immune regulatory

  • 文章类型: Journal Article
    伤口愈合的微环境易受细菌感染,慢性炎症,氧化应激,血管生成不足,需要开发具有抗菌作用的创新伤口敷料,抗炎,抗氧化剂,和血管生成能力。这项研究制作了一种新型的多功能细菌纤维素复合膜,该复合膜注入了铜掺杂的碳点(BC/Cu(II)-RCD)。研究结果验证了通过氢键相互作用将铜掺杂的碳点成功加载到BC膜上。与纯BC膜相比,BC/Cu(II)-RCDs复合膜显示出显著增强的亲水性,拉伸性能,和热稳定性。多种体外试验证明BC/Cu(II)-RCDs复合膜具有优异的生物相容性和抗菌活性,以及它们加速炎症阶段和刺激血管生成的能力。体内试验证实了膜促进上皮再生的能力,胶原蛋白沉积,大鼠全层皮肤伤口的组织再生,同时也抑制感染的全层皮肤伤口的炎症。更重要的是,BC/Cu(II)-RCDs复合膜的处理可能导致VEGF和MAPK信号蛋白的激活,它们是细胞迁移的关键参与者,血管生成,和皮肤组织发育。实质上,开发的BC/Cu(II)-RCDs复合膜显示出治疗感染伤口的希望,并作为药用绷带的可行替代材料。
    The microenvironment of wound healing is susceptible to bacterial infection, chronic inflammation, oxidative stress, and inadequate angiogenesis, requiring the development of innovative wound dressings with antibacterial, anti-inflammatory, antioxidant, and angiogenic capabilities. This research crafted a new multifunctional bacterial cellulose composite membrane infused with copper-doped carbon dots (BC/Cu(II)-RCDs). Findings validated the successful loading of copper-doped carbon dots onto the BC membrane via hydrogen bonding interactions. Compared to the pure BC membrane, the BC/Cu(II)-RCDs composite membrane exhibited significantly enhanced hydrophilicity, tensile properties, and thermal stability. Diverse in vitro assays demonstrated excellent biocompatibility and antibacterial activity of BC/Cu(II)-RCDs composite membranes, alongside their ability to expedite the inflammatory phase and stimulate angiogenesis. In vivo trials corroborated the membrane\'s ability to foster epithelial regeneration, collagen deposition, and tissue regrowth in full-thickness skin wounds in rats while also curbing inflammation in infected full-thickness skin wounds. More importantly, the treatment of the BC/Cu(II)-RCDs composite membrane may result in the activation of VEGF and MAPK signaling proteins, which are key players in cell migration, angiogenesis, and skin tissue development. In essence, the developed BC/Cu(II)-RCDs composite membrane shows promise for treating infected wounds and serves as a viable alternative material for medicinal bandages.
    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
    目的:鉴定类风湿关节炎(RA)早期诊断的生物标志物并探讨其可能的免疫调节机制。
    方法:对RA的差异表达基因进行筛选,并使用limma进行功能注释,RRA,批量校正,和clusterProfiler。从STRING数据库检索蛋白质-蛋白质相互作用网络,利用Cytoscape3.8.0和GeneMANIA选择关键基因并预测其相互作用机制。ROC曲线用于验证基于关键基因的诊断模型的准确性。通过机器学习选择疾病特异性免疫细胞,并使用Corplot软件包分析其与关键基因的相关性。使用GSEA方法探索关键基因的生物学功能。观察STAT1在胶原诱导性关节炎(CIA)大鼠滑膜组织中的表达。
    结果:我们在RA中确定了9个核心关键基因(CD3G,CD8A,SYK,LCK,IL2RG,STAT1、CCR5、ITGB2和ITGAL),主要通过细胞因子相关途径调节滑膜炎症。ROC曲线分析显示9个核心基因的预测准确率较高,其中STAT1的AUC最高(0.909)。相关分析显示CD3G具有很强的相关性,ITGAL,LCK,CD8A,和STAT1与疾病特异性免疫细胞,STAT1与M1型巨噬细胞的相关性最强(R=0.68,P=2.9e-08)。CIA大鼠踝关节滑膜组织显示STAT1和p-STAT1的高表达,滑膜成纤维细胞的细胞核和细胞质之间STAT1的表达显着差异。治疗后细胞核中p-STAT1和STAT1的蛋白表达显著降低。
    结论:CD3G,CD8A,SYK,LCK,IL2RG,STAT1、CCR5、ITGB2和ITGAL可作为RA早期诊断的生物标志物。基因免疫细胞通路,如CD3G/CD8A/LCK-γδT细胞,ITGAL-Tfh细胞,STAT1-M1型巨噬细胞可能与RA的发生发展密切相关。
    OBJECTIVE: To identify the biomarkers for early rheumatoid arthritis (RA) diagnosis and explore the possible immune regulatory mechanisms.
    METHODS: The differentially expressed genesin RA were screened and functionally annotated using the limma, RRA, batch correction, and clusterProfiler. The protein-protein interaction network was retrieved from the STRING database, and Cytoscape 3.8.0 and GeneMANIA were used to select the key genes and predicting their interaction mechanisms. ROC curves was used to validate the accuracy of diagnostic models based on the key genes. The disease-specific immune cells were selected via machine learning, and their correlation with the key genes were analyzed using Corrplot package. Biological functions of the key genes were explored using GSEA method. The expression of STAT1 was investigated in the synovial tissue of rats with collagen-induced arthritis (CIA).
    RESULTS: We identified 9 core key genes in RA (CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL), which regulate synovial inflammation primarily through cytokines-related pathways. ROC curve analysis showed a high predictive accuracy of the 9 core genes, among which STAT1 had the highest AUC (0.909). Correlation analysis revealed strong correlations of CD3G, ITGAL, LCK, CD8A, and STAT1 with disease-specific immune cells, and STAT1 showed the strongest correlation with M1-type macrophages (R=0.68, P=2.9e-08). The synovial tissues of the ankle joints of CIA rats showed high expressions of STAT1 and p-STAT1 with significant differential expression of STAT1 between the nucleus and the cytoplasm of the synovial fibroblasts. The protein expressions of p-STAT1 and STAT1 in the cell nuclei were significantly reduced after treatment.
    CONCLUSIONS: CD3G, CD8A, SYK, LCK, IL2RG, STAT1, CCR5, ITGB2, and ITGAL may serve as biomarkers for early diagnosis of RA. Gene-immune cell pathways such as CD3G/CD8A/LCK-γδ T cells, ITGAL-Tfh cells, and STAT1-M1-type macrophages may be closely related with the development of RA.
    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:患者从COVID-19中恢复后,很少有症状持续很长时间,称为“长期COVID”。我们探索了COVID-19的潜在微生物危险因素,以便更深入地了解和帮助这些后遗症的后续治疗。
    方法:使用在多个时间点从恢复的COVID-19患者和健康对照中收集的全血RNA-Seq数据进行微生物组重新注释。随后,进行了一系列下游分析,以揭示从SARS-CoV-2感染中恢复的患者的微生物特征。
    结果:感染后12周的血液微生物组明显受到干扰,包括芽孢杆菌/拟杆菌的比例增加和更高的微生物α多样性。此外,在感染后12周确定了一组病原微生物,包括金黄色葡萄球菌,肺炎克雷伯菌,肺炎链球菌,鲍曼不动杆菌,铜绿假单胞菌,与参与免疫调节和嗅觉转导途径的宿主基因呈正相关。几种微生物,例如肺炎链球菌与浸润的免疫细胞有关,如M2巨噬细胞。
    结论:这项研究提供了对血液微生物组和COVID-19后遗症之间关系的见解。在恢复的COVID-19患者中富集了几种病原微生物,从而影响了参与免疫和嗅觉转导途径的宿主基因,在COVID-19后遗症中起关键作用。
    Few symptoms persist for a long time after patients recover from COVID-19, called \"long COVID\". We explored the potential microbial risk factors for COVID-19 for a deeper understanding and assistance in the follow-up treatment of these sequelae.
    Microbiome re-annotation was performed using whole blood RNA-Seq data collected from recovered COVID-19 patients and healthy controls at multiple time points. Subsequently, a series of downstream analyses were conducted to reveal the microbial characteristics of patients who recovered from SARS-CoV-2 infection.
    The blood microbiome at 12 weeks post-infection was most evidently disturbed, including an increasing ratio of Bacillota/Bacteroidota and a higher microbial alpha diversity. In addition, a group of pathogenic microbes at 12 weeks post-infection were identified, including Staphylococcus aureus, Klebsiella pneumoniae, Streptococcus pneumoniae, Acinetobacter baumannii, and Pseudomonas aeruginosa, which were positively associated with host genes involved in immune regulatory and olfactory transduction pathways. Several microbes, such as Streptococcus pneumoniae were associated with infiltrating immune cells, such as M2 macrophages.
    This study provides insights into the relationship between the blood microbiome and COVID-19 sequelae. Several pathogenic microbes were enriched in recovered COVID-19 patients and thus affected host genes participating in the immune and olfactory transduction pathways, which play critical roles in COVID-19 sequelae.
    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
    肿瘤微环境与癌症中关键信号通路网络之间的关系在肿瘤的发生和发展中起着关键作用。肿瘤相关巨噬细胞(TAMs)是肿瘤微环境中重要的炎性细胞,在肿瘤发生、发展中起着重要作用。恶性肿瘤中的巨噬细胞,主要是M2亚型,通过产生细胞因子和下调抗炎免疫反应来促进肿瘤进展。几篇文章研究了巨噬细胞对癌症化疗药物敏感性的影响,但是很少有这样的文章在胆管癌中被报道,因此,我们研究了与M1相比,M2巨噬细胞对胆管癌细胞对Lenvatinib敏感性的影响。
    THP-1单核细胞被佛波醇12-肉豆蔻酸酯13-乙酸酯(PMA)极化为M0巨噬细胞,然后被LPS诱导分化为M1和M2巨噬细胞,分别为IFN-γ和IL-4和IL-13。巨噬细胞和胆管癌细胞在Lenvatinib给药24小时前共培养,通过蛋白质印迹检测癌细胞凋亡,膜联蛋白V和PI染色的FACS分析。此外,我们使用xCELLigenceRTCASP仪器(ACEABio-sciences)监测胆管癌细胞和巨噬细胞共培养中Lenvatinib给药的细胞活力。在免疫缺陷小鼠的肿瘤发生后,Lenvatinib给药,用免疫组织化学方法研究M2对胆管癌细胞生物学特性的影响。
    M1和M2标记的mRNA和蛋白表达证实了THP-1来源的巨噬细胞的极化,Lenvatinib诱导的胆管癌细胞凋亡显著减少,与M2巨噬细胞共培养,而与M1巨噬细胞共培养的胆管癌细胞凋亡增加。在CDX模型中,Lenvatinib诱导的癌细胞凋亡明显减少,在M2巨噬细胞的存在下,增殖细胞增加。与M2共培养的胆管癌细胞中血管生成相关因子显著增加。
    与M1相比,M2巨噬细胞可以通过免疫调节抑制Lenvatinib对胆管癌的抗肿瘤作用,可能与M2巨噬细胞的肿瘤血管生成因子作用有关。
    The relationship between the tumor microenvironment and the network of key signaling pathways in cancer plays a key role in the occurrence and development of tumors. Tumor-associated macrophages (TAMs) are important inflammatory cells in the tumor microenvironment and play an important role in tumorigenesis and progression. Macrophages in malignant tumors, mainly the M2 subtype, promote tumor progression by producing cytokines and down-regulating anti-inflammatory immune responses. Several articles have investigated the effect of macrophages on the sensitivity of cancer chemotherapeutic agents, but few such articles have been reported in cholangiocarcinoma, so we investigated the effect of M2 macrophage on the sensitivity of cholangiocarcinoma cells to Lenvatinib compared to M1.
    THP-1 monocytes were polarized to M0 macrophage by phorbol 12-myristate 13-acetate (PMA) and then induced to differentiate into M1 and M2 macrophages by LPS, IFN-γ and IL-4 and IL-13, respectively. Macrophages and cholangiocarcinoma cells were co-cultured prior to 24 hours of Lenvatinib administration, cancer cell apoptosis was detected by western-blot, FACS analysis of Annexin V and PI staining. Furthermore, we use xCELLigence RTCA SP Instrument (ACEA Bio-sciences) to monitor cell viability of Lenvatinib administration in co-culture of cholangiocarcinoma cells and macrophages. After tumorigenesis in immunodeficient mice, Lenvatinib was administered, and the effects of M2 on biological characteristics of cholangiocarcinoma cells were investigated by immuno-histochemistry.
    mRNA and protein expression of M1 and M2 markers confirmed the polarization of THP-1 derived macrophages, which provided a successful and efficient model of monocyte polarization to TAMs. Lenvatinib-induced apoptosis of cholangiocarcinoma cells was significantly reduced when co-cultured with M2 macrophage, whereas apoptosis of cholangiocarcinoma cells co-cultured with M1 macrophage was increased. In the CDX model, Lenvatinib-induced cancer cell apoptosis was markedly reduced, and proliferative cells increased in the presence of M2 macrophages. Angiogenesis related factors was significantly increased in cholangiocarcinoma cells co-cultured with M2.
    Compared with M1, M2 macrophages can inhibit the anti-tumor effect of Lenvatinib on cholangiocarcinoma through immune regulation, which may be related to the tumor angiogenesis factor effect of M2 macrophage.
    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
    肺腺癌(LUAD)是最常见的肺癌类型。尽管以前对LUAD的免疫机制和相关分子进行了研究,这些分子在免疫微环境中的具体调控机制尚不清楚.此外,调节基因或RNA对LUAD转移和生存时间的影响尚待了解。为了弥补这些差距,我们收集了大量的数据,包括来自1018个样本的17226个基因表达谱,来自461个样本的370,640个甲基化位点,和来自513个样品的248个miRNA。我们的目标是探索基因,miRNA,以及与LUAD进展相关的甲基化位点。利用miRNA和甲基化位点的调控功能,我们确定了靶基因和调节基因。通过LASSO的使用和生存分析,我们确定了22个在LUAD免疫调节机制中起关键作用的关键基因。值得注意的是,这22个基因的表达水平在预测LUAD患者生存时间方面具有显著的辨别能力.此外,我们的深度学习模型利用这些基因的表达水平准确预测了LUAD患者的远处转移.进一步的途径富集分析显示,这22个基因在与LUAD进展密切相关的途径中显著富集。通过免疫浸润试验,我们观察到T细胞CD4记忆静息,单核细胞,和巨噬细胞。M2是LUAD免疫微环境中最丰富的三种细胞类型。已知这些细胞在肿瘤生长中起关键作用,入侵,和转移。单细胞数据分析进一步验证了这些基因的功能意义,表明它们不仅参与免疫细胞,还参与上皮细胞,表现出显著的差异表达。总的来说,本研究通过鉴定与LUAD进展相关的关键基因,揭示了LUAD免疫微环境的调控机制.这些发现提供了对潜在预后标志物和治疗靶标的见解。
    Lung adenocarcinoma (LUAD) is the most common type of lung cancer. Despite previous research on immune mechanisms and related molecules in LUAD, the specific regulatory mechanisms of these molecules in the immune microenvironment remain unclear. Furthermore, the impact of regulatory genes or RNA on LUAD metastasis and survival time is yet to be understood. To address these gaps, we collected a substantial amount of data, including 17,226 gene expression profiles from 1,018 samples, 370,640 methylation sites from 461 samples, and 248 miRNAs from 513 samples. Our aim was to explore the genes, miRNAs, and methylation sites associated with LUAD progression. Leveraging the regulatory functions of miRNAs and methylation sites, we identified target and regulated genes. Through the utilization of LASSO and survival analysis, we pinpointed 22 key genes that play pivotal roles in the immune regulatory mechanism of LUAD. Notably, the expression levels of these 22 genes demonstrated significant discriminatory power in predicting LUAD patient survival time. Additionally, our deep learning model accurately predicted distant metastasis in LUAD patients using the expression levels of these genes. Further pathway enrichment analysis revealed that these 22 genes are significantly enriched in pathways closely linked to LUAD progression. Through Immune Infiltration Assay, we observed that T cell CD4 memory resting, monocytes, and macrophages.M2 were the three most abundant cell types in the immune microenvironment of LUAD. These cells are known to play crucial roles in tumor growth, invasion, and metastasis. Single-cell data analysis further validated the functional significance of these genes, indicating their involvement not only in immune cells but also in epithelial cells, showcasing significant differential expression. Overall, this study sheds light on the regulatory mechanisms underlying the immune microenvironment of LUAD by identifying key genes associated with LUAD progression. The findings provide insights into potential prognostic markers and therapeutic targets.
    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
    已经在肝脏和非肝脏疾病中描述了粘膜相关的不变T(MAIT)细胞,它们被归因于抗菌,免疫调节,保护,和致病作用。这篇综述的目的是描述它们的生物学特性,表明他们参与了慢性肝病,并鼓励调查澄清他们的行为和治疗影响。英文摘要在PubMed中通过多个搜索词识别,并开发了参考书目。MAIT细胞被有限多样性的限制性非肽和多种炎性细胞因子激活。不同的促炎,抗炎,和免疫调节细胞因子被释放;感染的细胞被消除;记忆细胞出现。循环MAIT细胞被过度激活,免疫疲惫,功能失调,并耗尽慢性肝病。这种表型缺乏疾病特异性,它不能预测生物效应。MAIT细胞推测在慢性病毒性肝炎中具有保护作用,酒精性肝炎,非酒精性脂肪性肝病,原发性硬化性胆管炎,和失代偿期肝硬化.它们在自身免疫性肝炎中具有致病和促纤维化作用,在原发性胆汁性胆管炎中具有混合作用。肝微环境中的局部因素(细胞因子,胆汁酸,肠道来源的细菌抗原,和代谢副产物)可能会调节其在个体疾病中的反应。有必要对功能进行研究操作,以建立与疾病严重程度和结局的关联。总之,MAIT细胞构成一种非特异性疾病,对慢性肝脏炎症和感染的免疫反应。它们的病理作用已经从它们在活动性肝病期间的缺陷中推断出来,未来的调查必须澄清这个角色,把它和结果联系起来,并探索治疗干预措施。
    Mucosal-associated invariant T (MAIT) cells have been described in liver and non-liver diseases, and they have been ascribed antimicrobial, immune regulatory, protective, and pathogenic roles. The goals of this review are to describe their biological properties, indicate their involvement in chronic liver disease, and encourage investigations that clarify their actions and therapeutic implications. English abstracts were identified in PubMed by multiple search terms, and bibliographies were developed. MAIT cells are activated by restricted non-peptides of limited diversity and by multiple inflammatory cytokines. Diverse pro-inflammatory, anti-inflammatory, and immune regulatory cytokines are released; infected cells are eliminated; and memory cells emerge. Circulating MAIT cells are hyper-activated, immune exhausted, dysfunctional, and depleted in chronic liver disease. This phenotype lacks disease-specificity, and it does not predict the biological effects. MAIT cells have presumed protective actions in chronic viral hepatitis, alcoholic hepatitis, non-alcoholic fatty liver disease, primary sclerosing cholangitis, and decompensated cirrhosis. They have pathogenic and pro-fibrotic actions in autoimmune hepatitis and mixed actions in primary biliary cholangitis. Local factors in the hepatic microenvironment (cytokines, bile acids, gut-derived bacterial antigens, and metabolic by-products) may modulate their response in individual diseases. Investigational manipulations of function are warranted to establish an association with disease severity and outcome. In conclusion, MAIT cells constitute a disease-nonspecific, immune response to chronic liver inflammation and infection. Their pathological role has been deduced from their deficiencies during active liver disease, and future investigations must clarify this role, link it to outcome, and explore therapeutic interventions.
    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
    免疫性血小板减少症(ITP)是一种自身免疫介导的疾病,其特征是血小板计数降低。细胞因子在调节免疫应答中起重要作用,并参与许多自身免疫性疾病的发病机理。这项研究旨在探索在新诊断的ITP患者(治疗前后)和脾切除的ITP患者中发挥免疫调节功能的一组核心细胞因子的血清水平。使用Bio-Plex悬浮阵列系统和ELISA,检测血清IL-10、IL-21、IL-27、IL-33、IL-35、IL-37和TGF-β1水平。数据显示,新诊断的ITP患者血清中免疫调节细胞因子IL-10,IL-35和TGF-β1的水平显着降低。在具有完全应答或基于类固醇的治疗后应答的患者中,可以改善降低的细胞因子水平。治疗前后血清TGF-β1浓度与血小板计数呈正相关。所有检测到的免疫调节细胞因子,除IL-37外,脾切除ITP患者的水平明显高于治疗前ITP患者和健康对照组。总之,这些数据提示,在ITP的发病机制中,免疫调节细胞因子水平较低,并且在ITP行脾切除患者中,免疫调节细胞因子存在长期持续的过表达.
    Immune thrombocytopenia (ITP) is an autoimmune-mediated disease characterized by decreased platelet counts. Cytokines play important roles in modulating the immune response and are involved in the pathogenesis of many autoimmune diseases. This study aimed at exploring the serum levels of a core set of cytokines that exert immune-regulatory functions in newly diagnosed ITP patients (both before and after treatment) and splenectomized ITP patients. Using the Bio-Plex suspension array system and ELISA, the serum levels of IL-10, IL-21, IL-27, IL-33, IL-35, IL-37, and TGF-β1 were detected. The data showed that the serum levels of the immune regulatory cytokines IL-10, IL-35, and TGF-β1 were significantly lower in newly diagnosed ITP patients. Decreased cytokine levels could be improved in patients with a complete response or a response after steroid-based treatment(s). The serum concentrations of TGF-β1 were positively correlated with the platelet counts both before and after treatment. All the detected immune-regulatory cytokines, except IL-37, showed significantly higher levels in splenectomized ITP patients than pretreatment ITP patients and healthy controls. In conclusion, these data suggest that lower levels of immune-regulatory cytokines are involved in the pathogenesis of ITP and that there is a long-lasting overexpression of immune-regulatory cytokines in ITP patients with splenectomy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    目的:自身免疫性疾病是一组对自身抗原失去免疫耐受的异质性疾病。众所周知,不规则激发的T细胞参与病理性免疫应答。作为一种具有广阔应用前景的新型纳米材料,研究发现四面体骨架核酸(TFNA)纳米结构对T细胞具有免疫调节作用。
    方法:为了验证TFNA的成功制造,通过原子力显微镜(AFM)和动态光散射观察TFNA的形貌。在与从健康供体分离的CD3+T细胞共培养后,通过流式细胞术评估TFNA的调节作用。此外,研究了相关的信号通路。最后,我们在视神经脊髓炎谱系障碍(NMOSD)患者的T细胞上验证了我们的结果,这是一种典型的由T细胞诱导的自身免疫性疾病。
    结果:我们通过JNK信号通路揭示了TFNA在稳定状态的人原代T细胞中的替代调节功能。此外,通过抑制JNK和ERK磷酸化,TFNA对来自激发T细胞的IFNγ分泌表现出显著的抑制作用,而不影响TNF分泌。在NMOSD患者的自身反应性T细胞中也观察到TFNA的类似免疫调节作用。
    结论:总体而言,我们的结果揭示了TFNA在调节适应性免疫系统中的潜在应用,以及阐明T细胞介导的自身免疫性疾病的治疗。
    OBJECTIVE: Autoimmune diseases are a heterogeneous group of diseases which lose the immunological tolerance to self-antigens. It is well recognized that irregularly provoked T cells participate in the pathological immune responses. As a novel nanomaterial with promising applications, tetrahedral framework nucleic acid (TFNA) nanostructure was found to have immune regulatory effects on T cells in this study.
    METHODS: To verify the successful fabrication of TFNA, the morphology of TFNA was observed by atomic force microscopy (AFM) and dynamic light scattering. The regulatory effect of TFNA was evaluated by flow cytometry after cocultured with CD3+ T cells isolated from healthy donors. Moreover, the associated signaling pathways were investigated. Finally, we verified our results on the T cells from patients with neuromyelitis optica spectrum disorder (NMOSD), which is a typical autoimmune disease induced by T cells.
    RESULTS: We revealed the alternative regulatory functions of TFNA in human primary T cells with steady status via the JNK signaling pathway. Moreover, by inhibiting both JNK and ERK phosphorylation, TFNA exhibited significant suppressive effects on IFNγ secretion from provoking T cells without affecting TNF secretion. Similar immune regulatory effects of TFNA were also observed in autoreactive T cells from patients with NMOSD.
    CONCLUSIONS: Overall, our results revealed a potential application of TFNA in regulating the adaptive immune system, as well as shed a light on the treatment of T cell-mediated autoimmune diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    重度抑郁症(MDD)是一种常见的,具有异质性病因的严重和致残性神经精神障碍。在最广泛认可的病因模型中,免疫发病机制是主要的。大量研究表明,外周血中的炎症标志物水平异常,MDD患者的脑脊液(CSF)和脑。包括荟萃分析在内的多项研究报道了急性期蛋白的外周水平增加,和促炎细胞因子,特别是IL-1β,TNF-α,和IL-6在MDD中。死后脑研究同样证明了这些促炎细胞因子的表达上调。这以及单核细胞的证据,淋巴细胞和小胶质细胞激活,提示MDD中炎症反应系统(IRS)激活。一些研究表明,在MDD患者中,抗炎细胞因子水平升高或炎症途径缺陷以及T细胞成熟和反应缺陷。这表明存在补偿性免疫反应系统(CIRS),可以抵消IRS对重度抑郁症的影响。最近,据报道,MDD患者大脑中促炎和抗炎细胞因子的水平同时升高;这表明MDD中IRS和CIRS的活性。IRS和CIRS是总体系统的进化保守和整体元素。失调的IRS-CIRS系统在MDD的神经生物学构建体中的相关性刚刚开始被理解。人们普遍猜测,被破坏的IRS-CIRS元素可能决定发作,情节,神经进展过程,MDD患者的治疗反应和恢复。值得注意的是,激活的IRS-CIRS的特征可能是MDD的潜在生物标志物.在这里,已经尝试强调MDD中IRS-CIRS激活的生物学和病理学相关性,并提供对这些成分在药物治疗中的作用的见解.
    Major depressive disorder (MDD) is a common, severe and disabling neuropsychiatric disorder with a heterogenous etiology. Among the most widely recognized etiological models, immunopathogenesis is a predominant one. Numerous studies have demonstrated aberrant levels of inflammatory markers in the peripheral blood, cerebrospinal fluid (CSF) and brain of patients with MDD. Multiple studies including meta-analyses have reported increased peripheral levels of acute phase proteins, and pro-inflammatory cytokines, particularly IL-1β, TNF-α, and IL-6 in MDD. Postmortem brain studies similarly demonstrated upregulated expressions of these pro-inflammatory cytokines. This along with evidence of monocytic, lymphocytic and microglial activation, suggest an activated inflammatory response system (IRS) in MDD. A few studies show increased levels of anti-inflammatory cytokines or defective inflammatory pathways and a deficit in T cell maturation and responses in MDD patients. This suggests the presence of a Compensatory Immune Response System (CIRS), which can counterbalance the effects of IRS in major depression. More recently, simultaneously increased levels of both the pro-and anti-inflammatory cytokines are reported in the brain of MDD patients; this indicates activity of both the IRS and CIRS in MDD. The IRS and CIRS are the evolutionarily conserved and integral elements of an overarching system. The relevance of a dysregulated IRS-CIRS system in the neurobiological construct of MDD is just beginning to be understood. Speculation is rife that the disrupted IRS-CIRS elements might determine the onset, episodes, neuroprogressive processes, treatment response as well as recovery of patients with MDD. Notably, the signatures of an activated IRS-CIRS might emerge as potential biomarkers of MDD. Herein, an attempt has been made to highlight the biology and pathobiological relevance of IRS-CIRS activation in MDD and provide an insight into the role of these components in pharmacological therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    2019年冠状病毒病(COVID-19)大流行的爆发正在世界各地迅速蔓延。这种病毒,被称为SARS-CoV-2,已经感染了成千上万的人。根据症状,急性呼吸道疾病的发病机制与高度同质的冠状病毒以及其他病原体有关。证据表明高炎症率,氧化,压倒性的免疫反应可能导致COVID-19的病理。COVID-19引起细胞因子风暴,随后导致急性呼吸窘迫综合征(ARDS),往往以病人的死亡告终。间充质干细胞(MSCs)是通过自我更新能力识别的多潜能干细胞,克隆种群的产生,和多谱系分化。MSCs存在于身体的几乎所有组织中,在组织的修复和生成中起着至关重要的作用。此外,MSCs通过先天和适应性免疫系统中免疫细胞的相互作用具有广泛的免疫调节特性,导致许多效应子活性的免疫抑制。MSCs可以减少冠状病毒感染产生的细胞因子风暴。在一些研究中,这些细胞的给药对COVID-19患者有益。此外,MSCs可能能够改善肺纤维化和肺功能。在这次审查中,我们将回顾COVID-19患者基于MSC的免疫调节的最新研究结果.
    The outbreak of coronavirus disease 2019 (COVID-19) pandemic is quickly spreading all over the world. This virus, which is called SARS-CoV-2, has infected tens of thousands of people. Based on symptoms, the pathogenesis of acute respiratory illness is responsible for highly homogenous coronaviruses as well as other pathogens. Evidence suggests that high inflammation rates, oxidation, and overwhelming immune response probably contribute to pathology of COVID-19. COVID-19 causes cytokine storm, which subsequently leads to acute respiratory distress syndrome (ARDS), often ending up in the death of patients. Mesenchymal stem cells (MSCs) are multipotential stem cells that are recognized via self-renewal capacity, generation of clonal populations, and multilineage differentiation. MSCs are present in nearly all tissues of the body, playing an essential role in repair and generation of tissues. Furthermore, MSCs have broad immunoregulatory properties through the interaction of immune cells in both innate and adaptive immune systems, leading to immunosuppression of many effector activities. MSCs can reduce the cytokine storm produced by coronavirus infection. In a number of studies, the administration of these cells has been beneficial for COVID-19 patients. Also, MSCs may be able to improve pulmonary fibrosis and lung function. In this review, we will review the newest research findings regarding MSC-based immunomodulation in patients with COVID-19.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号