TL1A

TL1A
  • 文章类型: Journal Article
    背景:气道上皮细胞(AEC)坏死导致气道过敏性炎症和哮喘加重。靶向肿瘤坏死因子样配体1A(TL1A)/死亡受体3(DR3)轴对哮喘气道炎症有治疗作用。TL1A在介导卵清蛋白(OVA)攻击的AECs坏死中的作用及其对气道炎症的贡献尚不清楚。
    方法:我们评估了受体相互作用的丝氨酸/苏氨酸蛋白激酶3(RIPK3)和混合谱系激酶结构域样蛋白(MLKL)在人血清和肺中的表达,并在组织学上验证了哮喘和OVA诱导的小鼠肺组织中MLKL磷酸化的水平。接下来,使用MLKL基因敲除小鼠和RIPK3抑制剂GSK872,我们研究了TL1A通过激活实验性哮喘的坏死性凋亡对气道炎症和气道屏障功能的影响。
    结果:在哮喘患者血清中观察到坏死标记蛋白的高表达,哮喘和OVA诱导的小鼠的气道上皮都激活了坏死。通过MLKL敲除或RIPK3抑制阻断坏死性凋亡可有效减轻支气管旁炎症,粘液分泌过多,和气道胶原纤维的积累,同时还抑制2型炎症因子的分泌。此外,通过在HBE细胞中沉默或过表达TL1A,TL1A/DR3显示在不存在胱天蛋白酶的情况下充当坏死的死亡触发因素。此外,发现重组TL1A蛋白在体内诱导坏死,MLKL的敲除部分逆转了TL1A诱导的病理变化。TL1A诱导的坏死通过降低紧密连接蛋白小带闭塞蛋白1(ZO-1)和闭塞蛋白的表达来破坏气道屏障功能,可能通过激活NF-κB信号通路。
    结论:TL1A诱导的气道上皮坏死在促进哮喘气道炎症和屏障功能障碍中起重要作用。抑制TL1A诱导的坏死途径可能是一种有前途的治疗策略。
    BACKGROUND: Airway epithelial cell (AEC) necroptosis contributes to airway allergic inflammation and asthma exacerbation. Targeting the tumor necrosis factor-like ligand 1 A (TL1A)/death receptor 3 (DR3) axis has a therapeutic effect on asthmatic airway inflammation. The role of TL1A in mediating necroptosis of AECs challenged with ovalbumin (OVA) and its contribution to airway inflammation remains unclear.
    METHODS: We evaluated the expression of the receptor-interacting serine/threonine-protein kinase 3(RIPK3) and the mixed lineage kinase domain-like protein (MLKL) in human serum and lung, and histologically verified the level of MLKL phosphorylation in lung tissue from asthmatics and OVA-induced mice. Next, using MLKL knockout mice and the RIPK3 inhibitor GSK872, we investigated the effects of TL1A on airway inflammation and airway barrier function through the activation of necroptosis in experimental asthma.
    RESULTS: High expression of necroptosis marker proteins was observed in the serum of asthmatics, and necroptosis was activated in the airway epithelium of both asthmatics and OVA-induced mice. Blocking necroptosis through MLKL knockout or RIPK3 inhibition effectively attenuated parabronchial inflammation, mucus hypersecretion, and airway collagen fiber accumulation, while also suppressing type 2 inflammatory factors secretion. In addition, TL1A/ DR3 was shown to act as a death trigger for necroptosis in the absence of caspases by silencing or overexpressing TL1A in HBE cells. Furthermore, the recombinant TL1A protein was found to induce necroptosis in vivo, and knockout of MLKL partially reversed the pathological changes induced by TL1A. The necroptosis induced by TL1A disrupted the airway barrier function by decreasing the expression of tight junction proteins zonula occludens-1 (ZO-1) and occludin, possibly through the activation of the NF-κB signaling pathway.
    CONCLUSIONS: TL1A-induced airway epithelial necroptosis plays a significant role in promoting airway inflammation and barrier dysfunction in asthma. Inhibition of the TL1A-induced necroptosis pathway could be a promising therapeutic strategy.
    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:朊病毒疾病是一种传染性和致命性的神经退行性疾病,其特征是错误折叠的朊病毒蛋白亚型(PrPSc)的积累,星形细胞增多症,小胶质细胞增生,海绵状体,和神经变性。细胞膜相关PrPSc蛋白和炎性细胞因子水平的升高提示着死亡受体(DR)途径的激活。DRs的激活调节,细胞存活或凋亡,自噬和坏死基于它们相互作用的衔接子。关于朊病毒疾病中DR途径的激活知之甚少。在正常小鼠大脑中表达的DR3和DR5从未在朊病毒疾病中研究过,它们的配体和任何DR衔接子也是如此。这一研究差距是值得注意的,并在本研究中进行了调查。
    方法:C57BL/6J小鼠用落基山实验室瘙痒病小鼠朊病毒株感染。通过观察形态和行为异常来检查朊病毒病的进展。使用检测印迹和脑切片上的蛋白质的基于抗体的技术,分别测量PrP同种型和GFAP的水平作为PrPSc积累和星形细胞增生的标志物。DR的水平,它们的糖基化和胞外域脱落,和相关因素保证他们在蛋白质水平上的检查,因此,这项研究采用了蛋白质印迹分析。
    结果:感染朊病毒的小鼠出现了运动障碍和神经病理学,如PrPSc积累和星形胶质细胞增多,类似于其他朊病毒疾病。这项研究的结果表明,所有DR配体的高表达,TNFR1、Fas和p75NTR但降低DR3和DR5水平。DR衔接蛋白如TRADD和TRAF2(主要调节促存活途径)的水平降低。FADD,主要调节细胞死亡,其水平保持不变。RIPK1调节促生存,凋亡和坏死,其表达和蛋白水解(抑制坏死但激活凋亡)增加。
    结论:本研究的结果为DR3,DR5,DR6,TL1A的参与提供了证据。TRAIL,TRADD,TRAF2、FADD和RIPK1为首次在朊病毒疾病中的应用。从这项研究中获得的知识讨论了这16种差异表达的DR因子对我们对朊病毒疾病的多方面神经病理学的理解以及对朊病毒疾病特异性神经病理学的潜在靶向治疗干预的未来探索的可能影响。
    BACKGROUND: Prion diseases are transmissible and fatal neurodegenerative diseases characterized by accumulation of misfolded prion protein isoform (PrPSc), astrocytosis, microgliosis, spongiosis, and neurodegeneration. Elevated levels of cell membrane associated PrPSc protein and inflammatory cytokines hint towards the activation of death receptor (DR) pathway/s in prion diseases. Activation of DRs regulate, either cell survival or apoptosis, autophagy and necroptosis based on the adaptors they interact. Very little is known about the DR pathways activation in prion disease. DR3 and DR5 that are expressed in normal mouse brain were never studied in prion disease, so also their ligands and any DR adaptors. This research gap is notable and investigated in the present study.
    METHODS: C57BL/6J mice were infected with Rocky Mountain Laboratory scrapie mouse prion strain. The progression of prion disease was examined by observing morphological and behavioural abnormalities. The levels of PrP isoforms and GFAP were measured as the marker of PrPSc accumulation and astrocytosis respectively using antibody-based techniques that detect proteins on blot and brain section. The levels of DRs, their glycosylation and ectodomain shedding, and associated factors warrant their examination at protein level, hence western blot analysis was employed in this study.
    RESULTS: Prion-infected mice developed motor deficits and neuropathology like PrPSc accumulation and astrocytosis similar to other prion diseases. Results from this research show higher expression of all DR ligands, TNFR1, Fas and p75NTR but decreased levels DR3 and DR5. The levels of DR adaptor proteins like TRADD and TRAF2 (primarily regulate pro-survival pathways) are reduced. FADD, which primarily regulate cell death, its level remains unchanged. RIPK1, which regulate pro-survival, apoptosis and necroptosis, its expression and proteolysis (inhibits necroptosis but activates apoptosis) are increased.
    CONCLUSIONS: The findings from the present study provide evidence towards the involvement of DR3, DR5, DR6, TL1A, TRAIL, TRADD, TRAF2, FADD and RIPK1 for the first time in prion diseases. The knowledge obtained from this research discuss the possible impacts of these 16 differentially expressed DR factors on our understanding towards the multifaceted neuropathology of prion diseases and towards future explorations into potential targeted therapeutic interventions for prion disease specific neuropathology.
    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
    TL1A在调节对炎症性肠病(IBD)和肠纤维化至关重要的免疫途径中的关键作用提供了有希望的治疗靶标。评估抗TL1A抗体的2期试验(TUSCANY和ARTEMIS-UC)显示在扩大IBD治疗选择方面取得了进展。首次在人类数据显示与抗TL1A治疗后的细胞外基质重塑和纤维化相关的基因表达降低。研究药物TEV-48574,可能发挥双重抗纤维化和抗炎作用,正在接受溃疡性结肠炎(UC)和克罗恩病(CD)的2期篮子研究。人们热切期待结果,标志着IBD治疗的进展。这篇批判性评论全面审查了现有的文献,照亮TL1A和DR3在IBD中的复杂作用,强调不断发展的治疗前景和正在进行的临床试验,对更有效的IBD管理有潜在的影响。
    The pivotal role of TL1A in modulating immune pathways crucial for inflammatory bowel disease (IBD) and intestinal fibrosis offers a promising therapeutic target. Phase 2 trials (TUSCANY and ARTEMIS-UC) evaluating an anti-TL1A antibody show progress in expanding IBD therapeutic options. First-in-human data reveal reduced expression of genes associated with extracellular matrix remodeling and fibrosis post-anti-TL1A treatment. Investigational drug TEV-48574, potentially exerting dual antifibrotic and anti-inflammatory effects, is undergoing a phase 2 basket study in both ulcerative colitis (UC) and Crohn disease (CD). Results are eagerly awaited, marking advancements in IBD therapeutics. This critical review comprehensively examines the existing literature, illuminating TL1A and the intricate role of DR3 in IBD, emphasizing the evolving therapeutic landscape and ongoing clinical trials, with potential implications for more effective IBD 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
    嗜酸性粒细胞性哮喘是最普遍和明确的哮喘表型。尽管大多数患者对皮质类固醇治疗和T2生物制剂有反应,仍然有一个子集有反复发作的哮喘加重,强调需要额外的治疗来完全改善气道嗜酸性粒细胞增多。第2组先天淋巴细胞(ILC2)被认为是通过产生大量的2型细胞因子在嗜酸性粒细胞性哮喘的发病机理中的关键参与者。即IL-5和IL-13。哮喘患者气道中的ILC2数量增加,并且在严重的强的松依赖性哮喘伴嗜酸性粒细胞不受控制的痰中检测到的活化ILC2数量最多。尽管上皮源性细胞因子是ILC2激活的重要介质,新出现的证据表明,其他途径刺激ILC2功能。肿瘤坏死因子超家族(TNFSF)及其受体(TNFRSF)促进ILC2活性。在这次审查中,我们讨论了支持ILC2和TNFSF/TNFRSF轴在嗜酸性粒细胞性哮喘中的关系的证据,以及这种关系在有气道自身免疫反应的重度哮喘中的作用.
    Eosinophilic asthma is the most prevalent and well-defined phenotype of asthma. Despite a majority of patients responding to corticosteroid therapy and T2 biologics, there remains a subset that have recurrent asthma exacerbations, highlighting a need for additional therapies to fully ameliorate airway eosinophilia. Group 2 innate lymphoid cells (ILC2) are considered key players in the pathogenesis of eosinophilic asthma through the production of copious amounts of type 2 cytokines, namely IL-5 and IL-13. ILC2 numbers are increased in the airways of asthmatics and with the greatest numbers of activated ILC2 detected in sputa from severe prednisone-dependent asthma with uncontrolled eosinophilia. Although epithelial-derived cytokines are important mediators of ILC2 activation, emerging evidence suggests that additional pathways stimulate ILC2 function. The tumor necrosis factor super family (TNFSF) and its receptors (TNFRSF) promote ILC2 activity. In this review, we discuss evidence supporting a relationship between ILC2 and TNFSF/TNFRSF axis in eosinophilic asthma and the role of this relationship in severe asthma with airway autoimmune responses.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    死亡受体3(DR3)是肿瘤坏死因子受体超家族的细胞因子受体,在先天和适应性免疫中起着多方面的作用。基于胞质尾部的死亡域基序,DR3已被提出并在功能上被确认为凋亡的触发因素。进一步研究,然而,还揭示了DR3在其他细胞途径中的作用,包括炎症,生存,和扩散。DR3在各种细胞类型中表达,包括T细胞,B细胞,先天淋巴细胞,骨髓细胞,成纤维细胞,甚至在免疫系统之外。因为DR3主要在T细胞上表达,导致自身免疫和其他疾病的DR3介导的免疫扰动主要归因于T细胞的DR3活化。然而,DR3控制哪些T细胞亚群和哪些T效应子功能来驱动这些过程仍未完全理解。以前发现DR3参与改变CD4T辅助亚群分化,扩大Foxp3+Treg细胞池,并在肠道内维持上皮内γδT细胞。最近的研究进一步揭示了DR3在调节先天样不变NKT(iNKT)细胞激活方面的一个以前未被承认的方面,扩大DR3介导的T谱系细胞免疫的范围。重要的是,在iNKT细胞的背景下,DR3结扎在激动性TCR信号传导中发挥了共刺激作用,揭示了T细胞活化和增殖的新调控框架。本综述旨在总结有关DR3对常规T细胞和先天样T细胞的作用的最新发现,并在免疫发病机制的背景下进行讨论。
    Death Receptor 3 (DR3) is a cytokine receptor of the Tumor Necrosis Factor receptor superfamily that plays a multifaceted role in both innate and adaptive immunity. Based on the death domain motif in its cytosolic tail, DR3 had been proposed and functionally affirmed as a trigger of apoptosis. Further studies, however, also revealed roles of DR3 in other cellular pathways, including inflammation, survival, and proliferation. DR3 is expressed in various cell types, including T cells, B cells, innate lymphocytes, myeloid cells, fibroblasts, and even outside the immune system. Because DR3 is mainly expressed on T cells, DR3-mediated immune perturbations leading to autoimmunity and other diseases were mostly attributed to DR3 activation of T cells. However, which T cell subset and what T effector functions are controlled by DR3 to drive these processes remain incompletely understood. DR3 engagement was previously found to alter CD4 T helper subset differentiation, expand the Foxp3+ Treg cell pool, and maintain intraepithelial γδ T cells in the gut. Recent studies further unveiled a previously unacknowledged aspect of DR3 in regulating innate-like invariant NKT (iNKT) cell activation, expanding the scope of DR3-mediated immunity in T lineage cells. Importantly, in the context of iNKT cells, DR3 ligation exerted costimulatory effects in agonistic TCR signaling, unveiling a new regulatory framework in T cell activation and proliferation. The current review is aimed at summarizing such recent findings on the role of DR3 on conventional T cells and innate-like T cells and discussing them in the context of immunopathogenesis.
    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
    产生白细胞介素(IL)-17的γδT(γδT17)细胞介导屏障组织中的炎症反应。γδT17细胞活化失调可导致IL-17和IL-22的过度产生和炎症性疾病的发生,包括牛皮癣。已知IL-23和IL-1β协同激活γδT17细胞,但γδT17细胞的调控机制尚未完全阐明。本研究旨在揭示炎性细胞因子肿瘤坏死因子样配体1A(TL1A)对γδT17细胞活化和银屑病发展的贡献。
    将抗TL1A抗体注射到咪喹莫特(IMQ)诱导的鼠银屑病模型中。在脾和真皮γδT细胞中分析TL1A受体表达。在IL-23,IL-1β刺激下,在体外和体内测试γδT细胞的细胞因子产生,和TL1A。将TL1A应用于皮内注射IL-23诱导的银屑病模型。将缺乏γδT细胞的小鼠皮内注射IL-23加TL1A,以验证TL1A依赖性γδT细胞活化对牛皮癣发展的贡献。
    TL1A的中和作用减弱了IMQ处理皮肤中的γδT17细胞活化。TL1A与IL-23协同诱导脾γδT17细胞产生细胞因子。真皮γδT17细胞以高水平组成型表达TL1A受体,并在皮内IL-23和TL1A注射后剧烈产生IL-22,但不单独产生IL-23。TL1A在野生型中但在γδT细胞缺陷型小鼠中不会加重IL-23注射诱导的皮肤症状。
    这些发现提示了γδT细胞通过TL1A及其参与银屑病发病机制的新调控机制,作为可能的治疗靶点。
    Interleukin (IL)-17-producing γδT (γδT17) cells mediate inflammatory responses in barrier tissues. Dysregulated γδT17 cell activation can lead to the overproduction of IL-17 and IL-22 and the development of inflammatory diseases, including psoriasis. IL-23 and IL-1β are known to synergistically activate γδT17 cells, but the regulatory mechanisms of γδT17 cells have not been fully elucidated. This study aimed to reveal the contribution of the inflammatory cytokine tumor necrosis factor-like ligand 1A (TL1A) to γδT17 cell activation and psoriasis development.
    Anti-TL1A antibody was injected into an imiquimod (IMQ)-induced murine psoriasis model. TL1A receptor expression was analyzed in splenic and dermal γδT cells. γδT cells were tested for cytokine production in vitro and in vivo under stimulation with IL-23, IL-1β, and TL1A. TL1A was applied to a psoriasis model induced by intradermal IL-23 injection. Mice deficient in γδT cells were intradermally injected with IL-23 plus TL1A to verify the contribution of TL1A-dependent γδT-cell activation to psoriasis development.
    Neutralization of TL1A attenuated γδT17 cell activation in IMQ-treated skin. TL1A induced cytokine production by splenic γδT17 cells in synergy with IL-23. Dermal γδT17 cells constitutively expressed a TL1A receptor at high levels and vigorously produced IL-22 upon intradermal IL-23 and TL1A injection but not IL-23 alone. TL1A exacerbated the dermal symptoms induced by IL-23 injection in wild-type but not in γδT cell-deficient mice.
    These findings suggest a novel regulatory mechanism of γδT cells through TL1A and its involvement in psoriasis pathogenesis as a possible therapeutic target.
    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
    肿瘤坏死因子样细胞因子1A(TL1A)与死亡受体3(DR3)的结合在树突状细胞(DC)和T细胞之间的相互作用中起重要作用,并有助于肠道炎症的发展。然而,在炎症性肠病发病过程中,表达TL1A的DC介导肠道固有层(LP)辅助性T(Th)细胞分化的机制尚不清楚.在这项研究中,我们发现TL1A/DR3以T-T和DC-T细胞相互作用依赖性方式促进Th1和Th17细胞分化。在T细胞转移结肠炎模型中,缺乏TL1A的CD4+T细胞未能极化成Th1/Th17细胞,也没有引起结肠炎症。值得注意的是,TL1A位于DCs的胞浆和胞核,正调节DC特异性ICAM抓取非整合素/RAF1/核因子κB信号通路,增强了DCs的抗原摄取能力,并促进TLR4介导的DC激活,诱导幼稚CD4+T细胞分化为Th1和Th17细胞。我们的工作揭示了TL1A在炎症性肠病发病机制中起着调节作用。
    The binding of tumor necrosis factor-like cytokine 1A (TL1A) to death receptor 3 (DR3) plays an important role in the interaction between dendritic cells (DCs) and T cells and contributes to intestinal inflammation development. However, the mechanism by which DCs expressing TL1A mediate helper T (Th) cell differentiation in the intestinal lamina propria (LP) during the pathogenesis of inflammatory bowel disease remains unclear. In this study, we found that TL1A/DR3 promoted Th1 and Th17 cell differentiation in T-T and DC-T cell interaction-dependent manners. TL1A-deficient CD4+ T cells failed to polarize into Th1/Th17 cells and did not cause colonic inflammation in a T cell transfer colitis model. Notably, TL1A was located in the cytoplasm and nuclei of DCs, positively regulated the DC-specific ICAM-grabbing nonintegrin/RAF1/nuclear factor κB signaling pathway, enhanced the antigen uptake ability of DCs, and promoted TLR4-mediated DC activation, inducing naive CD4+ T cell differentiation into Th1 and Th17 cells. Our work reveals that TL1A plays a regulatory role in inflammatory bowel disease pathogenesis.
    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:本研究旨在探讨黄芪甲苷(AS-IV)对坏死性小肠结肠炎(NEC)新生大鼠模型的可能影响,并验证TNF样配体1A(TL1A)和NF-κB信号通路的可能意义。
    方法:通过配方喂养建立NEC新生大鼠模型,冷/窒息应激和脂多糖(LPS)灌胃法。外观,评估了进行NEC建模的大鼠的活动和皮肤以及病理状态。H&E染色后观察肠组织。氧化应激生物标志物(SOD,MDA和GSH-Px)和炎性细胞因子(TNF-α,通过ELISA和qRT-PCR检测IL-1β和IL-6)。免疫印迹法和免疫组化法检测TL1A和NF-κB信号通路相关蛋白的表达。通过TUNEL评估细胞凋亡。
    结果:NEC新生大鼠模型成功建立,TL1A高表达,NF-κB信号通路被激活,AS-IV治疗可以抑制NEC大鼠的TL1A和NF-κB信号通路。同时,NEC模型大鼠肠组织炎症反应增加,AS-IV可通过抑制TL1A和NF-κb信号通路减轻NEC大鼠的炎症反应。
    结论:AS-IV可抑制NEC新生大鼠模型TL1A的表达和NF-κb信号通路,从而减轻炎症反应。
    OBJECTIVE: This study aims to explore the possible effect of Astragaloside IV (AS-IV) on necrotizing enterocolitis (NEC) neonatal rat models and verify the possible implication of TNF-like ligand 1 A (TL1A) and NF-κB signal pathway.
    METHODS: NEC neonatal rat models were established through formula feeding, cold/asphyxia stress and Lipopolysaccharide (LPS) gavage method. The appearance, activity and skin as well as the pathological status of rats subjected to NEC modeling were assessed. The intestinal tissues were observed after H&E staining. The expression of oxidative stress biomarkers (SOD, MDA and GSH-Px) and inflammatory cytokines (TNF-α, IL-1β and IL-6) were detected by ELISA and qRT-PCR. Western blotting and immunohistochemistry were applied to detect expressions of TL1A and NF-κB signal pathway-related proteins. Cell apoptosis was assessed by TUNEL.
    RESULTS: NEC neonatal rat models were established successfully, in which TL1A was highly expressed and NF-κB signal pathway was activated, while TL1A and NF-κB signal pathway can be suppressed by AS-IV treatment in NEC rats. Meanwhile, inflammatory response in intestinal tissues was increased in NEC rat models and AS-IV can attenuate inflammatory response in NEC rats through inhibiting TL1A and NF-κb signal pathway.
    CONCLUSIONS: AS-IV can inhibit TL1A expression and NF-κb signal pathway to attenuate the inflammatory response in NEC neonatal rat models.
    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:我们研究了其机制,其中肿瘤坏死因子样配体1A(TL1A)介导术后认知功能障碍(POCD)中星形胶质细胞的A1分化。
    方法:通过Morris水迷宫和旷场试验评估小鼠的认知和行为能力,同时通过RT-qPCR检测关键A1和A2星形胶质细胞因子的水平。免疫组化(IHC)染色检测GFAP的表达,蛋白质印迹用于测定相关蛋白质的水平,采用酶联免疫吸附试验(ELISA)检测炎性细胞因子水平。
    结果:结果表明,TL1A可以促进小鼠认知功能障碍的进展。星形胶质细胞分化为A1表型,而星形胶质细胞A2生物标志物的变化不明显。NLRP3基因敲除或NLRP3抑制剂干预可以抑制TL1A的作用,改善认知功能障碍和抑制A1分化。
    结论:我们的结果表明TL1A在小鼠POCD中起重要作用,通过NLRP3促进星形胶质细胞的A1分化,从而加剧认知功能障碍的进展。
    We investigated the mechanism, whereby tumor necrosis factor-like ligand 1A (TL1A) mediates the A1 differentiation of astrocytes in postoperative cognitive dysfunction (POCD).
    The cognitive and behavioral abilities of mice were assessed by Morris water maze and open field tests, while the levels of key A1 and A2 astrocyte factors were detected by RT-qPCR. Immunohistochemical (IHC) staining was used to examine the expression of GFAP, western blot was used to assay the levels of related proteins, and enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of inflammatory cytokines.
    The results showed that TL1A could promote the progression of cognitive dysfunction in mice. Astrocytes differentiated into A1 phenotype, while unobvious changes were noted in astrocyte A2 biomarkers. Knockout of NLRP3 or intervention with NLRP3 inhibitor could inhibit the effect of TL1A, improving the cognitive dysfunction and suppressing the A1 differentiation.
    Our results demonstrate that TL1A plays an important role in POCD in mice, which promotes the A1 differentiation of astrocytes through NLRP3, thereby exacerbating the progression of cognitive dysfunction.
    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
    肿瘤坏死因子样配体1A(TL1A),TNF超家族的一员,调节炎症反应和免疫防御。最近在鱼类中发现了TL1A同源物,但是它们的功能还没有被研究过。在这项研究中,在草鱼(Ctenpharyngodonidella)中鉴定出TL1A同源物,并研究了其生物活性。草鱼tl1a(Citl1a)基因在组织中组成型表达,在肝脏中检测到最高的表达。对嗜水气单胞菌感染的反应上调。重组CiTL1A在细菌中产生,并显示出刺激IL1β的表达,tnfα,原发性头肾白细胞中的caspase8和ifnγ。此外,免疫共沉淀实验显示CiTL1A与DR3相互作用并通过激活DR3诱导细胞凋亡。结果表明,TL1A调节炎症和细胞凋亡,并参与鱼类对细菌感染的免疫防御。
    Tumor necrosis factor like ligand 1A (TL1A), a member of TNF superfamily, regulates inflammatory response and immune defense. TL1A homologues have recently been discovered in fish, but their functions have not been studied. In this study, a TL1A homologue was identified in grass carp (Ctenopharyngodon idella) and its bioactivities were investigated. The grass carp tl1a (Citl1a) gene was constitutively expressed in tissues, with the highest expression detected in the liver. It was upregulated in response to infection with Aeromonas hydrophila. The recombinant CiTL1A was produced in bacteria and was shown to stimulate the expression of il1β, tnfα, caspase 8 and ifnγ in the primary head kidney leucocytes. In addition, co-immunoprecipitation assay revealed that CiTL1A interacted with DR3 and induced apoptosis via activation of DR3. The results demonstrate that TL1A regulates inflammation and apoptosis and is involved in the immune defense against bacterial infection in fish.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号