immunological synapse

免疫突触
  • 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
    Glofitamab,抗CD20抗体,被批准为复发性或难治性(r/r)弥漫性大细胞B淋巴瘤(DLBCL)的三线治疗,在近40%的患者中实现完全反应。这种人源化IgG1双特异性单克隆抗体结合恶性B淋巴细胞上的CD20和细胞毒性T细胞上的CD3。这种双重结合形成了免疫突触,激活T淋巴细胞并导致肿瘤细胞裂解。挽救性放疗对r/rDLBCL也有效,但它与glofitamab等全身治疗相结合可能会增加辐射诱导的毒性。我们报告了首例r/rDLBCL患者同时接受抢救放疗和glofitamab。一名被诊断为IV期DLBCL的68岁女性接受了R-CHOP的初始治疗,然后是汽车T细胞疗法,其次是glofitamab复发。在18FDG-PET/CT检测到早期代谢进展时,与glofitamab同时对难治性部位进行挽救性放疗。患者在放疗后出现轻度脊髓旁疼痛,但无其他明显毒性。治疗后三个月,她表现出完全的代谢反应,没有放疗毒性,正如PET-CT所证明的那样,也没有放射性肺炎的迹象.这种情况表明glofitamab联合抢救放疗是可以忍受的,并表明潜在的疗效。在r/rDLBCL的前瞻性研究中需要进一步调查。
    Glofitamab, an anti-CD20 antibody, is approved as a third-line treatment for relapsed or refractory (r/r) diffuse large-cell B lymphoma (DLBCL), achieving a complete response in nearly 40% of patients. This humanized IgG1 bispecific monoclonal antibody binds to CD20 on malignant B lymphocytes and to CD3 on cytotoxic T cells. This dual binding forms an immunological synapse, activating T lymphocytes and leading to the lysis of tumor cells. Salvage radiotherapy is also effective for r/r DLBCL, but its combination with systemic treatments like glofitamab may increase radiation-induced toxicity. We report the first case of a patient with r/r DLBCL receiving concurrent salvage radiotherapy and glofitamab. A 68-year-old female diagnosed with stage IV DLBCL underwent initial treatment with R-CHOP, then Car-T cell therapy, followed by glofitamab for recurrence. Upon early metabolic progression detected by 18FDG-PET/CT, salvage radiotherapy was administered to the refractory site concurrently with glofitamab. The patient experienced mild para-spinal pain post-radiotherapy but no other significant toxicities. Three months post-treatment, she showed a complete metabolic response with no radiotherapy toxicity, as evidenced by PET-CT, and no signs of radiation pneumonitis. This case indicates that combining glofitamab with salvage radiotherapy is tolerable and suggests potential efficacy, warranting further investigation in prospective studies for r/r DLBCL.
    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
    树突状细胞(DC)和T细胞之间的串扰在调节自然和病理条件下的免疫应答中起着至关重要的作用。DC-T电池串扰是通过接触依赖(即,免疫突触)和不依赖接触的机制(即,细胞因子)。活化的DC上调共刺激信号并分泌促炎细胞因子以协调T细胞活化和分化。相反,激活的T辅助细胞“许可”DCs走向成熟,而调节性T细胞(Tregs)沉默DC以引发耐受性免疫。可以利用有效调节DC-T细胞串扰的策略来促进用于癌症免疫疗法的免疫激活或用于治疗自身免疫疾病的免疫耐受。这里,我们回顾了DC和T细胞之间的自然串扰机制。我们重点介绍了调节DC-T细胞串扰的生物工程方法,包括常规疫苗,合成疫苗,和DC模仿,以及利用这些方法引导免疫反应治疗癌症和自身免疫性疾病的关键开创性研究。
    Crosstalk between dendritic cells (DCs) and T cells plays a crucial role in modulating immune responses in natural and pathological conditions. DC-T cell crosstalk is achieved through contact-dependent (i.e., immunological synapse) and contact-independent mechanisms (i.e., cytokines). Activated DCs upregulate co-stimulatory signals and secrete proinflammatory cytokines to orchestrate T cell activation and differentiation. Conversely, activated T helper cells \"license\" DCs towards maturation, while regulatory T cells (Tregs) silence DCs to elicit tolerogenic immunity. Strategies to efficiently modulate the DC-T cell crosstalk can be harnessed to promote immune activation for cancer immunotherapy or immune tolerance for the treatment of autoimmune diseases. Here, we review the natural crosstalk mechanisms between DC and T cells. We highlight bioengineering approaches to modulate DC-T cell crosstalk, including conventional vaccines, synthetic vaccines, and DC-mimics, and key seminal studies leveraging these approaches to steer immune response for the treatment of cancer and 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
    常规的1型树突状细胞(cDC1)是抗肿瘤T细胞应答的关键调节因子。癌组织中cDC1和CD8T细胞之间的细胞间接触的结构和丰度对于确定T细胞应答的结果是重要的。然而,在癌症进展过程中控制cDC1-CD8相互作用稳定性的分子决定簇仍未得到充分研究.这里,我们建立了一个非小细胞肺癌的遗传模型,该模型与荧光cDC1报告基因(KP-XCR1venus)交叉,以允许在不同肿瘤分期的肿瘤组织中检测cDC1-CD8T细胞簇.我们发现cDC1-CD8簇在肿瘤发展的早期阶段是丰富和有生产力的,但在晚期肿瘤中逐渐减少。转录谱分析和流式细胞术鉴定了粘附分子ALCAM/CD166(活化的白细胞粘附分子,CD6的配体)在肺cDC1中高度表达,在晚期肿瘤中显着下调。对人类数据集的分析表明,ALCAM在非小细胞肺癌中下调,其表达与更好的预后相关。机械上,在肺cDC1上触发ALCAM诱导细胞骨架重塑和接触形成,而其阻断阻止T细胞活化。一起,我们的结果表明,ALCAM是重要的稳定cDC1-CD8相互作用在早期肿瘤阶段,而其在晚期肿瘤中的损失有助于免疫逃避。
    Conventional type 1 dendritic cells (cDC1) are critical regulators of anti-tumoral T-cell responses. The structure and abundance of intercellular contacts between cDC1 and CD8 T cells in cancer tissues is important to determine the outcome of the T-cell response. However, the molecular determinants controlling the stability of cDC1-CD8 interactions during cancer progression remain poorly investigated. Here, we generated a genetic model of non-small cell lung cancer crossed to a fluorescent cDC1 reporter (KP-XCR1venus) to allow the detection of cDC1-CD8T cell clusters in tumor tissues across tumor stages. We found that cDC1-CD8 clusters are abundant and productive at the early stages of tumor development but progressively diminish in advanced tumors. Transcriptional profiling and flow cytometry identified the adhesion molecule ALCAM/CD166 (Activated Leukocyte Cell Adhesion Molecule, ligand of CD6) as highly expressed by lung cDC1 and significantly downregulated in advanced tumors. Analysis of human datasets indicated that ALCAM is downregulated in non-small cell lung cancer and its expression correlates to better prognosis. Mechanistically, triggering ALCAM on lung cDC1 induces cytoskeletal remodeling and contact formation whereas its blockade prevents T-cell activation. Together, our results indicate that ALCAM is important to stabilize cDC1-CD8 interactions at early tumor stages, while its loss in advanced tumors contributes to immune evasion.
    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
    适应性免疫应答包括通过由抗原呈递细胞(APC)表面上的主要组织相容性复合物(MHC)的蛋白质呈递的肽抗原激活T细胞。由于T细胞受体(TCR)与某种肽-MHC复合物有效地相互作用,专门的细胞-细胞连接,免疫突触,形成并伴随着细胞内信号分子的时空模式和功能的变化。在活化的T细胞中的血浆和内膜的细胞质小叶处发生的关键修饰包括影响脂质双层内或附近的蛋白质的结合和分布的脂质开关。这里,我们描述了在这个关键的水/膜界面起作用的两大类脂质开关。磷酸肌醇衍生自磷脂酰肌醇,一种两亲性分子,含有两条脂肪酸链和一个将甘油主链与碳水化合物肌醇桥接的磷酸基团。肌醇环可以通过专用激酶和磷酸盐可变地(去)磷酸化,从而创建定义信号分子的组成和性质的磷酸肌醇特征,分子复合物或整个细胞器。棕榈酰化是指脂肪酸棕榈酸酯与底物蛋白的半胱氨酸残基的可逆连接。DHHC酶,以其活性位点的四个保守氨基酸命名,催化这种翻译后修饰,从而改变蛋白质的分布,膜之间和膜内。T细胞利用这两种类型的分子开关来调整它们的特性以适应需要运动变化的激活过程,运输,分泌和基因表达。
    Adaptive immune responses comprise the activation of T cells by peptide antigens that are presented by proteins of the Major Histocompatibility Complex (MHC) on the surface of an antigen-presenting cell. As a consequence of the T cell receptor interacting productively with a certain peptide-MHC complex, a specialized cell-cell junction known as the immunological synapse forms and is accompanied by changes in the spatiotemporal patterning and function of intracellular signaling molecules. Key modifications occurring at the cytoplasmic leaflet of the plasma and internal membranes in activated T cells comprise lipid switches that affect the binding and distribution of proteins within or near the lipid bilayer. Here, we describe two major classes of lipid switches that act at this critical water/membrane interface. Phosphoinositides are derived from phosphatidylinositol, an amphiphilic molecule that contains two fatty acid chains and a phosphate group that bridges the glycerol backbone to the carbohydrate inositol. The inositol ring can be variably (de-)phosphorylated by dedicated kinases and phosphatases, thereby creating phosphoinositide signatures that define the composition and properties of signaling molecules, molecular complexes, or whole organelles. Palmitoylation refers to the reversible attachment of the fatty acid palmitate to a substrate protein\'s cysteine residue. DHHC enzymes, named after the four conserved amino acids in their active site, catalyze this post-translational modification and thereby change the distribution of proteins at, between, and within membranes. T cells utilize these two types of molecular switches to adjust their properties to an activation process that requires changes in motility, transport, secretion, and gene expression.
    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
    肿瘤采用各种策略来逃避免疫监视。中枢神经体系(CNS)具有克制免疫反响的多重特征。肿瘤和中枢神经系统是否具有相似的免疫抑制方案是难以捉摸的。这里,我们分析了接受曲妥珠单抗和抗PD-L1抗体的HER2+乳腺癌患者肿瘤的多组学数据,发现富含CNS的N-乙酰转移酶8样(NAT8L)及其代谢物N-乙酰天冬氨酸(NAA)在耐药肿瘤中过表达.在CNS中,NAA在脑炎症期间释放。NAT8L通过经由NAA抑制自然杀伤(NK)细胞和CD8+T细胞的细胞毒性来减轻脑部炎症并损害抗肿瘤免疫。NAA通过促进PCAF诱导的层粘连蛋白A-K542的乙酰化来破坏免疫突触的形成,从而抑制层粘连蛋白A和SUN2之间的整合并削弱裂解颗粒的极化。我们发现肿瘤细胞模拟CNS的抗炎机制以逃避抗肿瘤免疫,NAT8L是增强抗癌剂功效的潜在靶标。
    Tumors employ various strategies to evade immune surveillance. Central nervous system (CNS) has multiple features to restrain immune response. Whether tumors and CNS share similar programs of immunosuppression is elusive. Here, we analyze multi-omics data of tumors from HER2+ breast cancer patients receiving trastuzumab and anti-PD-L1 antibody and find that CNS-enriched N-acetyltransferase 8-like (NAT8L) and its metabolite N-acetylaspartate (NAA) are overexpressed in resistant tumors. In CNS, NAA is released during brain inflammation. NAT8L attenuates brain inflammation and impairs anti-tumor immunity by inhibiting cytotoxicity of natural killer (NK) cells and CD8+ T cells via NAA. NAA disrupts the formation of immunological synapse by promoting PCAF-induced acetylation of lamin A-K542, which inhibits the integration between lamin A and SUN2 and impairs polarization of lytic granules. We uncover that tumor cells mimic the anti-inflammatory mechanism of CNS to evade anti-tumor immunity and NAT8L is a potential target to enhance efficacy of anti-cancer agents.
    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
    过继性T细胞免疫疗法已成为治疗对传统癌症治疗已复发或变得难治(R/R)的癌症患者的相关方法。嵌合抗原受体(CAR)T细胞疗法改善了各种血液恶性肿瘤的生存率。然而,显著的局限性仍然阻碍了这些疗法在大多数癌症中的广泛采用.为了在这个领域取得进展,六个研究小组在马德里社区创建了“下一代CARTMAD联盟”(NEXTCART),旨在开发针对R/R和不良预后癌症的新型基于细胞的免疫疗法。在下一个cart,马德里的各种基础和转化研究小组和医院同意分享和协同他们在免疫疗法方面的基本专业知识,基因治疗,和免疫突触,以及儿科和成人肿瘤学的临床专业知识。NEXTCART的目标是为R/R成人和儿童肿瘤开发新的细胞工程方法和治疗方法,以在多中心临床试验中进行评估。这里,我们讨论了基于T细胞的疗法目前的局限性,并介绍了我们对未来发展的看法。发展机会包括开发同种异体产品,优化CAR信号域,结合细胞免疫疗法,多目标策略,改善肿瘤浸润淋巴细胞(TIL)/T细胞受体(TCR)治疗。此外,基础研究旨在识别新的肿瘤靶点,肿瘤微环境中影响CAR功效的肿瘤分子,以及增强免疫细胞和肿瘤细胞之间免疫突触效率的策略。我们对当前细胞免疫疗法的观点强调了这些治疗的潜力,同时承认需要创新解决方案来充分开发其癌症治疗潜力的现有障碍。
    Adoptive T cellular immunotherapies have emerged as relevant approaches for treating cancer patients who have relapsed or become refractory (R/R) to traditional cancer treatments. Chimeric antigen receptor (CAR) T-cell therapy has improved survival in various hematological malignancies. However, significant limitations still impede the widespread adoption of these therapies in most cancers. To advance in this field, six research groups have created the \"NEXT Generation CART MAD Consortium\" (NEXT CART) in Madrid\'s Community, which aims to develop novel cell-based immunotherapies for R/R and poor prognosis cancers. At NEXT CART, various basic and translational research groups and hospitals in Madrid concur to share and synergize their basic expertise in immunotherapy, gene therapy, and immunological synapse, and clinical expertise in pediatric and adult oncology. NEXT CART goal is to develop new cell engineering approaches and treatments for R/R adult and pediatric neoplasms to evaluate in multicenter clinical trials. Here, we discuss the current limitations of T cell-based therapies and introduce our perspective on future developments. Advancement opportunities include developing allogeneic products, optimizing CAR signaling domains, combining cellular immunotherapies, multi-targeting strategies, and improving tumor-infiltrating lymphocytes (TILs)/T cell receptor (TCR) therapy. Furthermore, basic studies aim to identify novel tumor targets, tumor molecules in the tumor microenvironment that impact CAR efficacy, and strategies to enhance the efficiency of the immunological synapse between immune and tumor cells. Our perspective of current cellular immunotherapy underscores the potential of these treatments while acknowledging the existing hurdles that demand innovative solutions to develop their potential for cancer treatment fully.
    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
    基于自然杀伤(NK)细胞的免疫疗法有望用于癌症治疗;然而,它的功效仍然有限,需要制定替代战略。这里,我们报告说维尼托克,FDA批准的BCL-2抑制剂,直接激活NK细胞,在体外和体内增强其对急性髓性白血病(AML)的细胞毒性,可能与BCL-2抑制无关。通过全面的方法,包括批量和单细胞RNA测序,亲和力测量,和功能测定,我们证明,维奈托克增加NK细胞对AML细胞的亲和力,并促进免疫突触(IS)形成过程中的溶解颗粒极化。值得注意的是,我们确定了一个独特的CD161低CD218bNK细胞亚群,对维奈托克治疗表现出显著的敏感性。此外,维奈托克通过NF-κB途径促进线粒体呼吸和ATP合成,从而促进NK细胞中的IS形成。总的来说,我们的研究结果确立了维奈托克作为NK细胞功能的多方面免疫代谢调节剂,并为增强基于NK细胞的癌症免疫治疗提供了一个有前景的策略.
    Natural killer (NK) cell-based immunotherapy holds promise for cancer treatment; however, its efficacy remains limited, necessitating the development of alternative strategies. Here, we report that venetoclax, an FDA-approved BCL-2 inhibitor, directly activates NK cells, enhancing their cytotoxicity against acute myeloid leukemia (AML) both in vitro and in vivo, likely independent of BCL-2 inhibition. Through comprehensive approaches, including bulk and single-cell RNA sequencing, avidity measurement, and functional assays, we demonstrate that venetoclax increases the avidity of NK cells to AML cells and promotes lytic granule polarization during immunological synapse (IS) formation. Notably, we identify a distinct CD161lowCD218b+ NK cell subpopulation that exhibits remarkable sensitivity to venetoclax treatment. Furthermore, venetoclax promotes mitochondrial respiration and ATP synthesis via the NF-κB pathway, thereby facilitating IS formation in NK cells. Collectively, our findings establish venetoclax as a multifaceted immunometabolic modulator of NK cell function and provide a promising strategy for augmenting NK cell-based cancer immunotherapy.
    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淋巴细胞微弱浸润的肿瘤对免疫疗法的反应较差。我们旨在揭示调节T细胞进入的恶性肿瘤相关程序,逮捕,和肿瘤环境中的激活。细胞粘附和组织结构程序的差异表达,特别是膜四跨膜蛋白claudin(CLDN)18作为签名基因的存在,从免疫耗尽的小鼠胰腺肿瘤中划分出免疫浸润。在人类胰腺导管腺癌(PDAC)和非小细胞肺癌中,CLDN18表达与分化程度高的组织学和良好的预后呈正相关。细胞表面的CLDN18促进细胞毒性T淋巴细胞(CTL)的积累,通过肌动蛋白驱动粘附蛋白ALCAM动员到肿瘤细胞膜的脂筏上,从而促进CTL与肿瘤细胞的直接接触。这个过程有利于在CTL和CLDN18阳性癌细胞之间形成强大的免疫突触(ISs)。导致T细胞活化增加。我们的数据揭示了CLDN18在协调T细胞浸润和塑造肿瘤免疫背景中的免疫作用。
    Tumors weakly infiltrated by T lymphocytes poorly respond to immunotherapy. We aimed to unveil malignancy-associated programs regulating T cell entrance, arrest, and activation in the tumor environment. Differential expression of cell adhesion and tissue architecture programs, particularly the presence of the membrane tetraspanin claudin (CLDN)18 as a signature gene, demarcated immune-infiltrated from immune-depleted mouse pancreatic tumors. In human pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer, CLDN18 expression positively correlated with more differentiated histology and favorable prognosis. CLDN18 on the cell surface promoted accrual of cytotoxic T lymphocytes (CTLs), facilitating direct CTL contacts with tumor cells by driving the mobilization of the adhesion protein ALCAM to the lipid rafts of the tumor cell membrane through actin. This process favored the formation of robust immunological synapses (ISs) between CTLs and CLDN18-positive cancer cells, resulting in increased T cell activation. Our data reveal an immune role for CLDN18 in orchestrating T cell infiltration and shaping the tumor immune contexture.
    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
    分子力越来越被认为是理解细胞信号传导过程的重要参数。近年来,积累的证据表明,在抗原识别过程中,T细胞也通过其T细胞受体施加拉力。为了测量这种细胞间拉力,可以利用蜘蛛丝肽的弹性特性,其作用类似于Hookean弹簧:增加的应变对应于施加到肽的增加的应力。结合福斯特共振能量转移(FRET)读出应变,这样的肽代表强大的和通用的纳米级力传感工具。在本文中,我们提供了一个详细的协议如何合成分子力传感器应用于T细胞抗原识别和实践指南的实验和分析获得的单分子FRET数据。
    Molecular forces are increasingly recognized as an important parameter to understand cellular signaling processes. In the recent years, evidence accumulated that also T-cells exert tensile forces via their T-cell receptor during the antigen recognition process. To measure such intercellular pulling forces, one can make use of the elastic properties of spider silk peptides, which act similar to Hookean springs: increased strain corresponds to increased stress applied to the peptide. Combined with Förster resonance energy transfer (FRET) to read out the strain, such peptides represent powerful and versatile nanoscopic force sensing tools. In this paper, we provide a detailed protocol how to synthesize a molecular force sensor for application in T-cell antigen recognition and hands-on guidelines on experiments and analysis of obtained single molecule FRET data.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号