Lymphocyte activation

淋巴细胞活化
  • 文章类型: Journal Article
    组蛋白脱乙酰酶1和2通过与无数共调节因子的相互作用在T调节(Treg)细胞的转录调节中起主要作用。Sin3a已被确立为Hdac1/2辅因子,虽然它在Tregs中的作用尚未确立。在这项研究中,评估了Sin3a在Foxp3+Tregs中条件性缺失的影响。Sin3a从Foxp3+Tregs的发育缺失导致致命的自身免疫的快速发作。Treg数量大大减少,而残留的Tregs抑制功能受损。小鼠还表现出效应T细胞活化,自身抗体生产,和广泛的组织损伤。机械上,Sin3a缺失导致Foxp3的转录降低,完全缺乏CNS2CpG去甲基化。此外,Foxp3蛋白稳定性随着ex-Treg群体的增加而受损。因此,Sin3a在Treg身份和功能的维持中起关键作用,并且对于Foxp3的表达和稳定性至关重要。
    Histone deacetylases 1 and 2 play a major role in the transcriptional regulation of T-regulatory (Treg) cells via interactions with a myriad of coregulatory factors. Sin3a has been well established as a Hdac1/2 cofactor, while its role within Tregs has not been established. In this study, the effects of conditional deletion of Sin3a within Foxp3+ Tregs were evaluated. Developmental deletion of Sin3a from Foxp3+ Tregs resulted in the rapid onset of fatal autoimmunity. Treg numbers were greatly reduced, while residual Tregs had impaired suppressive function. Mice also showed effector T-cell activation, autoantibody production, and widespread tissue injury. Mechanistically, Sin3a deletion resulted in decreased transcription of Foxp3 with a complete lack of CNS2 CpG demethylation. In addition, Foxp3 protein stability was impaired with an increased ex-Treg population. Thus, Sin3a plays a critical role in the maintenance of Treg identity and function and is essential for the expression and stability of Foxp3.
    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
    T细胞重新连接它们的代谢活动以满足免疫反应的需求,但是如何通过代谢调节因子在激活的T细胞中协调免疫反应是未知的。这里,我们确定自分泌VEGF-B是一种代谢调节因子,可控制脂质合成并维持活化T细胞存活的线粒体内膜完整性.T细胞中自分泌VEGF-B信号的中断减少了心磷脂质量,导致线粒体损伤,细胞凋亡增加,记忆发育减少。添加心磷脂或调节VEGF-B信号可改善T细胞线粒体适应性和存活率。自分泌VEGF-B信号通过GA结合蛋白α(GABPα)诱导sentrin/SUMO特异性蛋白酶2(SENP2)表达,进一步去SUMO化PPARγ并增强磷脂合成,导致活化T细胞的心磷脂增加。这些数据表明自分泌VEGF-B介导信号以协调脂质合成和线粒体适合性与T细胞活化的存活和免疫应答。此外,T细胞中的自分泌VEGF-B信号传导提供了针对感染和肿瘤的治疗靶标以及治疗自身免疫性疾病的途径。
    T cells rewire their metabolic activities to meet the demand of immune responses, but how to coordinate the immune response by metabolic regulators in activated T cells is unknown. Here, we identified autocrine VEGF-B as a metabolic regulator to control lipid synthesis and maintain the integrity of the mitochondrial inner membrane for the survival of activated T cells. Disruption of autocrine VEGF-B signaling in T cells reduced cardiolipin mass, resulting in mitochondrial damage, with increased apoptosis and reduced memory development. The addition of cardiolipin or modulating VEGF-B signaling improved T cell mitochondrial fitness and survival. Autocrine VEGF-B signaling through GA-binding protein α (GABPα) induced sentrin/SUMO-specific protease 2 (SENP2) expression, which further de-SUMOylated PPARγ and enhanced phospholipid synthesis, leading to a cardiolipin increase in activated T cells. These data suggest that autocrine VEGF-B mediates a signal to coordinate lipid synthesis and mitochondrial fitness with T cell activation for survival and immune response. Moreover, autocrine VEGF-B signaling in T cells provides a therapeutic target against infection and tumors as well as an avenue for the treatment of autoimmune diseases.
    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
    针对非适应性免疫细胞的治疗策略目前正在临床开发中。γδT细胞是T细胞的小亚型(占总T细胞的1-10%),无需抗原呈递机制即可介导其效应子功能。并与先天细胞共享功能特性。在不同的γδT亚型中,抗Vγ9Vδ2T抗体在早期临床研究中报告了临床疗效的迹象.在这篇综述中,我们描述了这种非常规T细胞亚型的生物学,并提供了激活这些细胞的新型抗体的作用机制的见解。我们将专注于靶向BTN3A配体和双特异性γδT细胞衔接剂的抗体。我们将详细回顾这些策略的优势,包括克服检查点抑制剂耐药机制的潜力,或与激活经典T细胞的药物相比更充分的安全性。在人类的第一次研究中确定的限制和克服这些限制的策略将被修改和讨论。最后,将为未来的临床发展提出临床选择。
    Therapeutic strategies targeting non-adaptive immune cells are currently in clinical development. γδT cells are a small subtype of T cells (1-10% of total T cells) that mediate their effector function without the necessity of the antigen presenting machinery, and also share functional properties with innate cells. Among the different γδT subtypes, antibodies against Vγ9Vδ2T have reported signs of clinical efficacy in early clinical studies. In this review we describe the biology of this subtype of non-conventional T cells and provide insights into the mechanism of action of novel antibodies that activate these cells. We will focus on antibodies targeting the BTN3A ligand and bi-specific γδT cell engagers. We will review in detail the advantages of these strategies including the potential for overcoming mechanisms of resistance to check point inhibitors, or the much more adequate safety profile compared with agents activating classical T cells. Limitations identified during the first studies in humans and strategies to overcome them will be revised and discussed. Finally, clinical options for future clinical development will be suggested.
    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
    αβT细胞受体(TCR)主要识别与不健康细胞上的主要组织相容性复合物分子(pMHCs)结合的异常肽,在免疫监视过程中,通过放置在TCR-pMHC键上的物理负荷来增强特异性和敏感性。为了理解这种机械生物学,大量和稀疏排列的表位刺激的TCR(NP366-374/Db和PA224-233/Db,分别)用光学镊子研究了体内甲型流感病毒感染。虽然某些NP库CD8T淋巴细胞需要许多配体来激活,其他人是数字化的,只需要几个。相反,所有PATCR都以数字方式执行,表现出显著的键寿命通过持续增加,激励结构转变的凌空。最佳的数字性能在体内更优越,与ERK磷酸化相关,CD3损失,和体外激活标记物上调。鉴于新抗原阵列缺乏,数字TCR可能对免疫疗法至关重要.
    αβ T cell receptors (TCRs) principally recognize aberrant peptides bound to major histocompatibility complex molecules (pMHCs) on unhealthy cells, amplifying specificity and sensitivity through physical load placed on the TCR-pMHC bond during immunosurveillance. To understand this mechanobiology, TCRs stimulated by abundantly and sparsely arrayed epitopes (NP366-374/Db and PA224-233/Db, respectively) following in vivo influenza A virus infection were studied with optical tweezers. While certain NP repertoire CD8 T lymphocytes require many ligands for activation, others are digital, needing just few. Conversely, all PA TCRs perform digitally, exhibiting pronounced bond lifetime increases through sustained, energizing volleys of structural transitioning. Optimal digital performance is superior in vivo, correlating with ERK phosphorylation, CD3 loss, and activation marker up-regulation in vitro. Given neoantigen array paucity, digital TCRs are likely critical for immunotherapies.
    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
    B细胞抗原受体(BCR)的连接启动体液免疫。然而,没有适当共刺激的BCR信号使B细胞死亡而不是分化成免疫效应细胞。BCR激活如何耗尽潜在的自身反应性B细胞,同时启动从同源T淋巴细胞接受拯救和分化信号仍然未知。这里,我们使用基于质谱的蛋白质组学方法来鉴定胞质/核穿梭元件,并揭示转录因子EB(TFEB)作为驱动激活诱导的细胞凋亡的中央BCR控制变阻器,并通过支持B细胞迁移和抗原呈递同时促进共刺激拯救信号的接收。CD40共刺激可防止TFEB驱动的细胞死亡,在增强和延长TFEB的核停留时间的同时,这标志着记忆B细胞的抗原经历。在老鼠身上,TFEB塑造生发中心B细胞的转录景观。在生发中心内,TFEB通过调节趋化因子受体促进光区中心细胞的暗区进入,通过平衡Bcl-2/BH3-only家族成员的表达,整合抗原诱导的细胞凋亡与T细胞提供的CD40存活信号。因此,TFEB重新编程抗原引发的生发中心B细胞以决定细胞命运。
    Ligation of the B cell antigen receptor (BCR) initiates humoral immunity. However, BCR signaling without appropriate co-stimulation commits B cells to death rather than to differentiation into immune effector cells. How BCR activation depletes potentially autoreactive B cells while simultaneously primes for receiving rescue and differentiation signals from cognate T lymphocytes remains unknown. Here, we use a mass spectrometry-based proteomic approach to identify cytosolic/nuclear shuttling elements and uncover transcription factor EB (TFEB) as a central BCR-controlled rheostat that drives activation-induced apoptosis, and concurrently promotes the reception of co-stimulatory rescue signals by supporting B cell migration and antigen presentation. CD40 co-stimulation prevents TFEB-driven cell death, while enhancing and prolonging TFEB\'s nuclear residency, which hallmarks antigenic experience also of memory B cells. In mice, TFEB shapes the transcriptional landscape of germinal center B cells. Within the germinal center, TFEB facilitates the dark zone entry of light-zone-residing centrocytes through regulation of chemokine receptors and, by balancing the expression of Bcl-2/BH3-only family members, integrates antigen-induced apoptosis with T cell-provided CD40 survival signals. Thus, TFEB reprograms antigen-primed germinal center B cells for cell fate decisions.
    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
    细胞因子8(DOCK8)免疫缺陷综合征的特点是生发中心反应失败,涉及抗原特异性B细胞的增殖和阳性选择的过程。这里,我们描述了如何在免疫小鼠生发中心的光区检查点阻断DOCK8缺陷B细胞,它们无法对T细胞依赖性存活和选择信号做出反应,从而分化成浆细胞或记忆B细胞。尽管缺乏DOCK8的B细胞可以获得并呈递抗原以启动同源T细胞的激活,整合素上调,B细胞-T细胞缀合物形成,当抗原可用性有限时,共刺激不足以持续激活B细胞和T细胞。我们的发现为DOCK8免疫缺陷综合征的体液反应失败提供了解释,并深入了解了可用抗原水平如何调节B细胞-T细胞串扰以微调体液免疫反应和免疫记忆。
    Dedicator of cytokinesis 8 (DOCK8) immunodeficiency syndrome is characterized by a failure of the germinal center response, a process involving the proliferation and positive selection of antigen-specific B cells. Here, we describe how DOCK8-deficient B cells are blocked at a light-zone checkpoint in the germinal centers of immunized mice, where they are unable to respond to T cell-dependent survival and selection signals and consequently differentiate into plasma cells or memory B cells. Although DOCK8-deficient B cells can acquire and present antigen to initiate activation of cognate T cells, integrin up-regulation, B cell-T cell conjugate formation, and costimulation are insufficient for sustained B cell and T cell activation when antigen availability is limited. Our findings provide an explanation for the failure of the humoral response in DOCK8 immunodeficiency syndrome and insight into how the level of available antigen modulates B cell-T cell cross-talk to fine-tune humoral immune responses and immunological memory.
    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:EB病毒(EBV)感染在鼻咽癌(NPC)的发病机制中起着重要作用。对于复发性或转移性鼻咽癌患者,对常规治疗有抵抗力,使用EBV特异性细胞毒性T细胞(EBV-CTL)的过继细胞疗法是有希望的选择。然而,长的生产期(约3至4周)和低EBV-CTL纯度(约占总CD8T细胞的40%)限制了EBV-CTL在临床上的应用.因此,本研究旨在建立快速生产EBV-CTL的方案。
    方法:用EBV特异性肽和白细胞介素(IL)-2、IL-15和干扰素α(IFN-α)培养来自EBV血清阳性供体的外周血单核细胞(PBMC)9天,我们发现IL-15可以增强IL-2介导的CTL激活,并显著提高CTL的产量.
    结果:当IFN-α用于IL-2/IL-15介导的CTL产生时,与来自IL-2/IL-15处理的EBV-CTL相比,EBV-CTL的生产率和EBV特异性细胞毒性显著增强。此外,IFN-α诱导的病毒特异性CTL的生产改善不仅是EBV-CTL的情况,而且是巨细胞病毒特异性CTL的情况。
    结论:我们建立了一种新的方案,可以从PBMC中快速扩展高纯度EBV-CTL,它可以在9天内产生EBV-CTL,并且在培养过程中不需要饲养细胞。
    BACKGROUND: Lytic Epstein-Barr virus (EBV) infection plays a major role in the pathogenesis of nasopharyngeal carcinoma (NPC). For patients with recurrent or metastatic NPC and resistant to conventional therapies, adoptive cell therapy using EBV-specific cytotoxic T cells (EBV-CTLs) is a promising option. However, the long production period (around 3 to 4 weeks) and low EBV-CTL purity (approximately 40% of total CD8 T cells) in the cell product limits the application of EBV-CTLs in clinics. Thus, this study aimed to establish a protocol for the rapid production of EBV-CTLs.
    METHODS: By culturing peripheral blood mononuclear cells (PBMCs) from EBV-seropositive donors with EBV-specific peptides and interleukin (IL)-2, IL-15, and interferon α (IFN-α) for 9 days, we identified that IL-15 can enhance IL-2-mediated CTL activation and significantly increase the yield of CTLs.
    RESULTS: When IFN-α was used in IL-2/IL-15-mediated CTL production from days 0 to 6, the productivity of EBV-CTLs and EBV-specific cytotoxicity significantly were reinforced relative to EBV-CTLs from IL-2/IL-15 treatment. Additionally, IFN-α-induced production improvement of virus-specific CTLs was not only the case for EBV-CTLs but also for cytomegalovirus-specific CTLs.
    CONCLUSIONS: We established a novel protocol to rapidly expand highly pure EBV-CTLs from PBMCs, which can produce EBV-CTLs in 9 days and does not require feeder cells during cultivation.
    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
    CD8+细胞毒性T淋巴细胞(CTL)在防御病毒感染和肿瘤方面非常有效。它们通过T细胞受体(TCR)识别肽-MHC-I复合物和共刺激而被激活。这种同源相互作用促进涉及细胞骨架重排的紧密细胞-细胞连接的组织,以实现效应子功能和靶细胞的清除。这是裂解颗粒不对称运输和动员到细胞-细胞接触的关键,促进裂解介质如颗粒酶和穿孔素的定向分泌。线粒体通过控制钙通量等过程在调节CTL功能中发挥作用,通过氧化磷酸化提供必要的能量,以及它自己在70S核糖体上的蛋白质翻译。然而,在成熟的CTL中尚未研究过急性抑制细胞溶质翻译在TCR后的快速反应中的作用。
    这里,我们研究了早期TCR激活和CD28共刺激对细胞骨架动态重组后人类CTL中胞质蛋白合成的重要性,线粒体,和裂解颗粒通过环己酰亚胺对80S核糖体的短期化学抑制和嘌呤霉素对80S和70S的短期化学抑制。
    我们观察到真核核糖体功能是允许微管蛋白细胞骨架和线粒体的适当不对称重组以及在人原代CTL中TCR激活后的早期mTOR途径激活所必需的。
    胞质蛋白翻译是在TCR激活时增加葡萄糖代谢和脱粒能力并因此调节人CTL的全部效应子功能所必需的。
    UNASSIGNED: CD8+ cytotoxic T lymphocytes (CTLs) are highly effective in defending against viral infections and tumours. They are activated through the recognition of peptide-MHC-I complex by the T-cell receptor (TCR) and co-stimulation. This cognate interaction promotes the organisation of intimate cell-cell connections that involve cytoskeleton rearrangement to enable effector function and clearance of the target cell. This is key for the asymmetric transport and mobilisation of lytic granules to the cell-cell contact, promoting directed secretion of lytic mediators such as granzymes and perforin. Mitochondria play a role in regulating CTL function by controlling processes such as calcium flux, providing the necessary energy through oxidative phosphorylation, and its own protein translation on 70S ribosomes. However, the effect of acute inhibition of cytosolic translation in the rapid response after TCR has not been studied in mature CTLs.
    UNASSIGNED: Here, we investigated the importance of cytosolic protein synthesis in human CTLs after early TCR activation and CD28 co-stimulation for the dynamic reorganisation of the cytoskeleton, mitochondria, and lytic granules through short-term chemical inhibition of 80S ribosomes by cycloheximide and 80S and 70S by puromycin.
    UNASSIGNED: We observed that eukaryotic ribosome function is required to allow proper asymmetric reorganisation of the tubulin cytoskeleton and mitochondria and mTOR pathway activation early upon TCR activation in human primary CTLs.
    UNASSIGNED: Cytosolic protein translation is required to increase glucose metabolism and degranulation capacity upon TCR activation and thus to regulate the full effector function of human CTLs.
    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:SARS-CoV-2感染引起的妊娠免疫反应尚不清楚。我们旨在获取和比较健康妊娠和感染SARS-CoV-2的孕妇之间的抗病毒细胞反应和淋巴细胞激活。方法:检测健康非妊娠妇女外周血淋巴细胞的免疫学变化,非孕妇COVID-19,健康孕妇,流式细胞术检测COVID-19孕妇和康复组。体外阻断用于鉴定通过ICOS-ICOSL途径的NKT样细胞活化。结果:我们发现,与健康孕妇相比,COVID-19阳性孕妇的CD3+CD56+NKT样细胞明显降低。SARS-CoV-2感染后,孕妇的NKT样细胞表达较高水平的活化受体CD69和NKp46。特别是,它们还增加了共刺激分子ICOS的表达。COVID-19孕妇NKT样细胞上调IFN-γ表达,CD107a和Ki67。同时,我们发现,在患有COVID-19的孕妇中,ICOSL在pDC上的表达显着增加。体外阻断ICOS可显著降低COVID-19阳性孕妇NKT样细胞的抗病毒活性,提示ICOS-ICOSL可能在NKT样细胞清除病毒中发挥重要作用。结论:SARS-CoV-2感染期间,孕妇NKT样细胞通过ICOS-ICOSL途径激活,在抗病毒应答中发挥重要作用。
    Objective: The immune response initiated by SARS-CoV-2 infection in pregnancy is poorly elucidated. We aimed to access and compare the antiviral cellular responses and lymphocytes activation between healthy pregnancies and pregnant women infected with SARS-CoV-2. Methods: We detected the immunological changes of lymphocytes in peripheral blood of healthy non-pregnant women, non-pregnant women with COVID-19, healthy pregnant women, pregnant women with COVID-19 and convalescent group by flow cytometry. In vitro blockade was used to identify NKT-like cell activation through ICOS-ICOSL pathway. Results: We found that CD3+CD56+ NKT-like cells decreased significantly in COVID-19 positive pregnant women compared to healthy pregnant women. NKT-like cells of pregnant women expressed higher level of activating receptors CD69 and NKp46 after SARS-CoV-2 infection. Particularly, they also increased the expression of the co-stimulatory molecule ICOS. NKT-like cells of pregnant women with COVID-19 up-regulated the expression of IFN-γ, CD107a and Ki67. Meanwhile, we found that ICOSL expression was significantly increased on pDCs in pregnant women with COVID-19. Blocking ICOS in vitro significantly decreased the antiviral activity of NKT-like cells in COVID-19 positive pregnant women, suggesting that ICOS-ICOSL may play an important role in the virus clearance by NKT-like cells. Conclusions: During SARS-CoV-2 infection, NKT-like cells of pregnant women activated through ICOS-ICOSL pathway and played an important role in the antiviral response.
    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:重症肌无力(MG)是一种自身免疫性疾病,其特征是针对神经肌肉接头结构的致病性抗体。证据表明,由转录因子(TF)介导的长链非编码RNA(lncRNA)的调节在MG的病理生理学中起着关键作用。然而,MG中lncRNAs的详细分子机制仍未确定。
    方法:使用微阵列分析,我们分析了MG中的lncRNA水平。通过生物信息学分析,发现LINC01566可能在MG中起重要作用。首先,进行qRT-PCR以验证LINC1566在MG患者中的表达。然后,进行荧光原位杂交以确定LINC01566在CD4+T细胞中的定位。最后,我们还使用流式细胞术和CCK-8试验分析了LINC01566敲低或过表达对CD4+T细胞功能的影响.使用双荧光素酶报告基因测定来验证TFFOSL1与LINC01566启动子的结合。
    结果:基于lncRNA微阵列和差异表达分析,我们鉴定了563个差异表达(DE)lncRNAs,MG中450个DEmRNA和19个DETFs。然后我们构建了一个lncRNA-TF-mRNA网络。通过网络分析,我们发现LINC01566可能通过调节T细胞相关通路在MG中发挥关键作用.进一步的实验表明LINC01566在MG患者中以低水平表达。功能上,LINC01566主要分布在细胞核中,可以促进CD4+T细胞凋亡,抑制细胞增殖。机械上,我们假设LINC01566可能负向调节DUSP3、CCR2、FADD、SIRPB1,LGALS3和SIRPB1参与T细胞活化途径,进一步影响MG细胞增殖和凋亡。此外,我们发现LINC01566对MG中CD4+T细胞的作用是由TFFOSL1介导的,体外实验表明FOSL1可以与LINC01566的启动子区结合。
    结论:总之,我们的研究揭示了LINC01566在临床样本和细胞实验中的保护作用,说明FOSL1/LINC01566负调节MG中CD4+T细胞活化的潜在作用和机制。
    BACKGROUND: Myasthenia gravis (MG) is an autoimmune disease characterized by pathogenic antibodies that target structures of the neuromuscular junction. The evidence suggests that the regulation of long noncoding RNAs (lncRNAs) that is mediated by transcription factors (TFs) plays a key role in the pathophysiology of MG. Nevertheless, the detailed molecular mechanisms of lncRNAs in MG remain largely undetermined.
    METHODS: Using microarray analysis, we analyzed the lncRNA levels in MG. By bioinformatics analysis, LINC01566 was found to potentially play an important role in MG. First, qRT‒PCR was performed to verify the LINC1566 expressions in MG patients. Then, fluorescence in situ hybridization was conducted to determine the localization of LINC01566 in CD4 + T cells. Finally, the impact of LINC01566 knockdown or overexpression on CD4 + T-cell function was also analyzed using flow cytometry and CCK-8 assay. A dual-luciferase reporter assay was used to validate the binding of the TF FOSL1 to the LINC01566 promoter.
    RESULTS: Based on the lncRNA microarray and differential expression analyses, we identified 563 differentially expressed (DE) lncRNAs, 450 DE mRNAs and 19 DE TFs in MG. We then constructed a lncRNA-TF-mRNA network. Through network analysis, we found that LINC01566 may play a crucial role in MG by regulating T-cell-related pathways. Further experiments indicated that LINC01566 is expressed at low levels in MG patients. Functionally, LINC01566 is primarily distributed in the nucleus and can facilitate CD4 + T-cell apoptosis and inhibit cell proliferation. Mechanistically, we hypothesized that LINC01566 may negatively regulate the expressions of DUSP3, CCR2, FADD, SIRPB1, LGALS3 and SIRPB1, which are involved in the T-cell activation pathway, to further influence the cellular proliferation and apoptosis in MG. Moreover, we found that the effect of LINC01566 on CD4 + T cells in MG was mediated by the TF FOSL1, and in vitro experiments indicated that FOSL1 can bind to the promoter region of LINC01566.
    CONCLUSIONS: In summary, our research revealed the protective roles of LINC01566 in clinical samples and cellular experiments, illustrating the potential roles and mechanism by which FOSL1/LINC01566 negatively regulates CD4 + T-cell activation in MG.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号