lymphotoxin beta receptor

淋巴毒素 β 受体
  • 文章类型: Journal Article
    调节性T细胞(Treg)是免疫耐受的关键参与者,其通过涉及CD25+FoxP3-和CD25-Foxp3lo前体的两种不同的发育途径在胸腺中发展。然而,调节最近发现的Foxp3lo前体途径的机制尚不清楚.这里,我们发现,在TCR/CD28和IL-2刺激下,膜结合型淋巴毒素α1β2(LTα1β2)异质复合物在Treg发育过程中上调。我们表明Lta表达通过调节它们的增殖限制了Treg从Foxp3lo前体的成熟发育,生存,和代谢概况。转基因报告小鼠和转录组分析进一步揭示髓质胸腺上皮细胞(mTEC)构成IL-4的意外来源。我们证明了LTα1β2-淋巴毒素β受体介导的与mTEC的相互作用通过下调mTEC中IL-4的表达来限制Treg的发育。总的来说,我们的研究发现,淋巴毒素轴是胸腺Treg发育的第一个抑制性检查点,它通过限制IL-4的可用性来微调Foxp3loTreg前体途径.
    Regulatory T cells (Treg) are critical players of immune tolerance that develop in the thymus via two distinct developmental pathways involving CD25+Foxp3- and CD25-Foxp3lo precursors. However, the mechanisms regulating the recently identified Foxp3lo precursor pathway remain unclear. Here, we find that the membrane-bound lymphotoxin α1β2 (LTα1β2) heterocomplex is upregulated during Treg development upon TCR/CD28 and IL-2 stimulation. We show that Lta expression limits the maturational development of Treg from Foxp3lo precursors by regulating their proliferation, survival, and metabolic profile. Transgenic reporter mice and transcriptomic analyses further reveal that medullary thymic epithelial cells (mTEC) constitute an unexpected source of IL-4. We demonstrate that LTα1β2-lymphotoxin β receptor-mediated interactions with mTEC limit Treg development by down-regulating IL-4 expression in mTEC. Collectively, our findings identify the lymphotoxin axis as the first inhibitory checkpoint of thymic Treg development that fine-tunes the Foxp3lo Treg precursor pathway by limiting IL-4 availability.
    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
    嗜酸性粒细胞参与针对多细胞生物体的宿主保护。然而,在2型免疫期间,它们招募到肠系膜淋巴结(mLN)的研究不足。我们的结果表明,在B细胞上选择性缺乏白细胞介素(IL)-4Rα或淋巴毒素β(LTβ)表达的小鼠中,嗜酸性粒细胞与成纤维细胞网状细胞(FRC)和淋巴内皮细胞构建的淋巴间质壁ni的关联减少。此外,嗜酸性粒细胞存活,激活,和增强的Il1rl1受体表达由基质细胞和B细胞对话驱动。淋巴毒素β受体(LTβR)在FRC上的连接改善了嗜酸性粒细胞的存活,并显着增加了IL-33的表达和嗜酸性粒细胞归巢到mLN,从而证实了淋巴毒素信号对粒细胞募集的意义。嗜酸性粒细胞缺陷型ΔdblGATA-1小鼠显示mLN扩增减少,滤泡间区(IFR)alarmin表达减少,推迟了蠕虫清除,阐明它们在2型免疫中的重要性。这些发现为基质细胞和B细胞之间的对话提供了见解,控制MLN嗜酸性粒细胞增多,以及这些机制在2型免疫过程中的相关性。
    Eosinophils are involved in host protection against multicellular organisms. However, their recruitment to the mesenteric lymph node (mLN) during type 2 immunity is understudied. Our results demonstrate that eosinophil association with lymphoid stromal niches constructed by fibroblastic reticular cells (FRCs) and lymphatic endothelial cells is diminished in mice selectively lacking interleukin (IL)-4Rα or lymphotoxin-β (LTβ) expression on B cells. Furthermore, eosinophil survival, activation, and enhanced Il1rl1 receptor expression are driven by stromal cell and B cell dialogue. The ligation of lymphotoxin-β receptor (LTβR) on FRCs improves eosinophil survival and significantly augments IL-33 expression and eosinophil homing to the mLN, thus confirming the significance of lymphotoxin signaling for granulocyte recruitment. Eosinophil-deficient ΔdblGATA-1 mice show diminished mLN expansion, reduced interfollicular region (IFR) alarmin expression, and delayed helminth clearance, elucidating their importance in type 2 immunity. These findings provide insight into dialogue between stromal cells and B cells, which govern mLN eosinophilia, and the relevance of these mechanisms during type 2 immunity.
    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
    实体器官移植仍然是终末期器官衰竭的救命治疗,但慢性排斥反应仍然是影响长期同种异体移植结局的主要障碍,目前尚未有实质性改善.三级淋巴器官(TLO)是在慢性炎症条件下形成的异位淋巴结构,来自人类移植的证据表明,TLO在接受慢性排斥的同种异体移植物中经常形成。在这项研究中,我们利用小鼠肾移植模型和操作的淋巴毒素α(LTα)-淋巴毒素β受体(LTβR)途径,这对TLO的形成至关重要,明确TLO在移植中的作用。我们表明,在同种异体移植物中唯一的淋巴器官是TLO的模型中,移植物内TLO足以激活同种免疫反应并介导移植物排斥。当移植到具有正常次级淋巴器官的受体上时,移植物TLO或LTα过表达的存在加速了排斥反应。如果LTβR通路在供体移植物中被破坏,TLO的形成被取消,移植物存活时间延长。肾TLO的活体显微镜检查表明,TLO中的局部T和B细胞活化与在次级淋巴器官中观察到的相似。总之,我们证明了TLO中的免疫激活有助于局部免疫反应,导致早期同种异体移植失败。因此,TLO和LTαβ-LTβR途径是限制局部免疫应答和防止同种异体移植物排斥的主要靶标。这些发现适用于其他疾病,如自身免疫或肿瘤,其中限制或加强局部免疫反应是有益的,并改善疾病的结果。
    Solid organ transplantation remains the life-saving treatment for end-stage organ failure, but chronic rejection remains a major obstacle to long-term allograft outcomes and has not improved substantially. Tertiary lymphoid organs (TLOs) are ectopic lymphoid structures that form under conditions of chronic inflammation, and evidence from human transplantation suggests that TLOs regularly form in allografts undergoing chronic rejection. In this study, we utilized a mouse renal transplantation model and manipulation of the lymphotoxin αβ/lymphotoxin β receptor (LTαβ/LTβR) pathway, which is essential for TLO formation, to define the role of TLOs in transplantation. We showed that intragraft TLOs are sufficient to activate the alloimmune response and mediate graft rejection in a model where the only lymphoid organs are TLOs in the allograft. When transplanted to recipients with a normal set of secondary lymphoid organs, the presence of graft TLOs or LTα overexpression accelerated rejection. If the LTβR pathway was disrupted in the donor graft, TLO formation was abrogated, and graft survival was prolonged. Intravital microscopy of renal TLOs demonstrated that local T and B cell activation in TLOs is similar to that observed in secondary lymphoid organs. In summary, we demonstrated that immune activation in TLOs contributes to local immune responses, leading to earlier allograft failure. TLOs and the LTαβ/LTβR pathway are therefore prime targets to limit local immune responses and prevent allograft rejection. These findings are applicable to other diseases, such as autoimmune diseases or tumors, where either limiting or boosting local immune responses is beneficial and improves disease outcomes.
    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
    细胞毒性药物引起的肠道损伤后,控制粘膜愈合的复杂免疫机制仍然知之甚少。这项研究的目的是研究淋巴毒素β受体(LTβR)信号在化疗诱导的肠道损伤中的作用。LTβR缺陷小鼠表现出更高的体重减轻,加剧肠道病理,促炎细胞因子表达增加,IL-22表达减少,甲氨蝶呤(MTX)治疗后肠上皮细胞的增殖。此外,LTβR-/-IL-22-/-小鼠死于MTX治疗,提示LTβR-和IL-22-依赖性途径共同促进粘膜修复。尽管LTβR配体LIGHT和LTβ在MTX治疗后早期在肠道中上调,LIGHT-/-小鼠,但不是LTβ-/-小鼠,显示加剧的疾病。Further,我们揭示了T细胞在粘膜修复中的关键作用,因为T细胞缺陷小鼠未能上调肠道LIGHT表达,并表现出体重减轻和肠道病理增加。对LTβR条件性失活的小鼠的分析表明,LTβR在肠上皮细胞中的信号传导,但不是在Lgr5+肠干细胞中,巨噬细胞或树突状细胞对粘膜修复至关重要。此外,肠上皮细胞中非经典NF-kB途径成员RelB的失活促进了MTX诱导的疾病。基于这些结果,我们提出了一个模型,其中T细胞产生的LIGHT激活肠上皮细胞中的LTβR-RelB信号传导,以促进化疗后的粘膜修复.
    The intricate immune mechanisms governing mucosal healing following intestinal damage induced by cytotoxic drugs remain poorly understood. The goal of this study was to investigate the role of lymphotoxin beta receptor (LTβR) signaling in chemotherapy-induced intestinal damage. LTβR deficient mice exhibited heightened body weight loss, exacerbated intestinal pathology, increased proinflammatory cytokine expression, reduced IL-22 expression, and proliferation of intestinal epithelial cells following methotrexate (MTX) treatment. Furthermore, LTβR-/-IL-22-/- mice succumbed to MTX treatment, suggesting that LTβR- and IL-22- dependent pathways jointly promote mucosal repair. Although both LTβR ligands LIGHT and LTβ were upregulated in the intestine early after MTX treatment, LIGHT-/- mice, but not LTβ-/- mice, displayed exacerbated disease. Further, we revealed the critical role of T cells in mucosal repair as T cell-deficient mice failed to upregulate intestinal LIGHT expression and exhibited increased body weight loss and intestinal pathology. Analysis of mice with conditional inactivation of LTβR revealed that LTβR signaling in intestinal epithelial cells, but not in Lgr5+ intestinal stem cells, macrophages or dendritic cells was critical for mucosal repair. Furthermore, inactivation of the non-canonical NF-kB pathway member RelB in intestinal epithelial cells promoted MTX-induced disease. Based on these results, we propose a model wherein LIGHT produced by T cells activates LTβR-RelB signaling in intestinal epithelial cells to facilitate mucosal repair following chemotherapy treatment.
    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
    银屑病(PS)和特应性皮炎(AD)是以高反应性角质形成细胞为特征的常见皮肤炎症性疾病。虽然,一些细胞因子被认为对每种疾病都有特异性,其他细胞因子可能是这两种疾病的核心。这里,我们显示肿瘤坏死因子超家族成员14(TNFSF14),被称为光,是实验PS所需要的,类似于其在实验AD中的要求。老鼠没有光,或缺失其任一受体,淋巴毒素β受体(LTβR)和疱疹病毒进入介质(HVEM),在角质形成细胞中,被保护免受咪喹莫特诱导的银屑病特征的发展,包括表皮增厚和增生,和PS相关基因的表达。相应地,在PS患者活检的单细胞RNA-seq分析中,发现LTβR转录物在角质形成细胞中与HVEM强烈表达,T细胞中LIGHT上调。在AD活检中也看到了类似的转录物表达谱,角质形成细胞中的LTβR缺失也保护小鼠免受过敏原诱导的AD特征。此外,在体外,LIGHT上调了人角质形成细胞中广谱的基因,这些基因是PS和AD皮肤病变的临床特征。我们的数据表明,阻断LIGHT活性的药物可能对PS和AD的治疗干预有用。
    Psoriasis (PS) and atopic dermatitis (AD) are common skin inflammatory diseases characterized by hyper-responsive keratinocytes. Although, some cytokines have been suggested to be specific for each disease, other cytokines might be central to both diseases. Here, we show that Tumor necrosis factor superfamily member 14 (TNFSF14), known as LIGHT, is required for experimental PS, similar to its requirement in experimental AD. Mice devoid of LIGHT, or deletion of either of its receptors, lymphotoxin β receptor (LTβR) and herpesvirus entry mediator (HVEM), in keratinocytes, were protected from developing imiquimod-induced psoriatic features, including epidermal thickening and hyperplasia, and expression of PS-related genes. Correspondingly, in single cell RNA-seq analysis of PS patient biopsies, LTβR transcripts were found strongly expressed with HVEM in keratinocytes, and LIGHT was upregulated in T cells. Similar transcript expression profiles were also seen in AD biopsies, and LTβR deletion in keratinocytes also protected mice from allergen-induced AD features. Moreover, in vitro, LIGHT upregulated a broad spectrum of genes in human keratinocytes that are clinical features of both PS and AD skin lesions. Our data suggest that agents blocking LIGHT activity might be useful for therapeutic intervention in PS as well as in AD.
    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
    淋巴毒素β受体(LTBR)是一种阳性的T细胞增殖调控基因。它与肿瘤免疫微环境密切相关。然而,其在癌症和免疫治疗中的作用尚不清楚.首先,分析LTBR的表达水平和预后价值。其次,LTBR在临床分期中的表达,免疫亚型,并对分子亚型进行了分析。LTBR与免疫调节基因的相关性,免疫检查点基因,然后分析RNA修饰基因。LTBR与免疫细胞的相关性,分数,癌症相关的功能状态,肿瘤干性指数,错配修复(MMR)基因,和DNA甲基转移酶也被分析。此外,我们分析了LTBR在DNA甲基化中的作用,突变状态,肿瘤突变负荷(TMB),和微卫星不稳定性(MSI)。基因本体论(GO),京都基因和基因组百科全书(KEGG),和基因集富集分析(GSEA)用于探索LTBR在泛癌症中的作用。最后,分析了与LTBR相关的药物.使用定量实时PCR和Western印迹确认LTBR的表达。LTBR在大多数癌症中显著过表达,并且与低患者存活率相关。此外,LTBR表达与免疫细胞密切相关,得分,癌症相关的功能状态,肿瘤干性指数,MMR基因,DNA甲基转移酶,DNA甲基化,突变状态,TMB,和MSI。富集分析显示LTBR与细胞凋亡有关,坏死,和免疫相关途径。最后,确定了多种靶向LTBR的药物。LTBR在几种肿瘤中过表达并且与不良预后相关。它与免疫相关基因和免疫细胞浸润有关。
    Lymphotoxin beta receptor (LTBR) is a positive T cell proliferation regulator gene. It is closely associated with the tumor immune microenvironment. However, its role in cancer and immunotherapy is unclear. Firstly, the expression level and prognostic value of LTBR were analyzed. Secondly, the expression of LTBR in clinical stages, immune subtypes, and molecular subtypes was analyzed. The correlation between LTBR and immune regulatory genes, immune checkpoint genes, and RNA modification genes was then analyzed. Correlations between LTBR and immune cells, scores, cancer-related functional status, tumor stemness index, mismatch repair (MMR) genes, and DNA methyltransferase were also analyzed. In addition, we analyzed the role of LTBR in DNA methylation, mutational status, tumor mutation burden (TMB), and microsatellite instability (MSI). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA) were used to explore the role of LTBR in pan-cancer. Finally, the drugs associated with LTBR were analyzed. The expression of LTBR was confirmed using quantitative real-time PCR and Western blot. LTBR is significantly overexpressed in most cancers and is associated with low patient survival. In addition, LTBR expression was strongly correlated with immune cells, score, cancer-related functional status, tumor stemness index, MMR genes, DNA methyltransferase, DNA methylation, mutational status, TMB, and MSI. Enrichment analysis revealed that LTBR was associated with apoptosis, necroptosis, and immune-related pathways. Finally, multiple drugs targeting LTBR were identified. LTBR is overexpressed in several tumors and is associated with a poor prognosis. It is related to immune-related genes and immune cell infiltration.
    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
    肝细胞癌(HCC)是最常见的原发性肝肿瘤,在全球范围内发病率越来越高。HCC是一种异质性恶性肿瘤,通常在慢性损伤的肝脏中发展。核因子κB(NF-κB)信号网络由规范和非规范分支组成。记录了HCC中经典NF-κB的激活。然而,非规范NF-κB及其下游效应物的功能和临床相关作用尚未确定。
    评估了四个人HCC队列(总n=1,462)和四个小鼠HCC模型的NF-κB信号传导组分和激活配体的表达和定位。体外,NF-κB信号,测量增殖和细胞死亡,证明通过NIK激活的RELB的促增殖作用。在体内,淋巴毒素β(LTβ)被鉴定为RELB激活的主要诱导物。重要的是,小鼠HCC模型中肝细胞特异性RELB敲除导致较低的发病率和较低的最大肿瘤直径。在硅片中,使用推断的蛋白质活性和基因集富集分析(GSEA)在TCGAHCC队列上验证RELB活性和RELB定向转录组学。在RELB活性肝癌中,介导增殖的通路被显著激活。与RELA相比,非经典RELB表达的核富集以独立于病因学的方式确定了预后不良的患者。此外,RELB激活与恶性特征转移和复发有关。
    结论:这项研究表明,在肝癌发生中LTβ/LTβR/RELB轴的病因独立和跨物种一致激活。这些观察结果可能对HCC产生广泛的影响,包括可能的临床开发。
    OBJECTIVE: HCC is the most common primary liver tumor, with an increasing incidence worldwide. HCC is a heterogeneous malignancy and usually develops in a chronically injured liver. The NF-κB signaling network consists of a canonical and a noncanonical branch. Activation of canonical NF-κB in HCC is documented. However, a functional and clinically relevant role of noncanonical NF-κB and its downstream effectors is not established.
    RESULTS: Four human HCC cohorts (total n = 1462) and 4 mouse HCC models were assessed for expression and localization of NF-κB signaling components and activating ligands. In vitro , NF-κB signaling, proliferation, and cell death were measured, proving a pro-proliferative role of v-rel avian reticuloendotheliosis viral oncogene homolog B (RELB) activated by means of NF-κB-inducing kinase. In vivo , lymphotoxin beta was identified as the predominant inducer of RELB activation. Importantly, hepatocyte-specific RELB knockout in a murine HCC model led to a lower incidence compared to controls and lower maximal tumor diameters. In silico , RELB activity and RELB-directed transcriptomics were validated on the The Cancer Genome Atlas HCC cohort using inferred protein activity and Gene Set Enrichment Analysis. In RELB-active HCC, pathways mediating proliferation were significantly activated. In contrast to v-rel avian reticuloendotheliosis viral oncogene homolog A, nuclear enrichment of noncanonical RELB expression identified patients with a poor prognosis in an etiology-independent manner. Moreover, RELB activation was associated with malignant features metastasis and recurrence.
    CONCLUSIONS: This study demonstrates a prognostically relevant, etiology-independent, and cross-species consistent activation of a lymphotoxin beta/LTβR/RELB axis in hepatocarcinogenesis. These observations may harbor broad implications for HCC, including possible clinical exploitation.
    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
    尽管进行了十多年的临床试验,将抑制新出现的检查点与PD-1/L1抑制剂骨架相结合,PD-1/L1抑制剂耐药或难治性实体瘤尚未显示有意义的生存益处,尤其是以骨髓抑制微环境为主的肿瘤。因此,实现持久的抗肿瘤免疫可能需要适应性和先天免疫刺激的组合。骨髓复极化,增强APC活化和抗原加工/呈递,解除CD47/SIRPα(分化簇47/信号调节蛋白α)\'不吃我\'信号,提供凋亡\'pro-eatme\'或\'找到我\'信号,以及封锁免疫检查站.最近强调了有效靶向mLILRB2和SIRPAyeloid细胞以提高应答率的重要性,鉴于骨髓细胞在大多数实体瘤的肿瘤微环境中丰富。TNFSF14或LIGHT,是一种肿瘤坏死超家族配体,具有广泛的适应性和先天免疫活性,包括(1)通过淋巴毒素β受体(LTβR)激活骨髓细胞,(2)T/NK(T细胞和自然杀伤细胞)通过疱疹病毒进入介质(HVEM)诱导抗肿瘤免疫活性,(3)通过LTβR增强肿瘤基质细胞的促炎细胞因子/趋化因子分泌,(4)直接诱导体外肿瘤细胞凋亡,和(5)淋巴组织结构的重组,包括在肿瘤微环境(TME)内,通过促进高内皮小静脉(HEV)的形成和三级淋巴结构的诱导。LTBR(淋巴毒素β受体)和HVEM在实体瘤的一系列共刺激受体中排名很高,这引起了人们对考虑LIGHT介导的共刺激如何与越来越多的免疫疗法靶标不同的兴趣,这些靶标未能作为单一疗法或与PD-1抑制剂联合提供生存益处,特别是在检查点获得的抗性设置中。
    Despite over a decade of clinical trials combining inhibition of emerging checkpoints with a PD-1/L1 inhibitor backbone, meaningful survival benefits have not been shown in PD-1/L1 inhibitor resistant or refractory solid tumours, particularly tumours dominated by a myelosuppressive microenvironment. Achieving durable anti-tumour immunity will therefore likely require combination of adaptive and innate immune stimulation, myeloid repolarisation, enhanced APC activation and antigen processing/presentation, lifting of the CD47/SIRPα (Cluster of Differentiation 47/signal regulatory protein alpha) \'do not eat me\' signal, provision of an apoptotic \'pro-eat me\' or \'find me\' signal, and blockade of immune checkpoints. The importance of effectively targeting mLILRB2 and SIRPAyeloid cells to achieve improved response rates has recently been emphasised, given myeloid cells are abundant in the tumour microenvironment of most solid tumours. TNFSF14, or LIGHT, is a tumour necrosis superfamily ligand with a broad range of adaptive and innate immune activities, including (1) myeloid cell activation through Lymphotoxin Beta Receptor (LTβR), (2) T/NK (T cell and natural killer cell) induced anti-tumour immune activity through Herpes virus entry mediator (HVEM), (3) potentiation of proinflammatory cytokine/chemokine secretion through LTβR on tumour stromal cells, (4) direct induction of tumour cell apoptosis in vitro, and (5) the reorganisation of lymphatic tissue architecture, including within the tumour microenvironment (TME), by promoting high endothelial venule (HEV) formation and induction of tertiary lymphoid structures. LTBR (Lymphotoxin beta receptor) and HVEM rank highly amongst a range of costimulatory receptors in solid tumours, which raises interest in considering how LIGHT-mediated costimulation may be distinct from a growing list of immunotherapy targets which have failed to provide survival benefit as monotherapy or in combination with PD-1 inhibitors, particularly in the checkpoint acquired resistant setting.
    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
    已知急性成淋巴细胞和成髓性白血病(ALL和AML)改变骨髓微环境并破坏非恶性造血。然而,驱动这些改变的分子机制仍然不明确。使用ALL和AML的小鼠模型,在这里,我们显示白血病细胞在定植骨髓后不久就关闭了淋巴生成和红细胞生成。ALL和AML细胞表达淋巴毒素α1β2并激活间充质干细胞(MSCs)中的淋巴毒素β受体(LTβR)信号,从而关闭IL7的产生并防止非恶性淋巴细胞生成。我们发现DNA损伤应答途径和CXCR4信号促进白血病细胞中淋巴毒素α1β2的表达。在MSCs中LTβR信号的遗传或药理学破坏可恢复淋巴细胞生成而不是红细胞生成,减少白血病细胞生长,并显著延长移植受者的生存期。同样,CXCR4阻断还可以防止白血病诱导的IL7下调并抑制白血病生长。这些研究表明,急性白血病利用控制造血输出的生理机制作为获得竞争优势的策略。
    Acute lymphoblastic and myeloblastic leukemias (ALL and AML) have been known to modify the bone marrow microenvironment and disrupt non-malignant hematopoiesis. However, the molecular mechanisms driving these alterations remain poorly defined. Using mouse models of ALL and AML, here we show that leukemic cells turn off lymphopoiesis and erythropoiesis shortly after colonizing the bone marrow. ALL and AML cells express lymphotoxin α1β2 and activate lymphotoxin beta receptor (LTβR) signaling in mesenchymal stem cells (MSCs), which turns off IL7 production and prevents non-malignant lymphopoiesis. We show that the DNA damage response pathway and CXCR4 signaling promote lymphotoxin α1β2 expression in leukemic cells. Genetic or pharmacological disruption of LTβR signaling in MSCs restores lymphopoiesis but not erythropoiesis, reduces leukemic cell growth, and significantly extends the survival of transplant recipients. Similarly, CXCR4 blocking also prevents leukemia-induced IL7 downregulation and inhibits leukemia growth. These studies demonstrate that acute leukemias exploit physiological mechanisms governing hematopoietic output as a strategy for gaining competitive advantage.
    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
    雌激素是通过雌激素受体α(ERα)在多发性硬化症(MS)及其动物模型实验性自身免疫性脑脊髓炎(EAE)中的疾病修饰因子。然而,ERα信号传导导致疾病发病机制变化的机制尚未完全阐明.这里,我们证明,在小鼠EAE模型中,小鼠树突状细胞(DC)中的ERα缺失诱导中枢神经系统严重的神经变性和对干扰素β(IFNβ)的抗性,一线MS治疗。由性腺外来源合成的雌激素对于控制疾病表型至关重要。机械上,激活的ERα直接与TLR4下游信号分子TRAF3相互作用,通过泛素化降解TRAF3,导致膜淋巴毒素(mLT)和IFNβ成分的IRF3核易位和转录减少。DC中ERα信号传导减弱通过mLT-淋巴毒素β受体(LTβR)信号传导产生神经毒性效应CD4+T细胞。淋巴毒素β受体拮抗剂消除了DC特异性ERα缺陷小鼠的EAE疾病症状。这些发现表明,雌激素来源于性腺外来源,如淋巴结,在DC中控制TRAF3介导的细胞因子产生以调节EAE疾病表型。
    Estrogen is a disease-modifying factor in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE) via estrogen receptor alpha (ERα). However, the mechanisms by which ERα signaling contributes to changes in disease pathogenesis have not been completely elucidated. Here, we demonstrate that ERα deletion in dendritic cells (DCs) of mice induces severe neurodegeneration in the central nervous system in a mouse EAE model and resistance to interferon beta (IFNβ), a first-line MS treatment. Estrogen synthesized by extragonadal sources is crucial for controlling disease phenotypes. Mechanistically, activated ERα directly interacts with TRAF3, a TLR4 downstream signaling molecule, to degrade TRAF3 via ubiquitination, resulting in reduced IRF3 nuclear translocation and transcription of membrane lymphotoxin (mLT) and IFNβ components. Diminished ERα signaling in DCs generates neurotoxic effector CD4+ T cells via mLT-lymphotoxin beta receptor (LTβR) signaling. Lymphotoxin beta receptor antagonist abolished EAE disease symptoms in the DC-specific ERα-deficient mice. These findings indicate that estrogen derived from extragonadal sources, such as lymph nodes, controls TRAF3-mediated cytokine production in DCs to modulate the EAE disease phenotype.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号