TLR signaling

TLR 信号
  • 文章类型: Journal Article
    I型干扰素(IFN-I)和IFN-γ通过促进T细胞应答来促进抗肿瘤免疫。矛盾的是,IFN可以通过激活免疫检查点来促进T细胞耗尽。这些完全不同的响应的下游调节器还没有被完全理解。这里,我们描述了干扰素调节因子1(IRF1)如何协调IFN的这些相反作用.肿瘤细胞中的IRF1表达通过阻止免疫细胞中干扰素刺激的基因(ISG)和效应程序来阻断Toll样受体和IFN-I依赖性宿主抗肿瘤免疫。相比之下,IRF1在宿主中的表达是抗肿瘤免疫所必需的。机械上,IRF1在肿瘤细胞中的免疫抑制而非免疫刺激ISG的启动子处明显结合或与STAT1结合。在Irf1-/-肿瘤中程序性细胞死亡配体1(PD-L1)的过表达仅部分恢复肿瘤生长,提示IRF1对抗肿瘤免疫的多因素作用。因此,我们确定IRF1在肿瘤细胞中的表达与宿主IFN-I和IRF1依赖性抗肿瘤免疫相反,以促进免疫逃逸和肿瘤生长。
    Type I interferon (IFN-I) and IFN-γ foster antitumor immunity by facilitating T cell responses. Paradoxically, IFNs may promote T cell exhaustion by activating immune checkpoints. The downstream regulators of these disparate responses are incompletely understood. Here, we describe how interferon regulatory factor 1 (IRF1) orchestrates these opposing effects of IFNs. IRF1 expression in tumor cells blocks Toll-like receptor- and IFN-I-dependent host antitumor immunity by preventing interferon-stimulated gene (ISG) and effector programs in immune cells. In contrast, expression of IRF1 in the host is required for antitumor immunity. Mechanistically, IRF1 binds distinctly or together with STAT1 at promoters of immunosuppressive but not immunostimulatory ISGs in tumor cells. Overexpression of programmed cell death ligand 1 (PD-L1) in Irf1-/- tumors only partially restores tumor growth, suggesting multifactorial effects of IRF1 on antitumor immunity. Thus, we identify that IRF1 expression in tumor cells opposes host IFN-I- and IRF1-dependent antitumor immunity to facilitate immune escape and tumor growth.
    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
    内酶体外切核酸酶PLD3和PLD4(磷脂酶D3和D4)与自身炎症和自身免疫性疾病有关。我们报告了这些酶的结构,以及它们催化的分子基础。该结构揭示了在界面处形成基本活性位点的链内二聚体拓扑结构。像其他PLD超家族成员一样,PLD3和PLD4携带HxKxxxxD/E基序并参与磷酸二酯键裂解。这些酶以5'至3'的方式消化ssDNA和ssRNA,并被5'磷酸化阻断。我们在apo中捕获了结构,中间,和产品状态,并揭示了“链接和释放”两步催化。我们还意外地通过共价3-磷酸组氨酸中间体证明了磷酸酶活性。PLD4含有额外的疏水钳,其稳定底物并且可以影响寡核苷酸底物偏好和产物释放。PLD3/4的疾病相关突变体的生化和结构分析显示了降低的酶活性或热稳定性以及疾病关联的可能基础。此外,这些发现提供了对治疗设计的见解.
    Endolysosomal exonucleases PLD3 and PLD4 (phospholipases D3 and D4) are associated with autoinflammatory and autoimmune diseases. We report structures of these enzymes, and the molecular basis of their catalysis. The structures reveal an intra-chain dimer topology forming a basic active site at the interface. Like other PLD superfamily members, PLD3 and PLD4 carry HxKxxxxD/E motifs and participate in phosphodiester-bond cleavage. The enzymes digest ssDNA and ssRNA in a 5\'-to-3\' manner and are blocked by 5\'-phosphorylation. We captured structures in apo, intermediate, and product states and revealed a \"link-and-release\" two-step catalysis. We also unexpectedly demonstrated phosphatase activity via a covalent 3-phosphohistidine intermediate. PLD4 contains an extra hydrophobic clamp that stabilizes substrate and could affect oligonucleotide substrate preference and product release. Biochemical and structural analysis of disease-associated mutants of PLD3/4 demonstrated reduced enzyme activity or thermostability and the possible basis for disease association. Furthermore, these findings provide insight into therapeutic design.
    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
    细胞信号传导途径内的RNA-蛋白质相互作用对RNA结合蛋白(RBPs)效应子功能具有显著的调节作用。在先天免疫反应期间,特异性RNA-蛋白质相互作用已被报道为转录后控制的调节层。我们研究了脂多糖(LPS)处理后永生化小鼠巨噬细胞(IMM)的RNA结合蛋白质组的变化。在细胞的细胞培养物(SILAC)中通过氨基酸进行稳定同位素标记,然后在LPS刺激后的两个时间点对RNP复合物进行无偏倚的纯化,并通过LC-MS/MS进行自下而上的蛋白质组学分析,产生了一组明显受影响的RBP。全局RNA测序和LFQ蛋白质组学用于表征在不同时间点响应于LPS的转录物和蛋白质丰度变化与蛋白质-RNA结合变化的相关性。Il1α,MARCKS,和ACOD1被标记为参与先天免疫信号传导的RBP候选物。通过使用核酸酶P1从洗脱后剩余的肽中消化交联的寡核苷酸来研究RBP和RNA缀合物在氨基酸分辨率时的结合位点。组合的数据集为通过RBP与不同类型的RNA相互作用的先天免疫信号调节的进一步研究提供了方向。
    RNA-protein interactions within cellular signaling pathways have significant modulatory effects on RNA binding proteins\' (RBPs\') effector functions. During the innate immune response, specific RNA-protein interactions have been reported as a regulatory layer of post-transcriptional control. We investigated changes in the RNA-bound proteome of immortalized mouse macrophages (IMM) following treatment with lipopolysaccharide (LPS). Stable isotope labeling by amino acids in cell culture (SILAC) of cells followed by unbiased purification of RNP complexes at two time points after LPS stimulation and bottom-up proteomic analysis by LC-MS/MS resulted in a set of significantly affected RBPs. Global RNA sequencing and LFQ proteomics were used to characterize the correlation of transcript and protein abundance changes in response to LPS at different time points with changes in protein-RNA binding. Il1α, MARCKS, and ACOD1 were noted as RBP candidates involved in innate immune signaling. The binding sites of the RBP and RNA conjugates at amino acid resolution were investigated by digesting the cross-linked oligonucleotide from peptides remaining after elution using Nuclease P1. The combined data sets provide directions for further studies of innate immune signaling regulation by RBP interactions with different classes of RNA.
    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受体相关激酶4(IRAK4)在toll样受体(TLR)信号通路中具有重要作用,这种分子的功能障碍除了癌症之外,还可能导致各种感染和免疫相关的疾病。IRAK4基因变异与各种类型的疾病有关。因此,我们进行了全面的分析,以识别对IRAK4最具破坏性影响的错义变体,并采用不同的生物信息学工具来研究单核苷酸多态性对功能的影响,稳定性,二级结构,和3D结构。还研究了残基在蛋白质结构域上的位置及其保守状态。此外,对接工具和结构生物学参与分析SNP对一种已开发的IRAK4抑制剂的影响。通过分析IRAK4基因SNPs,分析将十个变体区分为最有害的错义变体。所有变体位于重要蛋白质结构域上的高度保守位置。L318S和L318F突变与IRAK4二级结构的变化有关。通过I-Mutant2.0和Mu-Pro工具,发现八个SNP对IRAK4的稳定性具有降低的作用,而Mu-Pro工具确定了G198ESNP的降低效应。此外,还发现了所选变体对IRAK4的3D结构的不利影响。分子建模研究强调了这些鉴定的SNP突变残基对IRAK4ATP结合位点对已知靶抑制剂的成药性的有害影响。HG-12-6,与天然蛋白质相比。失去重要的配体残基接触,改变蛋白质的整体灵活性,增加的空间碰撞,甚至在配体结合位点界面处的电子惩罚都被认为与SNP模型有关,以阻碍HG-12-6对IRAK4靶蛋白的亲和力。该给定模型为更好地预测与IRAK4故障相关的各种疾病奠定了基础,并阐明了有害的IRAK4变体对IRAK4抑制剂功效的影响。
    Interleukin-1-receptor-associated kinase 4 (IRAK4) possesses a crucial function in the toll-like receptor (TLR) signaling pathway, and the dysfunction of this molecule could lead to various infectious and immune-related diseases in addition to cancers. IRAK4 genetic variants have been linked to various types of diseases. Therefore, we conducted a comprehensive analysis to recognize the missense variants with the most damaging impacts on IRAK4 with the employment of diverse bioinformatics tools to study single-nucleotide polymorphisms\' effects on function, stability, secondary structures, and 3D structure. The residues\' location on the protein domain and their conservation status were investigated as well. Moreover, docking tools along with structural biology were engaged in analyzing the SNPs\' effects on one of the developed IRAK4 inhibitors. By analyzing IRAK4 gene SNPs, the analysis distinguished ten variants as the most detrimental missense variants. All variants were situated in highly conserved positions on an important protein domain. L318S and L318F mutations were linked to changes in IRAK4 secondary structures. Eight SNPs were revealed to have a decreasing effect on the stability of IRAK4 via both I-Mutant 2.0 and Mu-Pro tools, while Mu-Pro tool identified a decreasing effect for the G198E SNP. In addition, detrimental effects on the 3D structure of IRAK4 were also discovered for the selected variants. Molecular modeling studies highlighted the detrimental impact of these identified SNP mutant residues on the druggability of the IRAK4 ATP-binding site towards the known target inhibitor, HG-12-6, as compared to the native protein. The loss of important ligand residue-wise contacts, altered protein global flexibility, increased steric clashes, and even electronic penalties at the ligand-binding site interfaces were all suggested to be associated with SNP models for hampering the HG-12-6 affinity towards IRAK4 target protein. This given model lays the foundation for the better prediction of various disorders relevant to IRAK4 malfunction and sheds light on the impact of deleterious IRAK4 variants on IRAK4 inhibitor efficacy.
    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
    糖尿病患者更容易受到SARS-CoV-2神经系统表现的影响。SARS-CoV-2诱导糖尿病脑血管功能障碍的分子机制尚不清楚。我们假设SARS-CoV-2通过激活肾素-血管紧张素-醛固酮系统(RAAS)和Toll样受体(TLR)信号的破坏性臂,加剧了糖尿病引起的脑血管氧化应激和炎症。在人源化ACE2转基因敲入小鼠中注射SARS-CoV-2刺突蛋白。认知功能,脑血流量,脑血管结构,RAAS,和TLR信号用于确定SARS-CoV-2刺突蛋白在糖尿病中的作用。使用用高葡萄糖条件培养基处理的人脑微血管内皮细胞在体外模拟糖尿病状况。Spike蛋白加剧了糖尿病诱导的脑血管氧化应激,炎症,和内皮细胞死亡导致血管稀疏增加和脑血流量减少。与对照小鼠相比,SARS-CoV-2刺突蛋白使糖尿病的认知功能障碍恶化。尖峰蛋白以RAAS保护臂为代价增强了破坏性的RAAS臂。并行,spike蛋白显著加剧了糖尿病患者的TLR信号,加重炎症和细胞凋亡的恶性循环。我们的研究表明SAR-CoV-2刺突蛋白增强了糖尿病患者的RAAS和TLR信号,增加脑血管损伤和认知功能障碍。
    Diabetics are more vulnerable to SARS-CoV-2 neurological manifestations. The molecular mechanisms of SARS-CoV-2-induced cerebrovascular dysfunction in diabetes are unclear. We hypothesize that SARS-CoV-2 exacerbates diabetes-induced cerebrovascular oxidative stress and inflammation via activation of the destructive arm of the renin-angiotensin-aldosterone system (RAAS) and Toll-like receptor (TLR) signaling. SARS-CoV-2 spike protein was injected in humanized ACE2 transgenic knock-in mice. Cognitive functions, cerebral blood flow, cerebrovascular architecture, RAAS, and TLR signaling were used to determine the effect of SARS-CoV-2 spike protein in diabetes. Studies were mirrored in vitro using human brain microvascular endothelial cells treated with high glucose-conditioned media to mimic diabetic conditions. Spike protein exacerbated diabetes-induced cerebrovascular oxidative stress, inflammation, and endothelial cell death resulting in an increase in vascular rarefaction and diminished cerebral blood flow. SARS-CoV-2 spike protein worsened cognitive dysfunction in diabetes compared to control mice. Spike protein enhanced the destructive RAAS arm at the expense of the RAAS protective arm. In parallel, spike protein significantly exacerbated TLR signaling in diabetes, aggravating inflammation and cellular apoptosis vicious circle. Our study illustrated that SAR-CoV-2 spike protein intensified RAAS and TLR signaling in diabetes, increasing cerebrovascular damage and cognitive dysfunction.
    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
    一些研究已经确定MYD88L265P基因中的突变是几种B细胞淋巴瘤中的关键驱动突变。携带MYD88L265P突变的B细胞淋巴瘤与磷酸化布鲁顿酪氨酸激酶(BTK)形成复合物,并对BTK抑制有反应。然而,B细胞淋巴瘤中的BTK抑制很少导致完全缓解,并且大多数患者经历最终疾病复发。通过下游分子如白介素1受体相关激酶4(IRAK-4)持续存活信号,\"mydosome\"复合体的一个组成部分,已显示在用BTK抑制剂治疗的B细胞淋巴瘤患者中具有组成活性。新的证据表明IRAK-4抑制B细胞淋巴瘤的治疗益处,以及可能逆转BTK抑制剂抗性。虽然MYD88基因突变在骨髓性恶性肿瘤中不存在,在急性髓细胞性白血病(AML)和骨髓增生异常综合征(MDS)中发现了IRAK-4的致癌长链形式的下游过度表达,特别是在AML和MDS中含有剪接因子U2AF1和SF3B1突变。这些数据表明IRAK-4抑制的抗白血病活性可用于复发性/难治性(R/R)AML/MDS。在这篇评论文章中,我们讨论了目前可用的emavusertib的临床前和临床数据,一个选择性的,口服生物可利用的IRAK-4抑制剂治疗R/RB细胞淋巴瘤和髓样恶性肿瘤。
    Several studies have identified mutations in the MYD88L265P gene as a key driver mutation in several B-cell lymphomas. B-cell lymphomas that harbor the MYD88L265P mutation form a complex with phosphorylated Bruton\'s tyrosine kinase (BTK) and are responsive to BTK inhibition. However, BTK inhibition in B-cell lymphomas rarely results in a complete response and most patients experience eventual disease relapse. Persistent survival signaling though downstream molecules such as interleukin 1 receptor-associated kinase 4 (IRAK-4), an integral part of the \"myddosome\" complex, has been shown to be constitutively active in B-cell lymphoma patients treated with BTK inhibitors. Emerging evidence is demonstrating the therapeutic benefit of IRAK-4 inhibition in B-cell lymphomas, along with possibly reversing BTK inhibitor resistance. While MYD88 gene mutations are not present in myeloid malignancies, downstream overexpression of the oncogenic long form of IRAK-4 has been found in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS), particularly in AML and MDS that harbor mutations in splicing factors U2AF1 and SF3B1. These data suggest that the anti-leukemic activity of IRAK-4 inhibition can be exploited in relapsed/refractory (R/R) AML/MDS. In this review article, we discuss the currently available pre-clinical and clinical data of emavusertib, a selective, orally bioavailable IRAK-4 inhibitor in the treatment of R/R B-cell lymphomas and myeloid malignancies.
    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
    亚单位或灭活疫苗包含大多数针对病毒和细菌病原体使用的疫苗。然而,与它们的活/减毒对应物相比,这些疫苗通常表现出降低的免疫原性,需要多种加强剂和/或佐剂来引发保护性免疫反应。出于这个原因,佐剂及其改善灭活疫苗反应的机制的研究对于开发具有更高效力的疫苗至关重要。研究表明,佐剂与抗原的直接结合促进了疫苗的免疫原性,具有佐剂和抗原靶向同一细胞的优势。使用这种直接联系的策略,我们开发了一种灭活的甲型流感(IAV)疫苗,该疫苗通过异双功能交联剂与Toll样受体7/8激动剂瑞喹莫德(R848)直接偶联.以前,我们发现,与无佐剂疫苗相比,该疫苗可改善新生非人灵长类动物的保护和病毒清除能力.我们随后发现,用于将R848与病毒缀合的接头的选择改变了疫苗的刺激活性,促进体外分化的DC成熟和促炎细胞因子的产生。有了这些知识,我们探讨了交联剂的选择如何影响这些疫苗的刺激活性.我们发现,接头的选择改变了人单核细胞衍生的树突状细胞(moDC)中通过NF-κB途径的信号传导。Further,我们将分析扩展到人外周血中存在的体内分化APC,复制在体外分化细胞中发现的接头依赖性差异。最后,我们在小鼠模型中证明,接头的选择会影响接种疫苗时产生的IAV特异性IgG抗体的量.这些数据增强了我们对用于改善疫苗免疫原性的缀合方法的理解。
    Subunit or inactivated vaccines comprise the majority of vaccines used against viral and bacterial pathogens. However, compared to their live/attenuated counterparts, these vaccines often demonstrate reduced immunogenicity, requiring multiple boosters and or adjuvants to elicit protective immune responses. For this reason, studies of adjuvants and the mechanism through which they can improve inactivated vaccine responses are critical for the development of vaccines with increased efficacy. Studies have shown that the direct conjugation of adjuvant to antigen promotes vaccine immunogenicity, with the advantage of both the adjuvant and antigen targeting the same cell. Using this strategy of direct linkage, we developed an inactivated influenza A (IAV) vaccine that is directly conjugated with the Toll-like receptor 7/8 agonist resiquimod (R848) through a heterobifunctional crosslinker. Previously, we showed that this vaccine resulted in improved protection and viral clearance in newborn nonhuman primates compared to a non-adjuvanted vaccine. We subsequently discovered that the choice of linker used to conjugate R848 to the virus alters the stimulatory activity of the vaccine, promoting increased maturation and proinflammatory cytokine production from DC differentiated in vitro. With this knowledge, we explored how the choice of crosslinker impacts the stimulatory activity of these vaccines. We found that the linker choice alters signaling through the NF-κB pathway in human monocyte-derived dendritic cells (moDCs). Further, we extended our analyses to in vivo differentiated APC present in human peripheral blood, replicating the linker-dependent differences found in in vitro differentiated cells. Finally, we demonstrated in a mouse model that the choice of linker impacts the amount of IAV-specific IgG antibody produced in response to vaccination. These data enhance our understanding of conjugation approaches for improving vaccine immunogenicity.
    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
    系统性红斑狼疮(SLE)是一种自身免疫性疾病,其特征是失去免疫耐受并产生针对核酸和其他核抗原(Ags)的自身抗体。B淋巴细胞在SLE的免疫发病机制中起重要作用。多种受体控制SLE患者的异常B细胞活化,包括内在Toll样受体(TLRs),B细胞受体(BCRs),和细胞因子受体。TLRs的作用,特别是TLR7和TLR9,在SLE的病理生理学方面近年来得到了广泛的探讨。当内源性或外源性核酸配体被BCR识别并内化到B细胞中时,它们结合TLR7或TLR9以激活相关的信号通路,从而控制B细胞的增殖和分化。令人惊讶的是,TLR7和TLR9似乎在SLEB细胞中起相反的作用,它们之间的相互作用仍然知之甚少。此外,其他细胞可以通过释放促进B细胞分化为浆细胞的细胞因子来增强SLE患者B细胞中的TLR信号传导。因此,TLR7和TLR9如何调节SLE中B细胞的异常激活,可能有助于理解SLE的发病机制,并为TLR靶向治疗SLE提供指导.
    Systemic lupus erythematosus (SLE) is an autoimmune illness marked by the loss of immune tolerance and the production of autoantibodies against nucleic acids and other nuclear antigens (Ags). B lymphocytes are important in the immunopathogenesis of SLE. Multiple receptors control abnormal B-cell activation in SLE patients, including intrinsic Toll-like receptors (TLRs), B-cell receptors (BCRs), and cytokine receptors. The role of TLRs, notably TLR7 and TLR9, in the pathophysiology of SLE has been extensively explored in recent years. When endogenous or exogenous nucleic acid ligands are recognized by BCRs and internalized into B cells, they bind TLR7 or TLR9 to activate related signalling pathways and thus govern the proliferation and differentiation of B cells. Surprisingly, TLR7 and TLR9 appear to play opposing roles in SLE B cells, and the interaction between them is still poorly understood. In addition, other cells can enhance TLR signalling in B cells of SLE patients by releasing cytokines that accelerate the differentiation of B cells into plasma cells. Therefore, the delineation of how TLR7 and TLR9 regulate the abnormal activation of B cells in SLE may aid the understanding of the mechanisms of SLE and provide directions for TLR-targeted therapies for SLE.
    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
    虽然Toll样受体(TLR)信号诱导的炎症是抵抗感染所必需的,持续的炎症会损害宿主组织,并导致无数的急性和慢性炎症性疾病。因此,不仅TLR信号在病原体存在下被激活,而且TLR信号最终被终止是必要的。限制持续TLR信号传导的一种机制是选择性前mRNA剪接。除了编码产生促进炎症的蛋白质的典型mRNA,TLR信号通路中的许多基因也编码可选择的mRNAs,这些mRNAs产生的蛋白质是信号的显性负性抑制剂.许多这些负调节因子是由免疫攻击诱导的,因此,这些替代同工型的产生代表了一个限制持续炎症的负反馈回路。虽然这些选择性剪接事件已经在一个基因的基础上进行了研究,对这种终止TLR信号传导的机制的系统分析有限.在这里,我们回顾了在TLR信号通路中产生负面作用的替代亚型的已知情况,包括这些抑制剂如何发挥作用。它们是如何产生的,以及它们在炎症性疾病中可能扮演的角色。
    While inflammation induced by Toll-like receptor (TLR) signaling is required to combat infection, persistent inflammation can damage host tissues and contribute to a myriad of acute and chronic inflammatory disorders. Thus, it is essential not only that TLR signaling be activated in the presence of pathogens but that TLR signaling is ultimately terminated. One mechanism that limits persistent TLR signaling is alternative pre-mRNA splicing. In addition to encoding the canonical mRNAs that produce proteins that promote inflammation, many genes in the TLR signaling pathway also encode alternative mRNAs that produce proteins that are dominant negative inhibitors of signaling. Many of these negative regulators are induced by immune challenge, so production of these alternative isoforms represents a negative feedback loop that limits persistent inflammation. While these alternative splicing events have been investigated on a gene by gene basis, there has been limited systemic analysis of this mechanism that terminates TLR signaling. Here we review what is known about the production of negatively acting alternative isoforms in the TLR signaling pathway including how these inhibitors function, how they are produced, and what role they may play in inflammatory disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号