inhibitory receptor

抑制性受体
  • 文章类型: Journal Article
    人类染色体19q13.4上的白细胞免疫球蛋白(Ig)样受体(LILRs)编码11个免疫球蛋白超家族受体,在人群内部和人群之间表现出遗传多样性。在LILR基因中,LILRB3和LILRA6周围的基因组区域由于其显著的序列同源性而尚未被完全表征,这使得很难区分它们。为了检查LILRB3和LILRA6基因组区域,一个名为JoGo-LILRCNCaller的工具,可以从短阅读全基因组测序(srWGS)数据中调用拷贝数,应用于包含2,504个样本的广泛的国际srWGS数据集。在这个过程中,在3个样本中检测到以前未报告的LILRB3和LILRA6丢失.使用这些样本的长读数测序,我们在日本人群的LILRB3和LILRA6基因组区域中发现了一个新的大缺失(33,692bp)。这个缺失跨越了三个基因,LILRB3,LILRA6和LILRB5,导致LILRB3外显子12-13位于LILRB5外显子1-12的下游,而LILRA6的丢失,表明LILRB5和LILRB3(LILRB5-3)之间的杂合基因的潜在表达。还验证了LILRB5-3杂合基因的转录和随后的翻译。杂合连接位于胞内结构域内,导致LILRB5胞外结构域与具有三个免疫受体基于酪氨酸的抑制基序(ITIM)的部分LILRB3胞内结构域融合,表明LILRB5-3获得了一种新的信号功能。将JoGo-LILR工具进一步应用于srWGS样品表明CEU群体中存在LILRB5-3杂合基因。我们的发现为LILR家族的遗传和功能多样性提供了见解。
    Leukocyte immunoglobulin (Ig)-like receptors (LILRs) on human chromosome 19q13.4 encode 11 immunoglobulin superfamily receptors, exhibiting genetic diversity within and between human populations. Among the LILR genes, the genomic region surrounding LILRB3 and LILRA6 has yet to be fully characterized due to their significant sequence homology, which makes it difficult to differentiate between them. To examine the LILRB3 and LILRA6 genomic region, a tool named JoGo-LILR CN Caller, which can call copy number from short-read whole genome sequencing (srWGS) data, was applied to an extensive international srWGS dataset comprising 2,504 samples. During this process, a previously unreported loss of both LILRB3 and LILRA6 was detected in three samples. Using long-read sequencing of these samples, we have discovered a novel large deletion (33,692 bp) in the LILRB3 and LILRA6 genomic regions in the Japanese population. This deletion spanned three genes, LILRB3, LILRA6, and LILRB5, resulting in LILRB3 exons 12-13 being located immediately downstream of LILRB5 exons 1-12 with the loss of LILRA6, suggesting the potential expression of a hybrid gene between LILRB5 and LILRB3 (LILRB5-3). Transcription and subsequent translation of the LILRB5-3 hybrid gene were also verified. The hybrid junction was located within the intracellular domain, resulting in an LILRB5 extracellular domain fused to a partial LILRB3 intracellular domain with three immunoreceptor tyrosine-based inhibitory motifs (ITIMs), suggesting that LILRB5-3 acquired a novel signaling function. Further application of the JoGo-LILR tool to srWGS samples suggested the presence of the LILRB5-3 hybrid gene in the CEU population. Our findings provide insight into the genetic and functional diversity of the LILR family.
    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:幼年特发性关节炎(JIA)是儿童最常见的风湿性疾病,其发病机制与T细胞活化有关。T细胞活化可以通过抑制性受体(IR)如CTLA-4、PD-1、LAG-3和TIM-3产生的信号平衡。这里,我们确定了IRs在不同JIA亚型发病机制中的作用。
    方法:总共,我们纳入了67例少关节JIA,12IgM-RF阴性多关节JIA,17附着炎相关关节炎,11例全身JIA患者和10例健康对照。我们从患者的疾病发作或发作期间收集了血浆(和滑液)样品。我们测量了co-IR(IL-2Rα,4-1BB,CD86,TGF-β1,CTLA-4,PD-L1,PD-1,TIM-3,LAG-3,Galectin-9)通过细胞计数珠阵列试剂盒及其细胞表达(PD-1,CTLA-4,TIM-3,LAG-3)通过流式细胞术。我们比较了不同JIA亚组中不同co-IR的血浆水平和细胞表达。
    结果:多关节JIA组与其他三个接受检查的JIA亚组不同,血浆sCTLA-4水平较高(p<0.001),sPD-1(p<0.05),与其他JIA亚组和健康对照相比,s4-1BB(p<0.05)。我们分析了不同JIA亚型的PBMC上不同co-IR的细胞表面表达。类似于血浆水平,poly-JIA亚组CTLA4表达的百分比(p<0.05)和MFI(平均荧光强度)(p<0.01)均较高。
    结论:这是研究JIA不同亚型中不同co-IR表达谱的首次报道。多关节JIA患者有不同的共同红外光谱,血浆中的CTLA-4,PD-1和4-1BB比其他JIA亚型多。
    OBJECTIVE: Juvenile Idiopathic Arthritis (JIA) is the most common rheumatic disease of childhood; the pathogenesis is associated with T cell activation. T cell activation can be counter-balanced by signals generated by inhibitory receptors (IRs) such as CTLA-4, PD-1, LAG-3, and TIM-3. Here, we identify the role of IRs in the pathogenesis of different JIA subtypes.
    METHODS: In total, we included 67 oligoarticular JIA, 12 IgM-RF negative polyarticular JIA, 17 enthesitis related arthritis, 11 systemic JIA patients and 10 healthy controls. We collected plasma (and synovial fluid) samples from the patients either at the onset or during a flare of their disease. We measured the soluble levels of co-IRs (IL-2Rα, 4-1BB, CD86, TGF-β1, CTLA-4, PD-L1, PD-1, TIM-3, LAG- 3, Galectin-9) by cytometric bead array kits and their cellular expression (PD-1, CTLA-4, TIM-3, LAG-3) by flow cytometry. We compared the plasma levels and cellular expressions of different co-IRs within different JIA subgroups.
    RESULTS: The polyarticular-JIA group was different from the three other examined JIA subgroups, having higher levels of plasma sCTLA-4(p< 0.001), sPD-1(p< 0.05), and s4-1BB(p< 0.05) when compared with the other JIA subgroups and healthy controls. We analyzed the cellular surface expression of different co-IRs on the PBMCs of different JIA subtypes. Similar to plasma levels, both the percentage(p< 0.05) and the MFI (mean fluorescence intensity) (p< 0.01) of CTLA4 expression were higher in the poly-JIA subgroup.
    CONCLUSIONS: This is the first report studying the expression profile of different co-IRs in different subtypes of JIA. Polyarticular JIA patients had a different co-IR profile, having more CTLA-4, PD-1 and 4-1BB in their plasma than the other subtypes of JIA.
    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
    抑制性自然杀伤(NK)细胞受体识别靶细胞上反式的I类MHC(MHC-I)并抑制细胞毒性。一些NK细胞受体识别顺式MHC-I,但是这种相互作用的作用是不确定的。Ly49Q,在非NK细胞中表达的非典型Ly49受体,顺式结合MHC-I并介导嗜中性粒细胞的趋化性和浆细胞样树突状细胞产生I型干扰素。我们在Ly49Q的近膜区域中鉴定了脂质结合基序,并发现Ly49Q通过硫酸盐结合组织了包含鞘脂的功能性膜结构域。Ly49Q将肌动蛋白重塑分子招募到基于免疫受体酪氨酸的抑制基序,这使鞘脂富集的膜结构域能够在吞噬作用期间介导层状足和吞噬体膜上复杂的肌动蛋白重塑。因此,Ly49Q促进蛋白质和脂质种类的整合调节以构建细胞类型特异性膜平台。其他Ly49成员具有脂质结合基序;因此,膜平台组织可能是一些NK细胞受体的主要作用。
    Inhibitory natural killer (NK) cell receptors recognize MHC class I (MHC-I) in trans on target cells and suppress cytotoxicity. Some NK cell receptors recognize MHC-I in cis, but the role of this interaction is uncertain. Ly49Q, an atypical Ly49 receptor expressed in non-NK cells, binds MHC-I in cis and mediates chemotaxis of neutrophils and type I interferon production by plasmacytoid dendritic cells. We identified a lipid-binding motif in the juxtamembrane region of Ly49Q and found that Ly49Q organized functional membrane domains comprising sphingolipids via sulfatide binding. Ly49Q recruited actin-remodeling molecules to an immunoreceptor tyrosine-based inhibitory motif, which enabled the sphingolipid-enriched membrane domain to mediate complicated actin remodeling at the lamellipodia and phagosome membranes during phagocytosis. Thus, Ly49Q facilitates integrative regulation of proteins and lipid species to construct a cell type-specific membrane platform. Other Ly49 members possess lipid binding motifs; therefore, membrane platform organization may be a primary role of some NK cell receptors.
    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
    在过继性T细胞疗法中,经工程改造的肿瘤特异性T细胞治疗的患者的长期治疗益处受到输注细胞产物缺乏长期持久性和肿瘤微环境中活跃的免疫抑制机制的限制.浸润肿瘤的耗尽的T细胞的特征在于由多种抑制性受体(IR)触发的效应子功能的丧失。在患者中,IR阻断逆转T细胞耗尽,但选择性低,可能释放自身反应性克隆并导致临床自身免疫副作用。此外,细胞治疗中长期保护性免疫力的丧失归因于输注细胞的效应记忆表型。
    我们同时通过TCR基因编辑(TCRED)将T细胞特异性重定向到NY-ESO-1抗原,并通过CRISPR/Cas9永久破坏LAG3,TIM-3或2B4基因(IRKO),以扩增早期分化的长寿命记忆T干细胞。TCRED-IRKO和IR感受态(TCRED-IRCOMP)细胞的效应子功能在短期共培养测定中和在体外慢性刺激设置下进行测试。最后,在多发性骨髓瘤异种移植模型中评估了所开发细胞产物的疗效.
    我们表明,在慢性刺激下,TCRED-IRKO细胞在通过不同机制抵抗功能衰竭方面优于TCRED-IRCOMP细胞,并在体内肿瘤再攻击时有效消除癌细胞。我们的数据表明,TIM-3和2B4破坏保留T细胞脱颗粒能力,而LAG-3破坏阻止T细胞中其他抑制性受体的上调。
    这些结果突出显示TIM-3、LAG-3和2B4破坏可增加肿瘤特异性细胞产物的治疗益处,IRs在抗肿瘤反应中的非冗余作用。
    In adoptive T cell therapy, the long term therapeutic benefits in patients treated with engineered tumor specific T cells are limited by the lack of long term persistence of the infused cellular products and by the immunosuppressive mechanisms active in the tumor microenvironment. Exhausted T cells infiltrating the tumor are characterized by loss of effector functions triggered by multiple inhibitory receptors (IRs). In patients, IR blockade reverts T cell exhaustion but has low selectivity, potentially unleashing autoreactive clones and resulting in clinical autoimmune side effects. Furthermore, loss of long term protective immunity in cell therapy has been ascribed to the effector memory phenotype of the infused cells.
    We simultaneously redirected T cell specificity towards the NY-ESO-1 antigen via TCR gene editing (TCRED) and permanently disrupted LAG3, TIM-3 or 2B4 genes (IRKO) via CRISPR/Cas9 in a protocol to expand early differentiated long-living memory stem T cells. The effector functions of the TCRED-IRKO and IR competent (TCRED-IRCOMP) cells were tested in short-term co-culture assays and under a chronic stimulation setting in vitro. Finally, the therapeutic efficacy of the developed cellular products were evaluated in multiple myeloma xenograft models.
    We show that upon chronic stimulation, TCRED-IRKO cells are superior to TCRED-IRCOMP cells in resisting functional exhaustion through different mechanisms and efficiently eliminate cancer cells upon tumor re-challenge in vivo. Our data indicate that TIM-3 and 2B4-disruption preserve T-cell degranulation capacity, while LAG-3 disruption prevents the upregulation of additional inhibitory receptors in T cells.
    These results highlight that TIM-3, LAG-3, and 2B4 disruptions increase the therapeutic benefit of tumor specific cellular products and suggest distinct, non-redundant roles for IRs in anti-tumor 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: Journal Article
    C型凝集素受体(CLRs)是模式识别受体家族,它通过小的碳水化合物识别结构域(CRD)检测广谱的配体。CLEC12A是一种抑制CLR,其识别晶体结构,例如尿酸单钠晶体。CLEC12A还识别霉菌酸,分枝杆菌细胞壁的主要组成部分,并抑制宿主的免疫反应。尽管CLEC12A可能是分枝杆菌感染的治疗靶标,CLEC12A的结构信息不可用.我们在这里报告了无配体形式的人CLEC12A的晶体结构,并与抑制性抗体50C1复合。50C1识别hCLEC12ACRD顶面上的人特异性残基。全面的丙氨酸扫描表明,霉菌酸和尿酸单钠晶体的配体结合位点可能相互重叠,这表明CLEC12A利用一个共同的界面来识别不同类型的配体。我们的结果为CLEC12A的阻断和配体识别机制提供了原子见解,并导致了CLR特异性抑制剂的设计。
    C-type lectin receptors (CLRs) are a family of pattern recognition receptors, which detect a broad spectrum of ligands via small carbohydrate-recognition domains (CRDs). CLEC12A is an inhibitory CLR that recognizes crystalline structures such as monosodium urate crystals. CLEC12A also recognizes mycolic acid, a major component of mycobacterial cell walls, and suppresses host immune responses. Although CLEC12A could be a therapeutic target for mycobacterial infection, structural information on CLEC12A was not available. We report here the crystal structures of human CLEC12A (hCLEC12A) in ligand-free form and in complex with 50C1, its inhibitory antibody. 50C1 recognizes human-specific residues on the top face of hCLEC12A CRD. A comprehensive alanine scan demonstrated that the ligand-binding sites of mycolic acid and monosodium urate crystals may overlap with each other, suggesting that CLEC12A utilizes a common interface to recognize different types of ligands. Our results provide atomic insights into the blocking and ligand-recognition mechanisms of CLEC12A and leads to the design of CLR-specific inhibitors.
    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
    T细胞可表达多种抑制性受体。在诱导T细胞耗竭以响应持久性抗原时,在抗肿瘤免疫反应中,许多是同时表达的。关键的抑制性受体是CTLA-4,PD-1,LAG3,TIM3和TIGIT,在这里调查。这些受体作为癌症免疫疗法中的中心治疗靶标是重要的。抑制性受体不在细胞表面组成型表达,但是大量的部分存在于细胞内囊泡结构中。仍未解决不同抑制性受体的亚细胞定位在多大程度上是不同的。使用亚细胞分布和质膜插入的定量成像,并辅以邻近蛋白质组学和生化分析抑制性受体与运输衔接子的关联,五种抑制性受体的亚细胞分布是离散的。CTLA-4的分布最为明显,与溶酶体来源的囊泡和分选nexin1/2/5/6运输机械优先相关。由于缺乏证据表明存在特定的囊泡亚型来解释不同的抑制性受体分布,我们建议,这种分布是由通过重叠的囊泡结构关节组的不同运输驱动的。五种抑制性受体的亚细胞定位的这种广泛表征为它们的贩运及其治疗开发的分子研究奠定了基础。
    T cells can express multiple inhibitory receptors. Upon induction of T cell exhaustion in response to a persistent antigen, prominently in the anti-tumor immune response, many are expressed simultaneously. Key inhibitory receptors are CTLA-4, PD-1, LAG3, TIM3, and TIGIT, as investigated here. These receptors are important as central therapeutic targets in cancer immunotherapy. Inhibitory receptors are not constitutively expressed on the cell surface, but substantial fractions reside in intracellular vesicular structures. It remains unresolved to which extent the subcellular localization of different inhibitory receptors is distinct. Using quantitative imaging of subcellular distributions and plasma membrane insertion as complemented by proximity proteomics and biochemical analysis of the association of the inhibitory receptors with trafficking adaptors, the subcellular distributions of the five inhibitory receptors were discrete. The distribution of CTLA-4 was most distinct, with preferential association with lysosomal-derived vesicles and the sorting nexin 1/2/5/6 transport machinery. With a lack of evidence for the existence of specific vesicle subtypes to explain divergent inhibitory receptor distributions, we suggest that such distributions are driven by divergent trafficking through an overlapping joint set of vesicular structures. This extensive characterization of the subcellular localization of five inhibitory receptors in relation to each other lays the foundation for the molecular investigation of their trafficking and its therapeutic exploitation.
    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
    自然杀伤(NK)细胞具有将细胞疗法从复杂的自体选择转变为通用的现成选择的潜力。尽管NK细胞在白血病的治疗中已经证明了疗效和安全性,基于NK细胞的免疫疗法对实体瘤的疗效有限仍然是一个主要障碍.在免疫抑制肿瘤微环境(TME)中,癌症和免疫细胞之间的抑制性相互作用损害抗肿瘤免疫。KLRC1基因编码NK细胞抑制性受体NKG2A,这是一个有效的NK细胞免疫检查点。NKG2A特异性结合HLA-E,一种在肿瘤中经常过表达的非经典HLAI类分子,导致强烈损害NK细胞功能的抑制信号的传递。
    为了恢复NK细胞对HLA-E+肿瘤的细胞毒性,我们通过使用CRISPR介导的KLRC1基因编辑靶向NKG2A/HLA-E免疫检查点.
    KLRC1敲除导致细胞分选后离体扩增的人NK细胞中NKG2A+细胞频率减少81%。体外,肿瘤细胞过度表达HLA-E显著抑制野生型(WT)NK细胞的细胞毒性,p值范围为0.0071~0.0473,这取决于肿瘤细胞系.相比之下,与WTNK细胞相比,KLRC1KONK细胞对四种不同的HLA-E实体瘤细胞系表现出明显更高的细胞毒性,p值范围从<0.0001到0.0154。有趣的是,编辑的NK细胞群中43.5%至60.2%的NKG2A-NK细胞比例足以最大程度地逆转HLA-E介导的NK细胞的细胞毒性抑制。活化受体NKG2C的表达在KLRC1KONK细胞中增加,并有助于改善NK细胞对HLA-E肿瘤的细胞毒性。在体内,人类KLRC1KONK细胞的过继转移在HLA-E+转移性乳腺癌的异种小鼠模型中显著延迟肿瘤进展和增加生存率,与WTNK细胞相比(p=0.0015)。
    我们的结果表明,KLRC1敲除是提高NK细胞对HLA-E肿瘤的抗肿瘤活性的有效策略,可应用于NK细胞治疗实体瘤的开发。
    Natural Killer (NK) cells hold the potential to shift cell therapy from a complex autologous option to a universal off-the-shelf one. Although NK cells have demonstrated efficacy and safety in the treatment of leukemia, the limited efficacy of NK cell-based immunotherapies against solid tumors still represents a major hurdle. In the immunosuppressive tumor microenvironment (TME), inhibitory interactions between cancer and immune cells impair antitumoral immunity. KLRC1 gene encodes the NK cell inhibitory receptor NKG2A, which is a potent NK cell immune checkpoint. NKG2A specifically binds HLA-E, a non-classical HLA class I molecule frequently overexpressed in tumors, leading to the transmission of inhibitory signals that strongly impair NK cell function.
    To restore NK cell cytotoxicity against HLA-E+ tumors, we have targeted the NKG2A/HLA-E immune checkpoint by using a CRISPR-mediated KLRC1 gene editing.
    KLRC1 knockout resulted in a reduction of 81% of NKG2A+ cell frequency in ex vivo expanded human NK cells post-cell sorting. In vitro, the overexpression of HLA-E by tumor cells significantly inhibited wild-type (WT) NK cell cytotoxicity with p-values ranging from 0.0071 to 0.0473 depending on tumor cell lines. In contrast, KLRC1 KO NK cells exhibited significantly higher cytotoxicity when compared to WT NK cells against four different HLA-E+ solid tumor cell lines, with p-values ranging from<0.0001 to 0.0154. Interestingly, a proportion of 43.5% to 60.2% of NKG2A- NK cells within the edited NK cell population was sufficient to reverse at its maximum the HLA-E-mediated inhibition of NK cell cytotoxicity. The expression of the activating receptor NKG2C was increased in KLRC1 KO NK cells and contributed to the improved NK cell cytotoxicity against HLA-E+ tumors. In vivo, the adoptive transfer of human KLRC1 KO NK cells significantly delayed tumor progression and increased survival in a xenogeneic mouse model of HLA-E+ metastatic breast cancer, as compared to WT NK cells (p = 0.0015).
    Our results demonstrate that KLRC1 knockout is an effective strategy to improve NK cell antitumor activity against HLA-E+ tumors and could be applied in the development of NK cell therapy for solid tumors.
    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
    淋巴细胞活化基因3蛋白(LAG3)是一种抑制性受体,在肿瘤中耗尽的T细胞上上调。LAG3是临床试验中使用许多抗LAG3抗体进行癌症免疫疗法的主要靶标。然而,这些抗体识别的表位没有结构信息。我们确定了与LAG3结合的治疗性抗体(favezelimab)的单颗粒冷冻EM结构,分辨率为3.5。揭示favezelimab靶向II类MHC的LAG3结合位点,它的典型配体。fusezelimab的常规(单价)Fab和LAG3(〜100kDa)之间的复合物的小尺寸对冷冻EM提出了挑战。因此,我们设计了一种二价版本的Fabfavezelimab,它使Fab-LAG3复合体的大小增加了一倍,并赋予复合体高度可识别的形状,从而促进了粒子选择和定位,用于图像处理.这项研究建立了二价Fab作为小蛋白冷冻EM分析的新基准标记。
    Lymphocyte activation gene 3 protein (LAG3) is an inhibitory receptor that is upregulated on exhausted T cells in tumors. LAG3 is a major target for cancer immunotherapy with many anti-LAG3 antibodies in clinical trials. However, there is no structural information on the epitopes recognized by these antibodies. We determined the single-particle cryoEM structure of a therapeutic antibody (favezelimab) bound to LAG3 to 3.5 Å resolution, revealing that favezelimab targets the LAG3-binding site for MHC class II, its canonical ligand. The small size of the complex between the conventional (monovalent) Fab of favezelimab and LAG3 (∼100 kDa) presented a challenge for cryoEM. Accordingly, we engineered a bivalent version of Fab favezelimab that doubled the size of the Fab-LAG3 complex and conferred a highly identifiable shape to the complex that facilitated particle selection and orientation for image processing. This study establishes bivalent Fabs as new fiducial markers for cryoEM analysis of small proteins.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    T细胞可表达多种抑制性受体。在诱导T细胞耗竭以响应持久性抗原时,在抗肿瘤免疫反应中,许多是同时表达的。关键的抑制性受体是CTLA-4,PD-1,LAG3,TIM3和TIGIT,在这里调查。这些受体作为癌症免疫疗法中的中心治疗靶标是重要的。抑制性受体不在细胞表面组成型表达,但是大量的部分存在于细胞内囊泡结构中。仍未解决不同抑制性受体的亚细胞定位在多大程度上是不同的。使用亚细胞分布和质膜插入的定量成像,并辅以邻近蛋白质组学和抑制性受体与运输衔接子的关联的生化分析,五种抑制性受体的亚细胞分布是离散的。CTLA-4的分布最明显,与溶酶体衍生的囊泡和分选nexin1/2/5/6转运机制优先相关。由于缺乏证据表明存在特定的囊泡亚型来解释不同的抑制性受体分布,我们建议,这种分布是由通过重叠的囊泡结构关节组的不同运输驱动的。五种抑制性受体的亚细胞定位的这种广泛表征为它们的贩运及其治疗开发的分子研究奠定了基础。
    T cells can express multiple inhibitory receptors. Upon induction of T cell exhaustion in response to persistent antigen, prominently in the anti-tumor immune response, many are expressed simultaneously. Key inhibitory receptors are CTLA-4, PD-1, LAG3, TIM3 and TIGIT, as investigated here. These receptors are important as central therapeutic targets in cancer immunotherapy. Inhibitory receptors are not constitutively expressed on the cell surface, but substantial fractions reside in intracellular vesicular structures. It remains unresolved to which extent the subcellular localization of different inhibitory receptors is distinct. Using quantitative imaging of subcellular distributions and plasma membrane insertion as complemented by proximity proteomics and a biochemical analysis of the association of the inhibitory receptors with trafficking adaptors, the subcellular distributions of the five inhibitory receptors were discrete. The distribution of CTLA-4 was most distinct with preferential association with lysosomal-derived vesicles and the sorting nexin 1/2/5/6 transport machinery. With a lack of evidence for the existence of specific vesicle subtypes to explain divergent inhibitory receptor distributions, we suggest that such distributions are driven by divergent trafficking through an overlapping joint set of vesicular structures. This extensive characterization of the subcellular localization of five inhibitory receptors in relation to each other lays the foundation for the molecular investigation of their trafficking and its therapeutic exploitation.
    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
    白细胞介素-1受体家族成员(ILR)和Toll样受体(TLR)在免疫和炎症中起关键作用,并由大多数细胞类型表达,包括先天和适应性免疫系统的细胞。在这种情况下,IL-1超家族成员在调节适应性和先天淋巴细胞的功能和分化中也是重要的参与者。这个系统是严格的调节,以避免不受控制的激活,这可能导致导致自身免疫或过敏反应的有害炎症。IL-1R8(也称为TIR8或SIGIRR)是IL-1R家族的成员,其充当抑制ILR和TLR信号传导的负调节因子并且充当人IL-37的共受体。人类和小鼠NK细胞,是肿瘤和感染免疫监视的关键角色,表达高水平的IL-1R8。在这次审查中,我们将总结我们目前对结构的理解,IL-1R8的表达和功能,我们还将讨论IL-1R8作为重要的检查点在包括癌症和病毒感染在内的病理条件下调节NK细胞功能的新作用。
    Interleukin-1 receptor family members (ILRs) and Toll-Like Receptors (TLRs) play pivotal role in immunity and inflammation and are expressed by most cell types including cells of both the innate and adaptive immune system. In this context, IL-1 superfamily members are also important players in regulating function and differentiation of adaptive and innate lymphoid cells. This system is tightly regulated in order to avoid uncontrolled activation, which may lead to detrimental inflammation contributing to autoimmune or allergic responses. IL-1R8 (also known as TIR8 or SIGIRR) is a member of the IL-1R family that acts as a negative regulator dampening ILR and TLR signaling and as a co-receptor for human IL-37. Human and mouse NK cells, that are key players in immune surveillance of tumors and infections, express high level of IL-1R8. In this review, we will summarize our current understanding on the structure, expression and function of IL-1R8 and we will also discuss the emerging role of IL-1R8 as an important checkpoint regulating NK cells function in pathological conditions including cancer and viral infections.
    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