T cell recruitment

T 细胞募集
  • 文章类型: Journal Article
    慢性阻塞性肺疾病(COPD)是一种呼吸道炎症性疾病。补骨脂素(PSO)是传统上用于治疗COPD的补肾方川配方中确定的主要药理成分,但其在COPD炎症中的疗效未见报道.在这项研究中,我们旨在阐明PSO在COPD中的抗炎潜力,并阐明其潜在机制,关注T淋巴细胞募集和趋化因子的调节,即干扰素-γ(CXCL9)诱导的单核因子,干扰素诱导蛋白10(CXCL10),和干扰素诱导型T细胞α化学引诱物(CXCL11)。体外,RAW264.7由干扰素(IFN)-γ+香烟烟雾提取物(CSE)刺激,并用PSO(2.5,5,10μM)处理,然后通过实时PCR和Westernblot分析趋化因子的水平和Janus激酶(JAK)/信号转导和转录激活因子1(STAT1)通路的激活。在体内,在第1、8、15和22天通过腹膜内注射CSE建立小鼠模型,然后用PSO(10mg/kg)治疗。我们的体外实验表明,PSO降低了CXCL9,CXCL10和CXCL11的水平,并降低了JAK2和STAT1的蛋白质磷酸化水平。此外,PSO有效改善CSE暴露小鼠的炎性浸润并降低CD8+T细胞的比例。此外,PSO降低支气管肺泡灌洗液(BALF)和肺组织中CXCL9、CXCL10和CXCL11的水平,并降低JAK2和STAT1的蛋白磷酸化水平。总之,我们的结果揭示了PSO对COPD炎症的治疗潜力,可能通过JAK2/STAT1信号通路调节CD8+T细胞募集和趋化因子。
    Chronic obstructive pulmonary disease (COPD) is a respiratory inflammatory disease. Psoralen (PSO) is the main pharmacological component identified from Bu-Shen-Fang-Chuan formula which has been traditionally used in treatment of COPD, yet its efficacy in COPD inflammation were unreported. In this study, we aimed to elucidate the anti-inflammatory potential of PSO in COPD and unravel the underlying mechanisms, focusing on T lymphocyte recruitment and the modulation of chemokines, namely monokine induced by interferon-gamma (CXCL9), interferon inducible protein 10 (CXCL10), and interferon inducible T-Cell alpha chemoattractant (CXCL11). In vitro, RAW264.7 was stimulated by interferon (IFN)-γ + cigarette smoke extract (CSE) and were treated with PSO (2.5, 5, 10 μM), then the levels of chemokines and the activation of Janus kinase (JAK)/Signal transducer and activator of transcription 1 (STAT1) pathway were analyzed by real time PCR and western blot. In vivo, a murine model was established by intraperitoneal injection of CSE on day 1, 8, 15, and 22, then treated with PSO (10 mg/kg). Our experiments in vitro illustrated that PSO reduced the levels of CXCL9, CXCL10, and CXCL11, and decreased the protein phosphorylation levels of JAK2 and STAT1. Additionally, PSO effectively improved inflammatory infiltration and decreased the proportion of CD8+ T cells in CSE-exposed mice. Furthermore, PSO reduced the levels of CXCL9, CXCL10, and CXCL11 in bronchoalveolar lavage fluid (BALF) and lung tissue, and decreased the protein phosphorylation levels of JAK2 and STAT1. In conclusion, our results revealed the therapeutic potential of PSO for COPD inflammation, possibly mediated through the regulation of CD8+ T cell recruitment and chemokines via the JAK2/STAT1 signaling pathway.
    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
    尽管免疫检查点封锁(ICB)具有开创性的影响,非小细胞肺癌的反应率仍然适中,特别是在免疫排斥或免疫沙漠微环境中。Toll样受体7(TLR7)作为一个潜在的靶点桥接先天和适应性免疫,为联合治疗增加ICB疗效提供了一个有希望的途径。这里,我们探索了新型口服TLR7激动剂TQ-A3334的抗肿瘤活性及其在同基因鼠肺癌模型中通过组合策略增强抗程序性死亡配体1(PD-L1)治疗的潜力.口服TQ-A3334可显着减轻C57BL/6J小鼠的肿瘤负担,由I型干扰素(IFN)调节,并表现出低毒性。这种疗法在肿瘤组织中引起先天和适应性免疫细胞的激活。特别是通过I型IFN途径和随后的CXCL10表达增加CD8TIL的丰度。体外检查证实,IFN-α刺激的肿瘤细胞显示CXCL10分泌增加,有利于促进CD8T细胞的运输。此外,TQ-A3334与抗PD-L1联合治疗超过了肿瘤控制,与单一疗法相比,CD8+TIL频率进一步增加。这些结果表明,TQ-A3334可以动员先天免疫并促进T细胞募集到肿瘤微环境;TQ-A3334和抗PD-L1抗体的组合可以增强肿瘤对抗PD-L1治疗的敏感性。这证明了治疗免疫能力差的浸润性肺癌的巨大潜力。
    Despite the groundbreaking impact of immune checkpoint blockade (ICB), response rates in non-small cell lung cancer remain modest, particularly in immune-excluded or immune-desert microenvironments. Toll-like receptor 7 (TLR7) emerges as a latent target bridging innate and adaptive immunity, offering a promising avenue for combination therapies to augment ICB efficacy. Here, we explored the anti-tumor activity of the novel oral TLR7 agonist TQ-A3334 and its potential to enhance anti-programmed death ligand 1 (PD-L1) therapy through a combination strategy in a syngeneic murine lung cancer model. Oral administration of TQ-A3334 significantly alleviated tumor burden in C57BL/6J mice, modulated by type I interferon (IFN), and exhibited low toxicity. This therapy elicited activation of both innate and adaptive immune cells in tumor tissue, particularly increasing the abundance of CD8+ TILs through type I IFN pathway and subsequent CXCL10 expression. In vitro examinations validated that IFN-α-stimulated tumor cells exhibited increased secretion of CXCL10, conducive to the promoted trafficking of CD8+ T cells. Furthermore, combining TQ-A3334 with anti-PD-L1 treatment exceeded tumor control, with a further increase in CD8+ TIL frequency compared to monotherapy. These findings suggest that TQ-A3334 can mobilize innate immunity and promote T cell recruitment into the tumor microenvironment; a combination of TQ-A3334 and anti-PD-L1 antibodies can intensify the sensitivity of tumors to anti-PD-L1 therapy, which demonstrates significant potential for treating poorly immune-infiltrated lung cancer.
    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
    结直肠癌是全球主要的健康负担,传统治疗方式在提高生存率方面的功效有限。然而,免疫治疗的最新进展改善了这种癌症患者的治疗效果.为了满足对改善治疗效果的持续需求,这项研究引入了一种新型的三特异性抗体,IMT030122,目标是EpCAM,4-1BB,和CD3。我们在体外和体内评估了IMT030122的药理功效和作用机制。在体外研究中,IMT030122表现出与抗原和表达EpCAM的细胞的差异结合,4-1BB,和CD3。此外,IMT030122依赖于EpCAM靶向的细胞内CD3和4-1BB信号的激活和介导的对HCT116结直肠癌细胞特异性的T细胞毒性。在体内,IMT030122显示出有效的抗肿瘤活性,显著抑制结肠癌HCT116和MC38-hEpCAM皮下移植瘤的生长。进一步的药理学分析显示,IMT030122从外周血中招募淋巴细胞进入结直肠癌组织并发挥持久的抗肿瘤活性,主要通过促进激活,扩散,和CD8T细胞的分化。值得注意的是,即使在结肠直肠癌组织中存在显著耗竭的淋巴细胞,IMT030122仍表现出抗肿瘤功效。IMT030122的有效药理活性和抗肿瘤作用表明,它可能会增强治疗功效,并在未来显着延长结直肠癌患者的生存期。
    Colorectal cancer is a major global health burden, with limited efficacy of traditional treatment modalities in improving survival rates. However, recently advances in immunotherapy has improved treatment outcomes for patients with this cancer. To address the continuing need for improved treatment efficacy, this study introduced a novel tri-specific antibody, IMT030122, that targets EpCAM, 4-1BB, and CD3. We evaluated the pharmacological efficacy and mechanism of action of IMT030122 in vitro and in vivo. In in vitro studies, IMT030122 exhibited differential binding to antigens and cells expressing EpCAM, 4-1BB, and CD3. Moreover, IMT030122 relied on EpCAM-targeted activation of intracellular CD3 and 4-1BB signaling and mediated T cell cytotoxicity specific to HCT116 colorectal cancer cells. In vivo, IMT030122 demonstrated potent anti-tumor activity, significantly inhibiting the growth of colon cancer HCT116 and MC38-hEpCAM subcutaneous grafts. Further pharmacological analysis revealed that IMT030122 recruited lymphocytes from peripheral blood into colorectal cancer tissue and exerted durable anti-tumor activity, predominantly by promoting the activation, proliferation, and differentiation of CD8T cells. Notably, IMT030122 still exhibited anti-tumor efficacy even in the presence of significantly depleted lymphocytes in colorectal cancer tissue. The potent pharmacological activity and anti-tumor effects of IMT030122 suggest it may enhance treatment efficacy and substantially extend the survival of patients with colorectal cancer in the future.
    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
    食管腺癌(OAC)中免疫抑制性肿瘤微环境的存在是反应不良的主要原因。需要新的治疗策略来补充目前的治疗方案并提高患者的生存率。这项研究检查了放射治疗和趋化因子受体拮抗作用对OAC中T细胞表型的免疫调节作用,主要目标是确定潜在的治疗靶标以与放射结合以改善抗肿瘤反应。与健康对照相比,OAC患者的抗肿瘤T细胞功能受损,由CD4+T辅助细胞产生较低的IFN-γ和较低的CD8+T细胞的细胞毒性潜力证明。在用临床相关剂量的辐射治疗后,这种减少的T细胞效应子功能得到增强。有趣的是,与健康对照相比,OAC患者血液中的CCR5+T细胞明显更丰富,临床相关剂量的照射进一步增强了T细胞的CCR5表面表达。此外,辐射增强了T细胞向OAC患者来源的肿瘤条件培养基(TCM)的迁移。用CCR5拮抗剂Maraviroc体外治疗可增强CD4T细胞产生的IFN-γ,并增加辐照的CD8T细胞向辐照的中药的迁移,表明其与辐射结合的协同治疗潜力。总的来说,这项研究强调了辐射在促进OAC中抗肿瘤T细胞反应和增加肿瘤中T细胞向趋化线索迁移方面的免疫刺激特性。重要的是,CCR5拮抗剂Maraviroc有望与放射疗法联合使用,以促进OAC中的抗肿瘤T细胞反应。
    The presence of an immunosuppressive tumour microenvironment in oesophageal adenocarcinoma (OAC) is a major contributor to poor responses. Novel treatment strategies are required to supplement current regimens and improve patient survival. This study examined the immunomodulatory effects that radiation therapy and chemokine receptor antagonism impose on T cell phenotypes in OAC with a primary goal of identifying potential therapeutic targets to combine with radiation to improve anti-tumour responses. Compared with healthy controls, anti-tumour T cell function was impaired in OAC patients, demonstrated by lower IFN-γ production by CD4+ T helper cells and lower CD8+ T cell cytotoxic potential. Such diminished T cell effector functions were enhanced following treatment with clinically relevant doses of irradiation. Interestingly, CCR5+ T cells were significantly more abundant in OAC patient blood compared with healthy controls, and CCR5 surface expression by T cells was further enhanced by clinically relevant doses of irradiation. Moreover, irradiation enhanced T cell migration towards OAC patient-derived tumour-conditioned media (TCM). In vitro treatment with the CCR5 antagonist Maraviroc enhanced IFN-γ production by CD4+ T cells and increased the migration of irradiated CD8+ T cells towards irradiated TCM, suggesting its synergistic therapeutic potential in combination with irradiation. Overall, this study highlights the immunostimulatory properties of radiation in promoting anti-tumour T cell responses in OAC and increasing T cell migration towards chemotactic cues in the tumour. Importantly, the CCR5 antagonist Maraviroc holds promise to be repurposed in combination with radiotherapy to promote anti-tumour T cell responses in OAC.
    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 cell ignorance is a specific form of immunological tolerance. It describes the maintenance of naivety in antigen-specific T cells in vivo despite the presence of their target antigen. It is thought to mainly play a role during the steady state, when self-antigens are presented in absence of costimulatory signals and at low density or to T cells of low affinity. In how far antigen-specific T cells can also remain clonally ignorant to foreign antigens, presented in the inflammatory context of systemic infection, remains unclear. Using single-cell in vivo fate mapping and high throughput flow cytometric enrichment, we find that high-affinity antigen-specific CD8+ T cells are efficiently recruited upon systemic infection. In contrast, most low-affinity antigen-specific T cells ignore the priming antigen and persist in the naïve state while remaining fully responsive to subsequent immunization with a high-affinity ligand. These data establish the widespread clonal ignorance of low-affinity T cells as a major factor shaping the composition of antigen-specific CD8+ T cell responses to systemic infection.
    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淋巴细胞(T细胞)对于肿瘤免疫治疗至关重要。然而,激活的T细胞数量不足极大地限制了肿瘤免疫治疗的疗效。在这里,我们提出了一种模拟溶瘤病毒的策略,以增强T细胞募集和活化,用于肿瘤治疗.我们构建了溶瘤病毒样纳米平台(PolyIC@ZIF-8),该平台在酸性肿瘤环境中降解以释放PolyIC和Zn2。释放的PolyIC表现出溶瘤病毒样功能,通过肿瘤抗原依赖性方式诱导肿瘤细胞凋亡并促进T细胞募集和活化。更重要的是,释放的Zn2+不仅通过诱导CXCL9/10/11表达来增强T细胞募集,而且通过不依赖肿瘤抗原的方式诱导ZAP-70磷酸化来促进T细胞活化以增加干扰素-γ(INF-γ)表达。这种Zn2+增强的溶瘤病毒模拟策略为肿瘤免疫治疗提供了新的方法。
    T lymphocytes (T cells) are essential for tumor immunotherapy. However, the insufficient number of activated T cells greatly limits the efficacy of tumor immunotherapy. Herein, we proposed an oncolytic virus-mimicking strategy to enhance T cell recruitment and activation for tumor treatment. We constructed an oncolytic virus-like nanoplatform (PolyIC@ZIF-8) that was degraded in the acidic tumor environment to release PolyIC and Zn2+ . The released PolyIC exhibited an oncolytic virus-like function that induced tumor cell apoptosis and promoted T cell recruitment and activation through a tumor antigen-dependent manner. More importantly, the released Zn2+ not only enhanced T cell recruitment by inducing CXCL9/10/11 expression but also promoted T cell activation to increase interferon-γ (INF-γ) expression by inducing the phosphorylation of ZAP-70 via a tumor antigen-independent manner. This Zn2+ -enhanced oncolytic virus-mimicking strategy provides a new approach for tumor immunotherapy.
    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
    克氏锥虫是恰加斯病的病因。在T细胞介导的急性期感染抑制后,这种细胞内真核病原体在极低水平的有限组织中长期存在。这种组织特异性慢性的原因尚不清楚。使用双生物发光荧光报告菌株和高灵敏度的组织成像,允许以单细胞分辨率监测实验性感染,我们对小鼠结肠中稀有寄生细胞的免疫微环境进行了系统分析,持久性的关键站点。我们证明,T细胞不完全募集到结肠感染灶的一部分,会导致细胞内寄生虫复制和分化为活动色素动物的重复循环,其频率足以使慢性感染永存。尽管存在,但寄生虫在该组织部位的终生持久性仍在继续,在系统层面,高效的T细胞反应。克服这种低水平的动态宿主-寄生虫平衡是疫苗开发的主要挑战。
    Trypanosoma cruzi is the etiological agent of Chagas disease. Following T cell-mediated suppression of acute-phase infection, this intracellular eukaryotic pathogen persists long-term in a limited subset of tissues at extremely low levels. The reasons for this tissue-specific chronicity are not understood. Using a dual bioluminescent-fluorescent reporter strain and highly sensitive tissue imaging that allows experimental infections to be monitored at single-cell resolution, we undertook a systematic analysis of the immunological microenvironments of rare parasitized cells in the mouse colon, a key site of persistence. We demonstrate that incomplete recruitment of T cells to a subset of colonic infection foci permits the occurrence of repeated cycles of intracellular parasite replication and differentiation to motile trypomastigotes at a frequency sufficient to perpetuate chronic infections. The lifelong persistence of parasites in this tissue site continues despite the presence, at a systemic level, of a highly effective T cell response. Overcoming this low-level dynamic host-parasite equilibrium represents a major challenge for vaccine 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
    The tumour microenvironment (TME) is the complex environment in which various non-cancerous stromal cell populations co-exist, co-evolve and interact with tumour cells, having a profound impact on the progression of solid tumours. The TME is comprised of various extracellular matrix (ECM) proteins in addition to a variety of immune and stromal cells. These include tumour-associated macrophages, regulatory T cells (Tregs), myeloid-derived suppressor cells, as well as endothelial cells, pericytes and cancer-associated fibroblasts (CAFs). CAFs are the most abundant stromal cell population in many tumours and support cancer progression, metastasis and resistance to therapies through bidirectional signalling with both tumour cells and other cells within the TME. More recently, CAFs have been shown to also affect the anti-tumour immune response through direct and indirect interactions with immune cells. In this review, we specifically focus on the interactions between CAFs and cytotoxic CD8+ T cells, and on how these interactions affect T cell recruitment, infiltration and function in the tumour. We additionally provide insight into the therapeutic implications of targeting these interactions, particularly in the context of cancer immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    Recently, the combination of platinum chemotherapy with PD-1/PD-L1 pathway blockades has shown synergistic efficacy in a few clinical trials. However, the exact mechanisms and the optimized sequence of such combinations are not fully clear. In this study, we combined different doses of platinum agents (cisplatin or oxaliplatin) with sequential therapy of PD-1 blockade therapy (anti-PD-1 antibody or anti-PD-L1 antibody) to treat established MC38 murine colon tumors. Although 10 mg/kg platinum (cisplatin or oxaliplatin) showed no significant effect on tumor growth, its combination with sequential anti-PD-1 antibody administration caused complete tumor remission in 80-100% mice. The synergic therapeutic efficacy was found to be associated with more effector and less exhausted CD8 T cell infiltration in the tumor sites. Platinum chemotherapy is generally considered immunosuppressive, with lymphopenia and neutropenia being common side effects. However, our data showed that high-dose (20 mg/kg) platinum treatment induced lymphopenia in MC38 tumor-bearing mice, and low-dose (10 mg/kg) treatment augmented the T cell response with an increased number of peripheral T cells. Notably, increased numbers of PD-1 positive CD8 T cells were found in draining lymph nodes, peripheral blood and tumor tissues three days after 10 mg/kg oxaliplatin treatment, and increased numbers of CD8 T cells and apoptotic tumor cells were detected at the edge of tumor tissues. Further investigation showed that the death of tumor cells induced by platinum compounds promoted T cell activation. Moreover, increased expression of T cell-attracting chemokines (CXCL9, CXCL10 and CCL5) was detected in MC38 cells after platinum treatment. These data indicated that the optimal dose of platinum chemotherapy could trigger T cell activation and recruitment into tumors, and sequential PD-1 blockade could prevent newly arriving T cell from becoming exhausted in tumor sites. These findings highlight the importance of optimizing the dose and timing of platinum chemotherapy combined with PD-1 blockade and provide an indication for the improvement of combined therapies in clinical trials.
    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
    Considering the existence of immune-desert in tumor microenvironment, the clinical efficacy of immunotherapy for lung adenocarcinoma is limited. This study aims to investigate the ability of transcription factors in regulating tumor immune microenvironment in lung adenocarcinoma. RNA-seq data were collected from the The Cancer Genome Atlas database. The relationships between transcription factors and immune infiltrates were assessed. Runt-related transcription factor 3 (RUNX3)-associated immune pathways were investigated by the Kyoto Encyclopedia of Genes and Genomes, Gene Ontology, and Gene set enrichment analysis. Upregulated chemokines in the RUNX3-overexpressed cell line were determined by quantitative real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assay. These chemokines were further confirmed in RUNX3-downregulated cell lines. Immunochemistry was conducted to determine the expression of RUNX3, CCL3, CCL20, and the numbers of CD8+ T lymphocytes in human lung cancer tissues. Chemokine receptors in CD8+ T cells were explored by flow cytometry and immunofluorescence. T cell recruitment was investigated by transwell assay. After screening 406 transcription factors, RUNX3 was found strongly correlated T cells, cytotoxic lymphocytes, and CD8+ T cells. RUNX3 was associated with a variety of immunomodulators, including LAG3, CTLA-4, PD-1, and TIGIT. More importantly, RUNX3 was involved in immune-related pathways, especially immune cell migration-related pathways. Further investigation exhibited RUNX3 could upregulate CCL3 and CCL20 whose receptors CCR5 and CCR6 were upregulated in CD8+ effector T cells, while downregulation of RUNX3 decreased the expression of CCL3 and CCL20 and the infiltration of CD8+ T cells in RUNX3-downregulated lung cancer cell lines. Immunochemistry exhibited positive correlations of RUNX3 with CCL3 and CD8+ T cells in clinical lung adenocarcinoma samples. The chemotaxis assay proved RUNX3 could promote CD8+ T cell recruitment by upregulating CCL3 and CCL20. This study unearths RUNX3 related molecular mechanisms of tumor immune microenvironment and may reverse the immune-desert condition in lung adenocarcinoma and be combined with immune checkpoint blockade and adoptive cell therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号