intestinal inflammatory conditions such as colitis and colorectal cancer (CRC) remain controversial.
    OBJECTIVE: To elucidate the function and mechanism of RNF128 in colitis and CRC.
    METHODS: Animal models of dextran sodium sulfate (DSS)-induced colitis and azoxymethane (AOM)/DSS-induced CRC were established in WT and Rnf128-deficient mice and evaluated by histopathology. Co-immunoprecipitation and ubiquitination analyses were employed to investigate the role of RNF128 in IL-6-STAT3 signaling.
    RESULTS: RNF128 was significantly downregulated in clinical CRC tissues compared with paired peritumoral tissues. Rnf128-deficient mice were hypersusceptible to both colitis induced by DSS and CRC induced by AOM/DSS or APC mutation. Loss of RNF128 promoted the proliferation of CRC cells and STAT3 activation during the early transformative stage of carcinogenesis in vivo and in vitro when stimulated by IL-6. Mechanistically, RNF128 interacted with the IL-6 receptor α subunit (IL-6Rα) and membrane glycoprotein gp130 and mediated their lysosomal degradation in ligase activity-dependent manner. Through a series of point mutations in the IL-6 receptor, we identified that RNF128 promoted K48-linked polyubiquitination of IL-6Rα at K398/K401 and gp130 at K718/K816/K866. Additionally, blocking STAT3 activation effectively eradicated the inflammatory damage of Rnf128-deficient mice during the transformative stage of carcinogenesis.
    CONCLUSIONS: RNF128 attenuates colitis and colorectal tumorigenesis by inhibiting IL-6-STAT3 signaling, which sheds novel insights into the modulation of IL-6 receptors and the inflammation-to-cancer transition.
    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
    苯醚环唑(DFZ),被归类为“低毒性农药”,\"近年来得到了广泛的应用。然而,物质的非目标毒性,特别是对水生生物,产生了相当大的忧虑。阿魏酸(FA)的抗炎和抗氧化作用在这种特殊情况下吸引了大量研究。本研究建立了DFZ的慢性暴露模型,并研究了FA对淡水鲤鱼慢性呼吸抑制导致g损伤的保护作用。通过HE染色的组织学分析表明,FA有效地减轻了由长期DFZ暴露引起的g组织损伤。qRT-PCR结果显示,添加FA降低了IL-1β的表达,IL-6和TNF-α同时增强IL-10和TGF-β1的表达。生化分析和DHE染色显示,FA降低MDA水平,增加CAT和GSH活性,还有T-AOC,响应于长期DFZ暴露的ROS积累减少。从Western印迹分析获得的结果表明,添加FA有效地抑制了长期暴露于DFZ的the中NF-κB信号传导途径和NLRP3炎性体途径的激活。总之,FA改善了暴露于慢性DFZ的鲤鱼的ill组织炎症并阻止了ROS的积累,通过NF-κB-NLRP3信号通路减轻组织炎症并恢复氧化还原稳态。因此,已经发现FA的应用对于改善呼吸抑制和减轻由水生生境中的DFZ污染引起的g组织炎症和氧化应激是有效的。
    Difenoconazole (DFZ), classified as a \"low-toxicity pesticide,\" has seen widespread application in recent years. Nevertheless, the non-target toxicity of the substance, particularly towards aquatic creatures, has generated considerable apprehension. The anti-inflammatory and antioxidant effects of Ferulic Acid (FA) have attracted considerable study in this particular setting. This study established a chronic exposure model to DFZ and investigated the protective effects of FA on chronic respiratory inhibition leading to gill damage in freshwater carp. Histological analyses via HE staining indicated that FA effectively alleviated gill tissue damage induced by chronic DFZ exposure. The qRT-PCR results showed that the addition of FA reduced the expression of IL-1β, IL-6 and TNF-α while boosting the expression of IL-10 and TGF-β1. Biochemical analyses and DHE staining revealed that FA reduced MDA levels and increased CAT and GSH activities, along with T-AOC, decreased ROS accumulation in response to chronic DFZ exposure. The results obtained from Western blotting analysis demonstrated that the addition of FA effectively suppressed the activation of the NF-κB signalling pathway and the NLRP3 inflammasome pathway in the gills subjected to prolonged exposure to DFZ. In summary, FA ameliorated gill tissue inflammation and blocked ROS accumulation in carp exposed to chronic DFZ, mitigating tissue inflammation and restoring redox homeostasis through the NF-κB-NLRP3 signaling pathway. Hence, the application of FA has been found to be efficacious for improving respiratory inhibition and mitigating gill tissue inflammation and oxidative stress resulting from DFZ pollution in aquatic habitats.
    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
    跟着二氧化锰纳米粒子(nanoMnO2)的普遍运用,健康危害也出现了。纳米MnO2可导致脑组织的炎症损伤,其机制尚不清楚。在这项研究中,我们旨在研究ROS介导的p38MAPK通路在纳米MnO2诱导的BV2小胶质细胞炎症反应中的作用。用2.5、5.0和10.0μg/mL纳米MnO2悬液处理BV2细胞12h,建立炎症损伤模型。活性氧(ROS)清除剂(20nMN-乙酰半胱氨酸,NAC)和p38MAPK途径抑制剂(10μMSB203580)用于阐明ROS和p38MAPK途径在纳米MnO2诱导的BV2细胞炎性病变中的作用。结果表明纳米MnO2增强了促炎细胞因子IL-1β和TNF-α的表达,BV2细胞内ROS水平升高并激活p38MAPK通路。用NAC控制细胞内ROS水平可以抑制p38MAPK通路的激活并减轻纳米MnO2诱导的炎症反应。此外,用SB203580抑制p38MAPK通路导致BV2细胞中炎症因子(IL-1β和TNF-α)的产生减少。总之,纳米MnO2可以通过增加细胞内ROS水平和进一步激活BV2小胶质细胞的p38MAPK通路来诱导炎症损伤。
    With the widespread use of manganese dioxide nanoparticles (nano MnO2), health hazards have also emerged. The inflammatory damage of brain tissues could result from nano MnO2, in which the underlying mechanism is still unclear. During this study, we aimed to investigate the role of ROS-mediated p38 MAPK pathway in nano MnO2-induced inflammatory response in BV2 microglial cells. The inflammatory injury model was established by treating BV2 cells with 2.5, 5.0, and 10.0 μg/mL nano MnO2 suspensions for 12 h. Then, the reactive oxygen species (ROS) scavenger (20 nM N-acetylcysteine, NAC) and the p38 MAPK pathway inhibitor (10 μM SB203580) were used to clarify the role of ROS and the p38 MAPK pathway in nano MnO2-induced inflammatory lesions in BV2 cells. The results indicated that nano MnO2 enhanced the expression of pro-inflammatory cytokines IL-1β and TNF-α, elevated intracellular ROS levels and activated the p38 MAPK pathway in BV2 cells. Controlling intracellular ROS levels with NAC inhibited p38 MAPK pathway activation and attenuated the inflammatory response induced by nano MnO2. Furthermore, inhibition of the p38 MAPK pathway with SB203580 led to a decrease in the production of inflammatory factors (IL-1β and TNF-α) in BV2 cells. In summary, nano MnO2 can induce inflammatory damage by increasing intracellular ROS levels and further activating the p38 MAPK pathway in BV2 microglial cells.
    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
    细颗粒物(PM2.5)可以通过多种途径进入人体,对人体健康产生不利影响。人的肺和眼睛长时间暴露在空气中,是最先暴露在PM2.5中的。“液体浸没暴露法”有一些局限性,使其无法充分反映颗粒物对人体的毒性作用。在这项研究中,对采集的PM2.5样品进行化学分析。基于人肺上皮细胞(A549)和永生化人角膜上皮细胞(HCE-T),建立了与体内环境高度相关的气液界面(ALI)模型。使用VITROCELLCloud12系统将PM2.5均匀地分布在细胞上。暴露6小时和24小时后,细胞活力,凋亡率,活性氧(ROS)水平,炎症因子的表达,和脱氧核糖核酸(DNA)损伤被测量。结果表明,PM2.5对细胞活力具有显著的剂量和时间依赖性。细胞凋亡,ROS生成,和ALI的DNA损伤,而炎症因子仅表现出剂量依赖性作用。值得注意的是,即使短时间暴露于低剂量的PM2.5也会导致细胞DNA双链断裂和γ-H2AX表达增加,表明PM2.5具有显著的遗传毒性。细胞中ROS丰度的增加在PM2.5暴露引起的细胞毒性中起着至关重要的作用。这些发现强调了短期暴露于低水平的PM2.5可能导致的明显的细胞损伤和遗传毒性。
    Fine particulate matter (PM2.5) can enter the human body in various ways and have adverse effects on human health. Human lungs and eyes are exposed to the air for a long time and are the first to be exposed to PM2.5. The \"liquid immersion exposure method\" has some limitations that prevent it from fully reflecting the toxic effects of particulate matter on the human body. In this study, the collected PM2.5 samples were chemically analyzed. An air-liquid interface (ALI) model with a high correlation to the in vivo environment was established based on human lung epithelial cells (A549) and immortalized human corneal epithelial cells (HCE-T). The VITROCELL Cloud 12 system was used to distribute PM2.5 on the cells evenly. After exposure for 6 h and 24 h, cell viability, apoptosis rate, reactive oxygen species (ROS) level, expression of inflammatory factors, and deoxyribonucleic acid (DNA) damage were measured. The results demonstrated significant dose- and time-dependent effects of PM2.5 on cell viability, cell apoptosis, ROS generation, and DNA damage at the ALI, while the inflammatory factors showed dose-dependent effects only. It should be noted that even short exposure to low doses of PM2.5 can cause cell DNA double-strand breaks and increased expression of γ-H2AX, indicating significant genotoxicity of PM2.5. Increased abundance of ROS in cells plays a crucial role in the cytotoxicity induced by PM2.5 exposure These findings emphasize the significant cellular damage and genotoxicity that may result from short-term exposure to low levels of PM2.5.
    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:炎症的持续存在是糖尿病足溃疡(DFU)的公认致病机制。我们旨在研究PLIN1在DFU患者组织中的表达,并评估其与炎症诱导的损伤的潜在关联。
    方法:我们对有或没有DFU的患者的足部皮肤进行了转录组测序和相关性分析。此外,我们通过分析PLIN1在组织和血清样本中的表达以及高糖刺激角质形成细胞(HaCaT细胞),检测了PLIN1与相关炎症指标之间的相关性.
    结果:PLIN1在DFU患者的组织和血清中上调。此外,PLIN1与白细胞呈正相关,中性粒细胞,单核细胞,C反应蛋白,血清中的降钙素原,以及组织中的IL-1β和TNF-α。细胞实验表明,PLIN1的表达降低导致iNOS的表达明显降低,IL-1β,IL-6、IL-18和TNF-α。PLIN1可能通过NF-κB信号通路介导创面炎性损伤。
    结论:我们的研究结果表明,PLIN1介导DFU的炎症损伤,为DFU的治疗提供了新的前景。
    BACKGROUND: The persistent presence of inflammation is a recognized pathogenic mechanisms of diabetic foot ulcers (DFUs). We aimed to investigate the expression of PLIN1 in tissues from DFU patients and assess its potential association with inflammation-induced damage.
    METHODS: We performed transcriptome sequencing and correlation analysis of the foot skin from patients with or without DFUs. Additionally, we examined the correlation between PLIN1 and related inflammatory indicators by analyzing PLIN1 expression in tissue and serum samples and through high-glucose stimulation of keratinocytes (HaCaT cells).
    RESULTS: PLIN1 is upregulated in the tissue and serum from DFU patients. Additionally, PLIN1 shows a positive correlation with leukocytes, neutrophils, monocytes, C-reactive protein, and procalcitonin in the serum, as well as IL-1β and TNF-α in the tissues. Experiments with Cells demonstrated that reduced expression of PLIN1 leads to significantly decreased expression of iNOS, IL-1β, IL-6, IL-18, and TNF-α. PLIN1 may mediate wound inflammatory damage through the NF-κB signaling pathway.
    CONCLUSIONS: Our findings suggest that PLIN1 mediates the inflammatory damage in DFU, offering new prospects for the treatment of DFU.
    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
    苯并(a)芘-7,8-二氢二醇-9,10-环氧化物(BPDE),环境污染物苯并(a)芘(B(a)P)的代谢产物可引起肺毒性和炎症。SIRT1,NAD+依赖性组蛋白脱乙酰酶,已知在各种疾病的发生和发展中调节炎症,但其对BPDE诱导的急性肺损伤的影响尚不清楚。本研究旨在探讨SIRT1在BPDE诱导的急性肺损伤中的作用。这里,用不同浓度(0.50、0.75和1.00μmol/L)的BPDE刺激人支气管上皮细胞(HBE)(BEAS-2B)24h,我们发现上清液中细胞因子的水平升高,细胞中SIRT1的表达下调,同时,BPDE刺激上调BEAS-2B细胞中HMGB1、TLR4和p-NF-κBp65的蛋白表达。然后在BPDE暴露前使用SIRT1的激活剂和抑制剂,研究表明,SIRT1的激活显着降低了炎症因子和HMGB1的水平,并降低了HMGB1,AC-HMGB1,TLR4和p-NF-κBp65蛋白的表达;而这些结果被SIRT1的抑制所逆转。本研究发现SIRT1活化可能通过调节HMGB1/TLR4/NF-κB通路保护BPDE诱导的BEAS-2B细胞炎症损伤。
    Benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), the metabolite of environmental pollutant benzo(a)pyrene (B(a)P) could induce pulmonary toxicity and inflammation. SIRT1, an NAD+ -dependent histone deacetylase, is known to regulate inflammation in the occurrence and development of various diseases, but its effects on BPDE-induced acute lung injury are still unknown. The present study aimed to explore the role of SIRT1 in BPDE-induced acute lung injury. Here, human bronchial epithelial (HBE) cells (BEAS-2B) cells were stimulated with BPDE at different concentrations (0.50, 0.75, and 1.00 μmol/L) for 24 h, we found that the levels of cytokines in the supernatant were increased and the expression of SIRT1 in cells was down-regulated, at the same time, BPDE stimulation up-regulated the protein expression of HMGB1, TLR4, and p-NF-κBp65 in BEAS-2B cells. Then the activator and inhibitor of SIRT1 were used before BPDE exposure, it was shown that the activation of SIRT1 significantly attenuated the levels of inflammatory cytokines and HMGB1, and reduced the expression of HMGB1, AC-HMGB1, TLR4, and p-NF-κBp65 protein; while these results were reversed by the inhibition of SIRT1. This study revealed that the SIRT1 activation may protect against BPDE-induced inflammatory damage in BEAS-2B cells by regulating the HMGB1/TLR4/NF-κB pathway.
    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
    糖尿病肾病(DN)是全球终末期肾病的主要原因。据报道,皂甙元(DSG)在DN足细胞损伤中起保护作用。本研究旨在探讨DSG在DN,及其在高葡萄糖(HG)诱导的足细胞DN体外模型中的作用机制。细胞活力,凋亡,炎症反应和胰岛素刺激的葡萄糖摄取使用细胞计数试剂盒-8,TUNEL,ELISA和2-脱氧-D-葡萄糖测定,分别。此外,免疫印迹法检测足细胞中AMP活化蛋白激酶(AMPK)/沉默蛋白1(SIRT1)/NF-κB信号相关蛋白的表达。结果表明,DSG增强了HG暴露后足细胞的活力,但抑制炎症损伤和减轻胰岛素抵抗。此外,DSG诱导AMPK/SIRT1/NF-κB信号通路的激活。此外,用化合物C处理,AMPK的抑制剂,DSG对HG诱导的足细胞的保护作用。因此,DSG可能是治疗糖尿病肾病的潜在治疗性化合物。
    Diabetic nephropathy (DN) is the predominant cause of end-stage renal disease globally. Diosgenin (DSG) has been reported to play a protective role in podocyte injury in DN. The present study aimed to explore the role of DSG in DN, as well as its mechanism of action in a high glucose (HG)-induced in vitro model of DN in podocytes. Cell viability, apoptosis, inflammatory response and insulin-stimulated glucose uptake were evaluated using Cell Counting Kit-8, TUNEL, ELISA and 2-deoxy-D-glucose assay, respectively. In addition, the expression of AMP-activated protein kinase (AMPK)/sirtuin 1 (SIRT1)/NF-κB signaling-related proteins in podocyte cells was measured using western blotting. The results indicated that DSG enhanced the viability of podocytes after HG exposure, but inhibited inflammatory damage and attenuated insulin resistance. Moreover, DSG induced the activation of the AMPK/SIRT1/NF-κB signaling pathway. Furthermore, treatment with compound C, an inhibitor of AMPK, counteracted the protective effects of DSG on HG-induced podocyte cells. Therefore, DSG may be a potential therapeutic compound for the treatment of diabetic nephropathy.
    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
    临床上治疗溃疡性结肠炎的药物主要影响炎症,但疗效有限,副作用多样。修复受损的肠粘膜是治疗溃疡性结肠炎的一种有希望的替代策略。三叶因子家族2(TFF2)可以修复肠粘膜,然而,它在体内的半衰期很短。为了提高TFF2的稳定性,我们制备了一种新的融合蛋白TFF2-Fc,研究TFF2-Fc对溃疡性结肠炎的治疗作用,并进一步说明了相关机制。我们发现,直肠内给药TFF2-Fc减轻了体重减轻,结肠缩短,疾病活动指数,肠组织损伤,葡聚糖硫酸钠(DSS)诱导的结肠炎小鼠的淋巴细胞浸润。体外,TFF2-Fc抑制Caco2细胞损伤和凋亡,促进细胞迁移,并通过在H2O2或炎症条件培养基(LPS-RAW264.7/CM)存在下激活P-ERK来增加Occludin和ZO-1的表达。此外,TFF2-Fc可以减少脂多糖(LPS)诱导的RAW264.7细胞中炎症细胞因子和活性氧的产生,并且还通过减少葡萄糖消耗和乳酸产生来抑制RAW264.7细胞向M1表型的极化。一起来看,在这项工作中,我们制备了一种新的融合蛋白TFF2-Fc,通过促进肠上皮细胞修复和抑制巨噬细胞炎症,和TFF2-Fc可能作为有希望的溃疡性结肠炎治疗剂。
    The clinical drugs for ulcerative colitis mainly affect the inflammatory symposiums with limited outcomes and various side effects. Repairing the damaged intestinal mucosa is a promising and alternative strategy to treat ulcerative colitis. Trefoil factor family 2 (TFF2) could repair the intestinal mucosa, however, it has a short half-life in vivo. To improve the stability of TFF2, we have prepared a new fusion protein TFF2-Fc with much stability, investigated the therapeutic effect of TFF2-Fc on ulcerative colitis, and further illustrated the related mechanisms. We found that intrarectally administered TFF2-Fc alleviated the weight loss, the colon shortening, the disease activity index, the intestinal tissue injury, and the lymphocyte infiltration in dextran sulfate sodium (DSS)-induced colitis mice. In vitro, TFF2-Fc inhibited Caco2 cells injury and apoptosis, promoted cellular migration, and increased the expression of Occludin and ZO-1 by activating P-ERK in the presence of H2O2 or inflammatory conditioned medium (LPS-RAW264.7/CM). Moreover, TFF2-Fc could reduce lipopolysaccharide (LPS)-induced production of inflammation cytokines and reactive oxygen species in RAW264.7 cells, and also inhibits the polarization of RAW264.7 cells to M1 phenotype by reducing glucose consumption and lactate production. Taken together, in this work, we have prepared a novel fusion protein TFF2-Fc, which could alleviate ulcerative colitis in vivo via promoting intestinal epithelial cells repair and inhibiting macrophage inflammation, and TFF2-Fc might serve as a promising ulcerative colitis therapeutic agent.
    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
    通过微调胸腺输出和初始T细胞的自我更新,将外周T细胞池维持在动态稳态。淋巴细胞减少或淋巴细胞数量减少与自身免疫性疾病有关,然而,对稳态机制知之甚少。这里,据报道,复制蛋白A1(RPA1)在T细胞稳态中起关键作用。利用T细胞特异性Rpa1缺陷(Rpa1fl/flCd4-cre)小鼠,Rpa1的缺失通过抑制外周T细胞群和限制TCR库多样性而导致淋巴细胞减少。此外,Rpa1fl/flCd4-cre小鼠对炎症性疾病的易感性增加,包括结肠炎和肝炎。临床分析表明,溃疡性结肠炎或其他自身炎症性疾病患者中RPA1的表达降低。机械上,RPA1的消耗通过触发基因组DNA泄漏到细胞质中激活ZBP1-RIPK3信号传导,从而导致T细胞坏死。这种由RPA1缺乏诱导的坏死T细胞死亡允许释放损伤相关分子模式(DAMPs),这反过来募集白细胞并加剧炎症反应。相互,对坏死信号的化学或遗传抑制可以改善Rpa1缺乏诱导的炎症损伤。因此,这些研究揭示了RPA1-ZBP1-RIPK3轴在T细胞稳态中的重要性,并为自身炎性疾病治疗提供了有希望的策略。
    The peripheral T cell pool is maintained at dynamic homeostasis through fine-tuning of thymic output and self-renewal of naïve T cells. Lymphopenia or reduced lymphocyte number is implicated in autoimmune diseases, yet little is known about the homeostatic mechanisms. Here, it is reported that the replication protein A1 (RPA1) plays a critical role in T cell homeostasis. Utilizing T cell-specific Rpa1-deficient (Rpa1fl/fl Cd4-cre) mice, loss of Rpa1 results in lymphopenia through restraining peripheral T cell population and limiting TCR repertoire diversity. Moreover, Rpa1fl/fl Cd4-cre mice exhibit increased susceptibility to inflammatory diseases, including colitis and hepatitis. Clinical analysis reveals that the expression of RPA1 is reduced in patients with ulcerative colitis or other autoinflammatory diseases. Mechanistically, depletion of RPA1 activates ZBP1-RIPK3 signaling through triggering the genomic DNA leakage into cytosol, consequently resulting in T cell necroptosis. This necroptotic T cell death induced by RPA1 deficiency allows the release of damage-associated molecular patterns (DAMPs), which in turn recruits leukocytes and exacerbates inflammatory response. Reciprocally, chemical or genetic inhibition of necroptosis signaling can ameliorate the Rpa1 deficiency-induced inflammatory damage. The studies thus uncover the importance of RPA1-ZBP1-RIPK3 axis in T cell homeostasis and provide a promising strategy for autoinflammatory disease treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号