MHC

MHC
  • 文章类型: Journal Article
    单核细胞增生李斯特菌是食源性细胞内细菌模型病原体。针对李斯特菌的保护性免疫取决于有效的CD8+T细胞反应,但是在小鼠中很少有T细胞表位被认为是李斯特菌病的常见动物感染模型。为了鉴定表位,我们通过基于质谱的免疫肽筛选了感染小鼠脾脏中存在的李斯特菌免疫肽。我们绘制了在MHCI类分子上呈递的6000多个小鼠自身肽,包括来自12种不同细菌蛋白的12种高度自信的李斯特菌肽。进一步测试了具有确认片段谱的细菌免疫肽激活CD8+T细胞的潜力。从推定的细胞壁表面锚家族蛋白LMON_0576中揭示VTYNYINI作为一种新型的真正的肽表位。该表位在初免加强模型中显示出高生物学效力,并且可以用作研究工具以探测李斯特菌感染的小鼠模型中的CD8+T细胞应答。一起,我们的结果证明了免疫肽用于细菌抗原鉴定的能力。
    Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6,000 mouse self-peptides presented on MHC Class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification.
    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
    遗传多样性是反映物种自然选择和环境适应的重要指标。孤立的小种群容易受到遗传漂移的影响,近亲繁殖,和有限的基因流动;因此,评估它们的遗传多样性对保护至关重要。在这项研究中,我们使用中性微卫星和五个适应性主要组织相容性复合体(MHC)基因研究了黑白金丝猴(Rhinopithecusbieti)的遗传多样性。两个DQA1等位基因,两个DQB1等位基因,两个DRB1等位基因,两个DRB5等位基因,从一个种群中分离出三个DPB1等位基因。结果表明,中性微卫星表现出高度的杂合性和多态性,而适应性MHC基因表现出高度杂合性和中度多态性。结果还表明,平衡选择在进化过程中显著影响了物种的MHC多样性:(1)在DRB1,DRB5和DQB1基因的几个氨基酸位点(主要在抗原结合位点及其附近)发现了显着的正选择;(2)系统发育分析显示了所有MHC基因座的跨物种进化模式。这项研究为黑白金丝猴提供了有价值的遗传多样性见解,自更新世以来,它们居住在最高海拔,经历了全球所有灵长类动物中最恶劣的环境选择。这些结果为制定或修改这种濒危灵长类动物的保护策略提供了宝贵的科学证据和参考。
    Genetic diversity is an essential indicator that echoes the natural selection and environmental adaptation of a species. Isolated small populations are vulnerable to genetic drift, inbreeding, and limited gene flow; thus, assessing their genetic diversity is critical in conservation. In this study, we studied the genetic diversity of black-and-white snub-nosed monkeys (Rhinopithecus bieti) using neutral microsatellites and five adaptive major histocompatibility complex (MHC) genes. Two DQA1 alleles, two DQB1 alleles, two DRB1 alleles, two DRB5 alleles, and three DPB1 alleles were isolated from a population. The results indicate that neutral microsatellites demonstrate a high degree of heterozygosity and polymorphism, while adaptive MHC genes display a high degree of heterozygosity and moderate polymorphism. The results also show that balancing selection has prominently influenced the MHC diversity of the species during evolution: (1) significant positive selection is identified at several amino acid sites (primarily at and near antigen-binding sites) of the DRB1, DRB5, and DQB1 genes; (2) phylogenetic analyses display the patterns of trans-species evolution for all MHC loci. This study provides valuable genetic diversity insights into black-and-white snub-nosed monkeys, which dwell at the highest altitude and have experienced the harshest environmental selection of all primates globally since the Pleistocene. Such results provide valuable scientific evidence and a reference for making or amending conservation strategies for this endangered primate species.
    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
    细胞外囊泡(EV)是由体内几乎所有细胞释放的纳米级热稳定囊泡,包括肿瘤细胞和肿瘤浸润性树突状细胞(DC)。通过携带来自原始细胞的分子,EV在稳态和癌症中作为细胞对细胞通信者工作,但也可能代表有价值的治疗和诊断工具。这篇综述的重点是肿瘤衍生的EV(TEV)在DC功能的调节中的作用,以及肿瘤和DC衍生的EV在免疫治疗和基于DC的疫苗设计中的治疗潜力。TEV最初的特征在于它们能够将肿瘤抗原转移到DC,但目前被认为主要是免疫抑制性的,因为DC抑制分子如PD-L1,HLA-G,PGE2等。然而,TEV仍然可以代表特权系统,以在适当的工程化后将抗原材料递送至DC以减少其免疫抑制货物或增加免疫原性。DC衍生的EV比肿瘤衍生的EV更有前途,因为它们暴露了抗原负载的MHC,在没有免疫抑制货物的情况下,共刺激分子和NK细胞激活配体。此外,与基于细胞的药物相比,DC衍生的EV具有几个优点,例如更高的抗原/MHC浓度和易于操作以及对免疫抑制微环境的更低敏感性。临床前模型表明,DC衍生的EV通过将抗原转移到肿瘤浸润的DC,直接或间接地有效激活肿瘤特异性NK和T细胞反应。相比之下,然而,I期和II期试验显示,基于EV的抗癌疫苗的临床疗效有限.我们讨论了基于EV的治疗的未来取决于我们克服重大挑战的能力,例如对其生物学和药代动力学的理解仍然不完整,以及缺乏高通量分离和纯化的标准化方法。尽管如此,作为癌症免疫疗法的候选者,电动汽车仍然备受关注,这可能会超过基于细胞的策略。
    Extracellular vesicles (EVs) are nanosized heat-stable vesicles released by virtually all cells in the body, including tumor cells and tumor-infiltrating dendritic cells (DCs). By carrying molecules from originating cells, EVs work as cell-to-cell communicators in both homeostasis and cancer but may also represent valuable therapeutic and diagnostic tools. This review focuses on the role of tumor-derived EVs (TEVs) in the modulation of DC functions and on the therapeutic potential of both tumor- and DC-derived EVs in the context of immunotherapy and DC-based vaccine design. TEVs were originally characterized for their capability to transfer tumor antigens to DCs but are currently regarded as mainly immunosuppressive because of the expression of DC-inhibiting molecules such as PD-L1, HLA-G, PGE2 and others. However, TEVs may still represent a privileged system to deliver antigenic material to DCs upon appropriate engineering to reduce their immunosuppressive cargo or increase immunogenicity. DC-derived EVs are more promising than tumor-derived EVs since they expose antigen-loaded MHC, costimulatory molecules and NK cell-activating ligands in the absence of an immunosuppressive cargo. Moreover, DC-derived EVs possess several advantages as compared to cell-based drugs such as a higher antigen/MHC concentration and ease of manipulation and a lower sensitivity to immunosuppressive microenvironments. Preclinical models showed that DC-derived EVs efficiently activate tumor-specific NK and T cell responses either directly or indirectly by transferring antigens to tumor-infiltrating DCs. By contrast, however, phase I and II trials showed a limited clinical efficacy of EV-based anticancer vaccines. We discuss that the future of EV-based therapy depends on our capability to overcome major challenges such as a still incomplete understanding of their biology and pharmacokinetic and the lack of standardized methods for high-throughput isolation and purification. Despite this, EVs remain in the limelight as candidates for cancer immunotherapy which may outmatch cell-based strategies in the fullness of their time.
    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
    个性化癌症疫苗已成为癌症治疗或预防策略的有希望的途径。这种方法针对个体患者肿瘤中的特定遗传改变,提供更个性化和有效的治疗选择。以前的研究表明,针对有限范围的基因突变的广义肽疫苗是无效的,强调需要个性化的方法。虽然研究已经探索了个性化的mRNA疫苗,在这种情况下,尚未研究个性化的肽疫苗。胰腺导管腺癌(PDAC)在肿瘤学中仍然具有挑战性,需要创新的治疗策略。在这项研究中,我们开发了一种个性化的肽疫苗设计方法,采用RNA测序(RNAseq)鉴定患者实体瘤组织中PDAC发展的流行基因突变。我们对修剪适配器进行了RNAseq分析,读取对齐,和体细胞变异呼叫。我们还开发了一个名为SCGeneID的Python程序,这验证了RNAseq分析的比对。Python程序可以免费下载。利用染色体数和基因座数据,SCGeneID沿着UCSChg38参考集鉴定靶基因。根据基因突变数据,我们开发了一种个性化的PDAC癌症疫苗,该疫苗针对两名患者的100个高度流行的基因突变.我们预测肽-MHC结合亲和力,免疫原性,抗原性,变应原性,和每个表位的毒性。然后,我们根据之前发表的疫苗设计方法选择了前50和100个表位.最后,我们产生了pMHC-TCR三维分子模型复杂结构,可免费下载。设计的个性化癌症疫苗含有PDAC实体瘤组织中常见的表位。我们的个性化疫苗由新抗原组成,允许针对癌细胞的更精确和有针对性的免疫反应。此外,我们发现了突变基因,在参考研究中也发现了这一点,我们获得了测序数据,从而验证我们的疫苗设计方法。这是第一个使用人类患者数据设计针对新抗原的个性化肽癌症疫苗的研究,以鉴定与感兴趣的特定肿瘤相关的基因突变。
    Personalized cancer vaccines have emerged as a promising avenue for cancer treatment or prevention strategies. This approach targets the specific genetic alterations in individual patient\'s tumors, offering a more personalized and effective treatment option. Previous studies have shown that generalized peptide vaccines targeting a limited scope of gene mutations were ineffective, emphasizing the need for personalized approaches. While studies have explored personalized mRNA vaccines, personalized peptide vaccines have not yet been studied in this context. Pancreatic ductal adenocarcinoma (PDAC) remains challenging in oncology, necessitating innovative therapeutic strategies. In this study, we developed a personalized peptide vaccine design methodology, employing RNA sequencing (RNAseq) to identify prevalent gene mutations underlying PDAC development in a patient solid tumor tissue. We performed RNAseq analysis for trimming adapters, read alignment, and somatic variant calling. We also developed a Python program called SCGeneID, which validates the alignment of the RNAseq analysis. The Python program is freely available to download. Using chromosome number and locus data, SCGeneID identifies the target gene along the UCSC hg38 reference set. Based on the gene mutation data, we developed a personalized PDAC cancer vaccine that targeted 100 highly prevalent gene mutations in two patients. We predicted peptide-MHC binding affinity, immunogenicity, antigenicity, allergenicity, and toxicity for each epitope. Then, we selected the top 50 and 100 epitopes based on our previously published vaccine design methodology. Finally, we generated pMHC-TCR 3D molecular model complex structures, which are freely available to download. The designed personalized cancer vaccine contains epitopes commonly found in PDAC solid tumor tissue. Our personalized vaccine was composed of neoantigens, allowing for a more precise and targeted immune response against cancer cells. Additionally, we identified mutated genes, which were also found in the reference study, where we obtained the sequencing data, thus validating our vaccine design methodology. This is the first study designing a personalized peptide cancer vaccine targeting neoantigens using human patient data to identify gene mutations associated with the specific tumor of interest.
    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
    核苷酸结合和寡聚化结构域样受体(NLR)NLR家族含有CARD结构域的蛋白5(NLRC5)和II类主要组织相容性复合物反式激活因子(CIITA)是主要组织相容性复合物(MHC)I类和II类基因的转录调节因子,分别。MHC分子是我们免疫系统的核心角色,允许检测危险的“非自身”抗原,因此,从生物体中识别和消除感染或转化的细胞。最近,CIITA和NLRC5已成为感兴趣地位于延伸的MHC基因座中的嗜丁基酶(BTN)家族的选定基因的调节因子。BTN是与B7家族共调节分子表现出结构相似性的跨膜蛋白。家族成员BTN2A2确实有助于控制T细胞活化,被发现受CIITA转录调控。相反,NLRC5作为BTN3A1、BTN3A2和BTN3A3基因的重要调节因子出现。与BTN2A1一起,BTN3As调节由微生物来源或在血液癌细胞中积累的选定代谢物触发的非常规Vγ9Vδ2T细胞反应。即使内源性代谢物符合“自我”的规范定义,代谢异常的细胞可能对生物体构成危险,应该由免疫系统细胞识别和控制。总的来说,关于NLRC5在BTN3As表达中的作用的新数据将调节典型的“非自身”呈现的机制与那些标记具有异常代谢构型的细胞进行免疫识别的机制联系起来,我们在这篇透视综述中讨论的进化平行。
    The nucleotide-binding and oligomerization domain-like receptors (NLRs) NLR family CARD domain-containing protein 5 (NLRC5) and Class II Major Histocompatibility Complex Transactivator (CIITA) are transcriptional regulators of major histocompatibility complex (MHC) class I and class II genes, respectively. MHC molecules are central players in our immune system, allowing the detection of hazardous \'non-self\' antigens and, thus, the recognition and elimination of infected or transformed cells from the organism. Recently, CIITA and NLRC5 have emerged as regulators of selected genes of the butyrophilin (BTN) family that interestingly are located in the extended MHC locus. BTNs are transmembrane proteins exhibiting structural similarities to B7 family co-modulatory molecules. The family member BTN2A2, which indeed contributes to the control of T cell activation, was found to be transcriptionally regulated by CIITA. NLRC5 emerged instead as an important regulator of the BTN3A1, BTN3A2, and BTN3A3 genes. Together with BTN2A1, BTN3As regulate non-conventional Vγ9Vδ2 T cell responses triggered by selected metabolites of microbial origin or accumulating in hematologic cancer cells. Even if endogenous metabolites conform to the canonical definition of \'self\', metabolically abnormal cells can represent a danger for the organism and should be recognized and controlled by immune system cells. Collectively, new data on the role of NLRC5 in the expression of BTN3As link the mechanisms regulating canonical \'non-self\' presentation and those marking cells with abnormal metabolic configurations for immune recognition, an evolutionary parallel that we discuss in this perspective review.
    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
    牛主要组织相容性复合物(MHC),也被称为牛白细胞抗原(BoLA)复合物,是编码用于抗原呈递以启动免疫应答的最重要分子的基因组区域。牛中MHC的第一个证据指出了一个含有2种抗原的基因座,一种通过细胞毒性抗血清(MHCI类)检测到,另一种通过混合淋巴细胞培养试验(MHCII类)研究。BoLA区域中研究最多的基因是高度多态的BoLA-DRB3,其编码具有参与T细胞的抗原呈递的肽沟结构域的β链,所述T细胞将产生并共刺激细胞和体液效应物应答。BoLA-DRB3等位基因与感染性疾病如乳腺炎的结果有关。锥虫病,和tick负载,并具有生产特征。为了对这些等位基因进行分类,提出了2种命名方法,目前这两种系统的使用使得很难列出,有效地理解和应用这些数据。在这篇综述中,我们组织了所有关于BoLA-DRB3等位基因频率的报告中可用的知识。它涵盖了至少26个国家对30多个品种进行的研究的信息;在重要的牲畜生产者国家缺乏研究。我们强调BoLA研究在鉴定与传染性和寄生虫病抗性相关的标记物中的实际应用。增加的生产性状和T细胞表位作图,除了商业和克里奥尔和当地适应品种的遗传多样性和保护研究。最后,我们为发现新的BoLA等位基因并揭示该基因座在生产性状中的未知作用的研究需求提供了支持。
    The bovine Major Histocompatibility Complex (MHC), also known as the Bovine Leucocyte Antigen (BoLA) complex, is the genomic region that encodes the most important molecules for antigen presentation to initiate immune responses. The first evidence of MHC in bovines pointed to a locus containing 2 antigens, one detected by cytotoxic antiserum (MHC class I) and another studied by mixed lymphocyte culture tests (MHC class II). The most studied gene in the BoLA region is the highly polymorphic BoLA-DRB3, which encodes a β chain with a peptide groove domain involved in antigen presentation for T cells that will develop and co-stimulate cellular and humoral effector responses. BoLA-DRB3 alleles have been associated with outcomes in infectious diseases such as mastitis, trypanosomiasis, and tick loads, and with production traits. To catalog these alleles, 2 nomenclature methods were proposed, and the current use of both systems makes it difficult to list, comprehend and apply these data effectively. In this review we have organized the knowledge available in all of the reports on the frequencies of BoLA-DRB3 alleles. It covers information from studies made in at least 26 countries on more than 30 breeds; studies are lacking in countries that are important producers of cattle livestock. We highlight practical applications of BoLA studies for identification of markers associated with resistance to infectious and parasitic diseases, increased production traits and T cell epitope mapping, in addition to genetic diversity and conservation studies of commercial and creole and locally adapted breeds. Finally, we provide support for the need of studies to discover new BoLA alleles and uncover unknown roles of this locus in production traits.
    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
    众所周知,预先形成的抗猪抗体对供体组织的攻击会导致异种移植中的移植物失败。对猪供体进行基因工程以消除这些预先形成的抗体的靶标,再加上免疫抑制药物的进步,现在可以在临床前猪到非人灵长类动物模型中实现延长的存活。尽管有这些改进,抗体在移植的一生中仍然存在风险,和许多患者继续预先形成的供体特异性抗体,甚至高度工程化的猪。虽然存在可以帮助减轻抗体有害影响的疗法,他们的行为广泛可能削弱有益的免疫力。识别额外的异种抗原可以实现更有针对性的方法,比如基因编辑,通过进一步消除供体组织上的抗体靶标来克服这些挑战。因为我们发现经典的I类猪白细胞抗原是人类抗体的靶标,我们现在研究相关的猪蛋白是否也可能被人类抗体靶向。我们在这里表明,非经典I类猪白细胞蛋白(SLA-6,-7,-8)可以在哺乳动物细胞表面表达并充当抗体靶标。
    Attack of donor tissues by pre-formed anti-pig antibodies is well known to cause graft failure in xenotransplantation. Genetic engineering of porcine donors to eliminate targets of these pre-formed antibodies coupled with advances in immunosuppressive medicines have now made it possible to achieve extended survival in the pre-clinical pig-to-non-human primate model. Despite these improvements, antibodies remain a risk over the lifetime of the transplant, and many patients continue to have pre-formed donor-specific antibodies even to highly engineered pigs. While therapeutics exist that can help mitigate the detrimental effects of antibodies, they act broadly potentially dampening beneficial immunity. Identifying additional xenoantigens may enable more targeted approaches, such as gene editing, to overcome these challenges by further eliminating antibody targets on donor tissue. Because we have found that classical class I swine leukocyte antigens are targets of human antibodies, we now examine whether related pig proteins may also be targeted by human antibodies. We show here that non-classical class I swine leukocyte proteins (SLA-6, -7, -8) can be expressed at the surface of mammalian cells and act as antibody 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: Congress
    暂无摘要。
    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:胃肠道恶性肿瘤包括对全球健康构成重大挑战的多种癌症。主要组织相容性复合体(MHC)在免疫监视中起着关键作用,协调免疫系统对肿瘤细胞的识别和消除。然而,MHC基因表达的复杂调控易受动态表观遗传修饰,会影响功能和病理结果。
    方法:通过了解驱动MHC下调的表观遗传学改变,深入了解免疫逃逸的分子机制,肿瘤进展,和免疫疗法抗性。本系统综述审查了当前有关食管MHC失调的表观遗传机制的文献。胃,胰腺,肝脏和结肠直肠恶性肿瘤。讨论了靶向异常表观遗传修饰以恢复MHC表达和免疫治疗干预的有效性的潜在临床意义。
    结论:表观遗传靶向疗法与免疫疗法的整合对于改善胃肠道恶性肿瘤患者的临床结果具有巨大的潜力,并且是未来研究和治疗发展的令人信服的途径。
    BACKGROUND: Gastrointestinal malignancies encompass a diverse group of cancers that pose significant challenges to global health. The major histocompatibility complex (MHC) plays a pivotal role in immune surveillance, orchestrating the recognition and elimination of tumor cells by the immune system. However, the intricate regulation of MHC gene expression is susceptible to dynamic epigenetic modification, which can influence functionality and pathological outcomes.
    METHODS: By understanding the epigenetic alterations that drive MHC downregulation, insights are gained into the molecular mechanisms underlying immune escape, tumor progression, and immunotherapy resistance. This systematic review examines the current literature on epigenetic mechanisms that contribute to MHC deregulation in esophageal, gastric, pancreatic, hepatic and colorectal malignancies. Potential clinical implications are discussed of targeting aberrant epigenetic modifications to restore MHC expression and 0 the effectiveness of immunotherapeutic interventions.
    CONCLUSIONS: The integration of epigenetic-targeted therapies with immunotherapies holds great potential for improving clinical outcomes in patients with gastrointestinal malignancies and represents a compelling avenue for future research and therapeutic development.
    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
    用于人类白细胞抗原(HLA)分型的基于SNP的插补方法利用了主要组织相容性复合物(MHC)区域内的单倍型结构。这些方法使用密集的SNP基因型预测HLA经典等位基因,通常在全基因组关联研究(GWAS)中使用的基于阵列的平台上发现。HLA经典等位基因的分析可以在没有额外成本的情况下在当前SNP数据集上进行。这里,我们描述了HIBAG的工作流程,一种带有属性装袋的插补方法,使用SNP数据推断样本的HLA经典等位基因。提供了两个示例来演示使用1000Genomes项目最新版本的公共HLA和SNP数据的功能:使用GWAS中预先构建的分类器进行基因型填补,和模型训练,以创建新的预测模型。GPU实现有助于模型构建,使它比单线程实现快数百倍。
    SNP-based imputation approaches for human leukocyte antigen (HLA) typing take advantage of the haplotype structure within the major histocompatibility complex (MHC) region. These methods predict HLA classical alleles using dense SNP genotypes, commonly found on array-based platforms used in genome-wide association studies (GWAS). The analysis of HLA classical alleles can be conducted on current SNP datasets at no additional cost. Here, we describe the workflow of HIBAG, an imputation method with attribute bagging, to infer a sample\'s HLA classical alleles using SNP data. Two examples are offered to demonstrate the functionality using public HLA and SNP data from the latest release of the 1000 Genomes project: genotype imputation using pre-built classifiers in a GWAS, and model training to create a new prediction model. The GPU implementation facilitates model building, making it hundreds of times faster compared to the single-threaded implementation.
    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