Memory T Cells

记忆 T 细胞
  • 文章类型: Journal Article
    CAR-T cell therapy targeting CD19 and BCMA for relapsed or refractory hematopoietic tumors has been adopted in routine practice and has shown dramatic results. However, half of patients who achieve remission with CAR-T therapy eventually relapse, and thus efforts to improve the efficacy of CAR-T therapy are gaining momentum. Notably, studies have described innovative technologies that enable control of cell kinetics after infusion, which is not possible with conventional CAR-T therapies. In this article, we review the challenges of CAR-T cell therapy and the development of new technologies.
    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:组织内记忆CD103+CD8+T细胞(CD103+CD8+TRMs)是抗肿瘤免疫的重要组成部分。然而,CD103+CD8+TRMs在结直肠癌(CRC)中的意义及其优势尚不清楚.
    方法:使用临床数据和标本评估CD103+CD8+TRMs在CRC中的意义。通过小鼠皮下肿瘤发生模型和集落形成实验来评估CD103CD8TRMs的抗肿瘤作用。最后,使用流式细胞术评估肿瘤中CD103CD8TRMs的浸润密度和功能。
    结果:在这项研究中,我们发现,在CRC患者中,高度浸润的CD103+CD8+TRMs与早期临床分期和VEGF阴性表达相关,并预测CRC/CRC肝转移患者的预后良好.有趣的是,我们还发现,CD103+CD8+TRMs可能对CRC中是否发生肝转移具有预测潜力.此外,我们发现CRC中α-SMA+血管数与α-SMA+和CD31+血管数之和呈正相关,和CD103+CD8+TRMs的浸润水平。此外,抗血管生成治疗促进CD103+CD8+TRMs的浸润并增强其分泌干扰素(IFN)-γ的能力,从而进一步提高抗肿瘤效果。此外,体内实验表明,与外周血CD8+T细胞相比,CD103+CD8+TRMs输注回体内还可进一步促进CD8+T细胞浸润肿瘤,它们分泌IFN-γ的能力更强,从而产生更好的抗肿瘤效果。
    结论:我们证明了CD103+CD8+TRMs具有临床应用的潜力,并为联合抗肿瘤治疗策略提供了新思路。如抗肿瘤血管生成治疗和CAR-T联合免疫疗法。
    BACKGROUND: Tissue-resident memory CD103+CD8+ T cells (CD103+CD8+ TRMs) are important components of anti-tumor immunity. However, the significance of CD103+CD8+ TRMs in colorectal cancer (CRC) and their advantages remain unclear.
    METHODS: Clinical data and specimens were used to evaluate the significance of CD103+CD8+ TRMs in CRC. A mouse subcutaneous tumorigenesis model and colony-formation assay were conducted to evaluate the anti-tumor effects of CD103+CD8+ TRMs. Finally, the infiltration density and function of CD103+CD8+ TRMs in the tumors were evaluated using flow cytometry.
    RESULTS: In this study, we showed that highly infiltrated CD103+CD8+ TRMs were associated with earlier clinical stage and negative VEGF expression in CRC patients and predicted a favorable prognosis for CRC/CRC liver metastases patients. Interestingly, we also found that CD103+CD8+ TRMs may have predictive potential for whether CRC develops liver metastasis in CRC. In addition, we found a positive correlation between the ratio of the number of α-SMA+ vessels to the sum of the number of α-SMA+ and CD31+ vessels in CRC, and the infiltration level of CD103+CD8+ TRMs. In addition, anti-angiogenic therapy promoted infiltration of CD103+CD8+ TRMs and enhanced their ability to secrete interferon (IFN)-γ, thus further improving the anti-tumor effect. Moreover, in vivo experiments showed that compared with peripheral blood CD8+ T cells, CD103+CD8+ TRMs infused back into the body could also further promote CD8+ T cells to infiltrate the tumor, and they had a stronger ability to secrete IFN-γ, which resulted in better anti-tumor effects.
    CONCLUSIONS: We demonstrated that CD103+CD8+ TRMs have the potential for clinical applications and provide new ideas for combined anti-tumor therapeutic strategies, such as anti-tumor angiogenesis therapy and CAR-T combined 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
    许多病原体通过粘膜部位进入宿主。因此,因此,通过粘膜部位的局部中和干扰病原体进入是预防疾病的有效策略。粘膜施用的疫苗具有在粘膜部位诱导保护性免疫应答的潜力。这份手稿深入研究了粘膜疫苗接种的一些最新进展,特别关注佐剂技术的进步以及这些佐剂在增强疫苗抗呼吸道病原体功效中的作用。它强调了呼吸道粘膜免疫系统的解剖学和免疫学复杂性,强调粘膜分泌型IgA和组织固有记忆T细胞在局部免疫反应中的重要性。我们进一步讨论了通过传统的肠胃外疫苗接种方法诱导的免疫应答与粘膜给药策略,并探索通过粘膜途径免疫提供的保护性优势。
    Many pathogens enter the host through mucosal sites. Thus, interfering with pathogen entry through local neutralization at mucosal sites therefore is an effective strategy for preventing disease. Mucosally administered vaccines have the potential to induce protective immune responses at mucosal sites. This manuscript delves into some of the latest developments in mucosal vaccination, particularly focusing on advancements in adjuvant technologies and the role of these adjuvants in enhancing vaccine efficacy against respiratory pathogens. It highlights the anatomical and immunological complexities of the respiratory mucosal immune system, emphasizing the significance of mucosal secretory IgA and tissue-resident memory T cells in local immune responses. We further discuss the differences between immune responses induced through traditional parenteral vaccination approaches vs. mucosal administration strategies, and explore the protective advantages offered by immunization through mucosal routes.
    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
    区域免疫监视依赖于不同记忆T细胞群体的共同努力。其中,组织驻留记忆T(TRM)细胞被策略性地定位在屏障组织中,在那里他们能够有效的前线防御感染和癌症。然而,这些细胞的长期持久性与多种免疫介导的病理有关。因此,调节TRM细胞群体代表了针对基于组织的疾病的新型疫苗接种和治疗性干预的有吸引力的策略。这里,我们提供了跨组织和疾病状态的TRM细胞异质性和功能的最新概述.我们讨论了TRM细胞介导的免疫保护机制及其对自身免疫性疾病的潜在贡献。最后,我们研究了TRM细胞反应如何持久增强或抑制治疗增益。
    Regionalized immune surveillance relies on the concerted efforts of diverse memory T cell populations. Of these, tissue-resident memory T (TRM) cells are strategically positioned in barrier tissues, where they enable efficient frontline defense against infections and cancer. However, the long-term persistence of these cells has been implicated in a variety of immune-mediated pathologies. Consequently, modulating TRM cell populations represents an attractive strategy for novel vaccination and therapeutic interventions against tissue-based diseases. Here, we provide an updated overview of TRM cell heterogeneity and function across tissues and disease states. We discuss mechanisms of TRM cell-mediated immune protection and their potential contributions to autoimmune disorders. Finally, we examine how TRM cell responses might be durably boosted or dampened for therapeutic gain.
    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
    KLRG1+CD8T细胞在急性感染清除后持续数月,并维持高水平的效应分子,提供针对全身病原体的保护性免疫。继发感染时,这些长寿效应细胞(LLEC)不能形成其他循环的KLRG1-记忆亚群,如中枢和效应记忆T细胞.因此,KLRG1+记忆T细胞通常被称为寿命相对较短的终末分化群体。这里,我们证明在病毒感染小鼠后,来自LLEC的效应细胞迅速进入非淋巴组织并减少病原体负担,但在很大程度上依赖于从血管内皮细胞接收抗原信号。单细胞RNA测序显示,由KLRG1或KLRG1-记忆前体产生的非淋巴组织中的次级记忆细胞会产生类似的常驻记忆转录签名。因此,虽然LLECs不能分化为其他循环记忆群体,他们仍然保留进入组织和建立居住权的灵活性。
    KLRG1+ CD8 T cells persist for months after clearance of acute infections and maintain high levels of effector molecules, contributing protective immunity against systemic pathogens. Upon secondary infection, these long-lived effector cells (LLECs) are incapable of forming other circulating KLRG1- memory subsets such as central and effector memory T cells. Thus, KLRG1+ memory T cells are frequently referred to as a terminally differentiated population that is relatively short lived. Here, we show that after viral infection of mice, effector cells derived from LLECs rapidly enter nonlymphoid tissues and reduce pathogen burden but are largely dependent on receiving antigen cues from vascular endothelial cells. Single-cell RNA sequencing reveals that secondary memory cells in nonlymphoid tissues arising from either KLRG1+ or KLRG1- memory precursors develop a similar resident memory transcriptional signature. Thus, although LLECs cannot differentiate into other circulating memory populations, they still retain the flexibility to enter tissues and establish residency.
    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: Clinical Trial, Phase II
    背景:在患有血液系统恶性肿瘤的儿科患者移植后复发仍然是一个挑战。用于疾病控制的清髓治疗方案与急性和长期不良反应相关。我们使用CD45RA耗尽的单倍体移植物过继转移记忆T细胞并结合NK细胞回输,并假设最大化移植物抗白血病(GVL)效应可能会降低预处理方案的强度。
    方法:在本II期临床试验(NCT01807611)中,72例血液恶性肿瘤患者(完全缓解(CR)1:25,≥CR2:28,难治性疾病:19)接受了富含CD34的单倍体和CD45RA的造血祖细胞移植物,然后进行NK细胞输注。调理包括氟达拉滨,Thiotepa,melphalan,环磷酰胺,总淋巴照射,移植物抗宿主病(GVHD)的预防包括短程西罗莫司或霉酚酸酯,无需血清疗法。
    结果:CR1患者的3年总生存率(OS)和无事件生存率(EFS)分别为92%(95%CI:72-98)和88%(95%CI:67-96);≥CR2为81%(95%CI:61-92)和68%(95%CI:47-82),难治性疾病为32%(95%CI:54-6)。所有患者形态CR的3年EFS为77%(95%CI:64-87),在有或没有微小残留病的接受者之间没有差异(P=0.2992)。免疫重建很快,在第30天,平均CD3和CD4T细胞计数为410/μL和140/μL。急性GVHD和慢性GVHD的累积发生率分别为36%和26%,但大多数急性GVHD患者通过治疗迅速恢复。NK细胞同种反应性供体观察到III-IV级急性GVHD的发生率较低(P=0.004),母体供者的中度/重度慢性GVHD发生率更高(P=0.035)。
    结论:CD45RA耗尽的移植物和NK细胞回补的组合导致了强大的免疫重建,最大限度地提高了GVL效应,并允许使用清髓性下,与优秀的EFS相关的无TBI预处理方案在该高危人群中产生有希望的长期结果。该试验在ClinicalTrials.gov(NCT01807611)注册。
    BACKGROUND: Relapse remains a challenge after transplantation in pediatric patients with hematological malignancies. Myeloablative regimens used for disease control are associated with acute and long-term adverse effects. We used a CD45RA-depleted haploidentical graft for adoptive transfer of memory T cells combined with NK-cell addback and hypothesized that maximizing the graft-versus-leukemia (GVL) effect might allow for reduction in intensity of conditioning regimen.
    METHODS: In this phase II clinical trial (NCT01807611), 72 patients with hematological malignancies (complete remission (CR)1: 25, ≥ CR2: 28, refractory disease: 19) received haploidentical CD34 + enriched and CD45RA-depleted hematopoietic progenitor cell grafts followed by NK-cell infusion. Conditioning included fludarabine, thiotepa, melphalan, cyclophosphamide, total lymphoid irradiation, and graft-versus-host disease (GVHD) prophylaxis consisted of a short-course sirolimus or mycophenolate mofetil without serotherapy.
    RESULTS: The 3-year overall survival (OS) and event-free-survival (EFS) for patients in CR1 were 92% (95% CI:72-98) and 88% (95% CI: 67-96); ≥ CR2 were 81% (95% CI: 61-92) and 68% (95% CI: 47-82) and refractory disease were 32% (95% CI: 11-54) and 20% (95% CI: 6-40). The 3-year EFS for all patients in morphological CR was 77% (95% CI: 64-87) with no difference amongst recipients with or without minimal residual disease (P = 0.2992). Immune reconstitution was rapid, with mean CD3 and CD4 T-cell counts of 410/μL and 140/μL at day + 30. Cumulative incidence of acute GVHD and chronic GVHD was 36% and 26% but most patients with acute GVHD recovered rapidly with therapy. Lower rates of grade III-IV acute GVHD were observed with NK-cell alloreactive donors (P = 0.004), and higher rates of moderate/severe chronic GVHD occurred with maternal donors (P = 0.035).
    CONCLUSIONS: The combination of a CD45RA-depleted graft and NK-cell addback led to robust immune reconstitution maximizing the GVL effect and allowed for use of a submyeloablative, TBI-free conditioning regimen that was associated with excellent EFS resulting in promising long-term outcomes in this high-risk population. The trial is registered at ClinicalTrials.gov (NCT01807611).
    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
    我们以前报道过纳米脉冲治疗(NPT),脉冲功率技术,导致4T1-luc乳腺肿瘤消除和强烈的原位疫苗接种,从而完全保护无肿瘤动物免受第二次活肿瘤攻击。NPT在4T1乳腺癌中主要是免疫抑制性肿瘤微环境(TME)中产生有效的抗肿瘤免疫反应的机制仍未得到解决。在这项研究中,用NPT治疗原位4T1小鼠乳腺肿瘤(100ns,50kV/cm,1000个脉冲,3Hz)。血,脾,脾引流淋巴结,肿瘤在4小时后被采集,8-h,1天,3天,7天,和3个月的治疗后间隔的频率分析,死亡,和各种免疫细胞的功能标志物,以及调节性T细胞(Tregs)的抑制功能。NPT被证实能引发针对乳腺癌的强原位疫苗接种(ISV)并促进急性和长期T细胞记忆。NPT通过大幅减少Tregs,在系统和TME中消除了免疫抑制优势,骨髓来源的抑制细胞(MDSCs),和肿瘤相关巨噬细胞(TAMs)。NPT诱导Tregs和TAMs细胞凋亡。它还在功能上降低了Treg抑制能力,通过激活标记的下调来解释,特别是4-1BB和TGFβ,和表型从主要活化的(CD44+CD62L-)转变为未活化的(CD44-CD62L+)Treg。重要的是,NPT在活化的Tregs和幸免效应CD4+和CD8+T细胞中选择性诱导凋亡。这些变化伴随着CD8CD103组织驻留的记忆T细胞和TAMM1极化的增加。这些发现表明,NPT有效地将TME和次级淋巴系统从免疫抑制状态转换为免疫刺激状态,允许细胞毒性T细胞功能和免疫记忆形成以消除癌细胞并解释NPT原位疫苗接种。
    We previously reported that nano-pulse treatment (NPT), a pulsed power technology, resulted in 4T1-luc mammary tumor elimination and a strong in situ vaccination, thereby completely protecting tumor-free animals against a second live tumor challenge. The mechanism whereby NPT mounts effective antitumor immune responses in the 4T1 breast cancer predominantly immunosuppressive tumor microenvironment (TME) remains unanswered. In this study, orthotopic 4T1 mouse breast tumors were treated with NPT (100 ns, 50 kV/cm, 1000 pulses, 3 Hz). Blood, spleen, draining lymph nodes, and tumors were harvested at 4-h, 8-h, 1-day, 3-day, 7-day, and 3-month post-treatment intervals for the analysis of frequencies, death, and functional markers of various immune cells in addition to the suppressor function of regulatory T cells (Tregs). NPT was verified to elicit strong in situ vaccination (ISV) against breast cancer and promote both acute and long-term T cell memory. NPT abolished immunosuppressive dominance systemically and in the TME by substantially reducing Tregs, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs). NPT induced apoptosis in Tregs and TAMs. It also functionally diminished the Treg suppression capacity, explained by the downregulation of activation markers, particularly 4-1BB and TGFβ, and a phenotypic shift from predominantly activated (CD44+CD62L-) to naïve (CD44-CD62L+) Tregs. Importantly, NPT selectively induced apoptosis in activated Tregs and spared effector CD4+ and CD8+ T cells. These changes were followed by a concomitant rise in CD8+CD103+ tissue-resident memory T cells and TAM M1 polarization. These findings indicate that NPT effectively switches the TME and secondary lymphatic systems from an immunosuppressive to an immunostimulatory state, allowing cytotoxic T cell function and immune memory formation to eliminate cancer cells and account for the NPT in situ vaccination.
    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
    巨细胞病毒(CMV)的再激活是同种异体干细胞移植后的重要问题。虽然先前的研究强调了γδT细胞在免疫受损移植患者中的抗CMV再激活作用,CMV再激活高危受者的特征仍然有限.
    本研究的重点是处于CMV再激活高风险的D+/R+受者(供者和受者均为CMV血清阳性)。我们分析了28例异基因造血干细胞移植后100天内出现CMV复发的患者,以及36名未经历CMV复发的匹配接受者.比较两组的临床数据,并确定了CMV再激活的危险因素。此外,测量CMV病毒载量,并进行流式细胞术分析以评估外周血γδT细胞比例的变化,亚群分布,和差异化地位。我们还分析了不同γδT细胞亚群中TCRδ链的CDR3库。通过在刺激时测量CMV感染的细胞的裂解来进行功能分析。
    移植后CMV的再激活与急性移植物抗宿主病(aGvHD)和非CMV疱疹病毒的再激活有关。值得注意的是,CMV再激活导致γδT细胞持续扩增,主要在Vδ2negγδT细胞亚群内,从幼稚细胞分化为效应记忆细胞的趋势。对δ链CDR3库的分析显示,CMV再激活后Vδ2negγδT细胞中克隆多样性的重建延迟,而Vδ2posT细胞未受影响。在用CMV感染的MRC5细胞刺激时,Vδ2negγδT细胞亚群作为产生IFN-γ并能够裂解CMV感染细胞的主要效应细胞群出现。此外,我们的发现表明,NKG2D不一定参与Vδ2negγδT细胞介导的抗CMV细胞毒性。
    这项研究为γδT细胞在CMV感染高风险的移植受体中对CMV再激活的免疫反应中的作用提供了新的见解。具体来说,Vδ2negγδT细胞亚群似乎与CMV再激活密切相关,强调它们在控制感染和反映HSCT患者CMV再激活方面的潜在作用。
    UNASSIGNED: Cytomegalovirus (CMV) reactivation is a significant concern following allogeneic stem cell transplantation. While previous research has highlighted the anti-CMV reactivation effect of γδ T cells in immunocompromised transplant patients, their characterization in recipients at high risk of CMV reactivation remains limited.
    UNASSIGNED: This study focused on D+/R+ recipients (where both donor and recipient are CMV seropositive) at high risk of CMV reactivation. We analyzed 28 patients who experienced CMV recurrence within 100 days post-allogeneic hematopoietic stem cell transplantation, along with 36 matched recipients who did not experience CMV recurrence. Clinical data from both groups were compared, and risk factors for CMV reactivation were identified. Additionally, CMV viral load was measured, and flow cytometric analysis was conducted to assess changes in peripheral blood γδ T cell proportions, subpopulation distribution, and differentiation status. We also analyzed the CDR3 repertoire of the TCR δ chain in different γδ T cell subsets. Functional analysis was performed by measuring the lysis of CMV-infected cells upon stimulation.
    UNASSIGNED: CMV reactivation post-transplantation was associated with acute graft-versus-host disease (aGvHD) and reactivation of non-CMV herpesviruses. Notably, CMV reactivation led to sustained expansion of γδ T cells, primarily within the Vδ2neg γδ T cell subpopulation, with a trend toward differentiation from Naive to effector memory cells. Analysis of the δ chain CDR3 repertoire revealed a delay in the reconstitution of clonal diversity in Vδ2neg γδ T cells following CMV reactivation, while Vδ2pos T cells remained unaffected. Upon stimulation with CMV-infected MRC5 cells, the Vδ2neg γδ T cell subpopulation emerged as the primary effector cell group producing IFN-γ and capable of lysing CMV-infected cells. Moreover, our findings suggest that NKG2D is not necessary involved in Vδ2neg γδ T cell-mediated anti-CMV cytotoxicity.
    UNASSIGNED: This study provides novel insights into the role of γδ T cells in the immune response to CMV reactivation in transplantation recipients at high risk of CMV infection. Specifically, the Vδ2neg γδ T cell subpopulation appears to be closely associated with CMV reactivation, underscoring their potential role in controlling infection and reflecting CMV reactivation in HSCT patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Editorial
    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
    记忆CD4T细胞对人体免疫至关重要,然而,目前尚不清楚记忆形成过程中的病毒性炎症是否具有长期后果。这里,我们比较了Spike(S)特异性记忆CD4T细胞的转录和表观遗传特征,这些个体首次暴露于S是通过SARS-CoV-2感染或mRNA疫苗接种.记忆形成近2年后,通过感染建立的S特异性CD4T细胞仍然富集了与细胞毒性相关的转录本和干扰素刺激的基因,可能是因为炎症改变了染色质可及性景观。此外,与疫苗引发的细胞相比,感染引发的S特异性CD4T细胞在体外的增殖能力降低。此外,S特异性记忆CD4T细胞的转录状态在加强免疫和/或突破性感染时发生了最小程度的改变.因此,感染相关的炎症持久印记CD4T细胞记忆,这会影响这些细胞的功能,并可能对长期免疫产生影响。
    Memory CD4 T cells are critical to human immunity, yet it is unclear whether viral inflammation during memory formation has long-term consequences. Here, we compared transcriptional and epigenetic landscapes of Spike (S)-specific memory CD4 T cells in 24 individuals whose first exposure to S was via SARS-CoV-2 infection or mRNA vaccination. Nearly 2 years after memory formation, S-specific CD4 T cells established by infection remained enriched for transcripts related to cytotoxicity and for interferon-stimulated genes, likely because of a chromatin accessibility landscape altered by inflammation. Moreover, S-specific CD4 T cells primed by infection had reduced proliferative capacity in vitro relative to vaccine-primed cells. Furthermore, the transcriptional state of S-specific memory CD4 T cells was minimally altered by booster immunization and/or breakthrough infection. Thus, infection-associated inflammation durably imprints CD4 T cell memory, which affects the function of these cells and may have consequences for long-term immunity.
    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