inflammatory monocytes

炎性单核细胞
  • 文章类型: Journal Article
    组织中的间质液作为淋巴通过传入淋巴管不断排入淋巴结(LN),并通过传出淋巴管从LN排入血液。LN被策略性地定位并且具有合适的细胞组成以充当针对入侵病原体的适应性免疫起始位点。然而,淋巴传播的病毒,通过淋巴系统从进入部位传播到其他组织,引流LN(dLN)中的免疫细胞在抑制原发性和继发性感染期间的全身性病毒传播中也起关键作用。组织中的淋巴携带病毒可以作为淋巴中或感染细胞内的游离病毒体被转运到dLN。不管进入机制如何,感染的骨髓抗原呈递细胞,包括树突状细胞的各种亚型,炎性单核细胞,和巨噬细胞,在启动dLN内的先天免疫应答中起关键作用。这种先天性免疫应答涉及感染和旁观者先天性免疫细胞之间的细胞串扰,所述先天性免疫细胞最终产生I型干扰素(IFN-Is)和其他细胞因子并募集炎性单核细胞和自然杀伤(NK)细胞。IFN-I和NK细胞的细胞毒性可以限制原发性感染期间的全身性病毒传播并预防严重的疾病。此外,驻留或快速迁移到dLN的记忆性CD8+T细胞可有助于在继发病毒感染期间预防疾病。这篇综述探讨了在含有原发性病毒感染的dLN中精心安排的复杂的先天免疫反应,以及继发性感染或CD8T细胞疫苗接种后记忆CD8T细胞的作用。
    The interstitial fluids in tissues are constantly drained into the lymph nodes (LNs) as lymph through afferent lymphatic vessels and from LNs into the blood through efferent lymphatics. LNs are strategically positioned and have the appropriate cellular composition to serve as sites of adaptive immune initiation against invading pathogens. However, for lymph-borne viruses, which disseminate from the entry site to other tissues through the lymphatic system, immune cells in the draining LN (dLN) also play critical roles in curbing systemic viral dissemination during primary and secondary infections. Lymph-borne viruses in tissues can be transported to dLNs as free virions in the lymph or within infected cells. Regardless of the entry mechanism, infected myeloid antigen-presenting cells, including various subtypes of dendritic cells, inflammatory monocytes, and macrophages, play a critical role in initiating the innate immune response within the dLN. This innate immune response involves cellular crosstalk between infected and bystander innate immune cells that ultimately produce type I interferons (IFN-Is) and other cytokines and recruit inflammatory monocytes and natural killer (NK) cells. IFN-I and NK cell cytotoxicity can restrict systemic viral spread during primary infections and prevent serious disease. Additionally, the memory CD8+ T-cells that reside or rapidly migrate to the dLN can contribute to disease prevention during secondary viral infections. This review explores the intricate innate immune responses orchestrated within dLNs that contain primary viral infections and the role of memory CD8+ T-cells following secondary infection or CD8+ T-cell vaccination.
    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
    我们已经获得了有关冠状病毒2(SARS-CoV-2)引起的严重急性呼吸道综合症的发病机理的重要知识。然而,导致疾病康复的潜在机制仍需充分了解.
    为了深入了解与COVID-19病因相关的关键免疫标志物,我们研究了从COVID-19中康复的患者外周血样本免疫谱的演变,并将其与患有严重急性呼吸道疾病但SARS-CoV-2检测阴性的受试者(对照)进行了比较.此外,确定了不同参数之间的线性和聚类相关性。
    获得的数据显示,出院时与出院时相比,炎性单核细胞(CD14CD16)的频率显着降低admission.值得注意的是,排出时单核细胞室产生的一氧化氮(NO)显着减少。此外,入院时白细胞介素(IL)-6的血浆水平与NOCD14CD16单核细胞的频率呈负相关。然而,在医院出院的时候,循环IL-6与单核细胞的NO产生率直接相关。根据这些观察,我们发现伴随着没有减少,在医院释放时,CD8T细胞上的硝基酪氨酸(NT)水平显着降低。考虑到嘌呤能信号构成另一个调节系统,我们分析了CD8T细胞中CD39和CD73胞外酶表达的动力学。我们发现,CD39+CD8+T细胞的频率显着减少,而CD73+细胞的百分比在出院时增加。体外,IL-6刺激COVID-19患者的PBMC会降低CD8T细胞上的NT水平。在NT+与NT-CD8+T细胞群。
    结果表明,感染后早期,IL-6控制NO的产生,它调节CD8T细胞上NT的水平,从而改变其效应子功能。有趣的是,在这种细胞毒性细胞群体中,嘌呤能胞外酶的表达与硝化表面分子的存在紧密相关。总的来说,获得的数据有助于更好地了解与COVID-19结局相关的致病机制.
    We have acquired significant knowledge regarding the pathogenesis of severe acute respiratory syndrome caused by coronavirus 2 (SARS-CoV-2). However, the underlying mechanisms responsible for disease recovery still need to be fully understood.
    To gain insights into critical immune markers involved in COVID-19 etiopathogenesis, we studied the evolution of the immune profile of peripheral blood samples from patients who had recovered from COVID-19 and compared them to subjects with severe acute respiratory illness but negative for SARS-CoV-2 detection (controls). In addition, linear and clustered correlations between different parameters were determined.
    The data obtained revealed a significant reduction in the frequency of inflammatory monocytes (CD14+CD16+) at hospital discharge vs. admission. Remarkably, nitric oxide (NO) production by the monocyte compartment was significantly reduced at discharge. Furthermore, interleukin (IL)-6 plasma levels were negatively correlated with the frequency of NO+CD14+CD16+ monocytes at hospital admission. However, at the time of hospital release, circulating IL-6 directly correlated with the NO production rate by monocytes. In line with these observations, we found that concomitant with NO diminution, the level of nitrotyrosine (NT) on CD8 T-cells significantly diminished at the time of hospital release. Considering that purinergic signaling constitutes another regulatory system, we analyzed the kinetics of CD39 and CD73 ectoenzyme expression in CD8 T-cells. We found that the frequency of CD39+CD8+ T-cells significantly diminished while the percentage of CD73+ cells increased at hospital discharge. In vitro, IL-6 stimulation of PBMCs from COVID-19 patients diminished the NT levels on CD8 T-cells. A clear differential expression pattern of CD39 and CD73 was observed in the NT+ vs. NT-CD8+ T-cell populations.
    The results suggest that early after infection, IL-6 controls the production of NO, which regulates the levels of NT on CD8 T-cells modifying their effector functions. Intriguingly, in this cytotoxic cell population, the expression of purinergic ectoenzymes is tightly associated with the presence of nitrated surface molecules. Overall, the data obtained contribute to a better understanding of pathogenic mechanisms associated with COVID-19 outcomes.
    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
    二甲双胍治疗可减弱实验性腹主动脉瘤(AAA)的形成,以及减少糖尿病患者的临床AAA直径扩大。二甲双胍介导的动脉瘤抑制机制,以及它在抑制已建立的实验性动脉瘤方面的功效,仍然不确定。
    通过主动脉内输注猪胰弹性蛋白酶在雄性C57BL/6J小鼠中产生实验性AAAs。单独使用二甲双胍(250mg/kg),或二甲双胍联合5'AMP活化蛋白激酶(AMPK)拮抗剂化合物C(10mg/kg),在AAA诱导后4天开始,每天给予相应的小鼠队列。其他AAA队列接受AMPK激动剂AICA核苷(500mg/kg)为阳性,或载体(盐水)为阴性,controls.通过连续体内超声检查和处死时的组织病理学评估所有组的AAA进展。通过流式细胞术分析确定产生细胞因子的T细胞和髓样细胞数量。
    与盐水对照相比,二甲双胍在治疗开始后3天(-85%)和10天(-68%)限制了已建立的实验性AAA进展。同时的化合物C处理使这种效果降低了大约50%。在二甲双胍治疗的小鼠中,AAA进展减少与相对弹性蛋白保存相关,平滑肌细胞保存,与载体对照组相比,壁白细胞浸润和新血管生成减少。二甲双胍还导致干扰素-γ-,但不是白细胞介素-10或-17,在动脉瘤小鼠中产生脾T细胞。此外,二甲双胍治疗增加循环和脾炎症单核细胞(CD11b+Ly-6Chigh),但不是中性粒细胞(CD11b+Ly-6G+),对各自的骨髓细胞群没有影响。
    二甲双胍治疗部分通过AMPK激动剂活性抑制现有的实验性AAA进展,限制产生干扰素γ的T细胞分化,同时增强循环和脾炎症单核细胞滞留。
    UNASSIGNED: Metformin treatment attenuates experimental abdominal aortic aneurysm (AAA) formation, as well as reduces clinical AAA diameter enlargement in patients with diabetes. The mechanisms of metformin-mediated aneurysm suppression, and its efficacy in suppressing established experimental aneurysms, remain uncertain.
    UNASSIGNED: Experimental AAAs were created in male C57BL/6J mice via intra-aortic infusion of porcine pancreatic elastase. Metformin alone (250 mg/kg), or metformin combined with the 5\' AMP-activated protein kinase (AMPK) antagonist Compound C (10 mg/kg), were administered to respective mouse cohorts daily beginning 4 days following AAA induction. Further AAA cohorts received either the AMPK agonist AICA riboside (500 mg/kg) as positive, or vehicle (saline) as negative, controls. AAA progression in all groups was assessed via serial in vivo ultrasonography and histopathology at sacrifice. Cytokine-producing T cells and myeloid cellularity were determined by flow cytometric analyses.
    UNASSIGNED: Metformin limited established experimental AAA progression at 3 (-85%) and 10 (-68%) days following treatment initiation compared with saline control. Concurrent Compound C treatment reduced this effect by approximately 50%. In metformin-treated mice, reduced AAA progression was associated with relative elastin preservation, smooth muscle cell preservation, and reduced mural leukocyte infiltration and neoangiogenesis compared with vehicle control group. Metformin also resulted in reduced interferon-γ-, but not interleukin-10 or -17, producing splenic T cells in aneurysmal mice. Additionally, metformin therapy increased circulating and splenic inflammatory monocytes (CD11b+Ly-6Chigh), but not neutrophils (CD11b+Ly-6G+), with no effect on respective bone marrow cell populations.
    UNASSIGNED: Metformin treatment suppresses existing experimental AAA progression in part via AMPK agonist activity, limiting interferon-γ-producing T cell differentiation while enhancing circulating and splenic inflammatory monocyte retention.
    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
    炎性单核细胞(iMO)和B细胞是小鼠淋巴结中痘病毒异位病毒(ECTV)的主要靶标,并在感染中发挥不同的作用。受感染和旁观者的IMO控制ECTV的系统传播,防止过早死亡,而B细胞产生抗体消除ECTV。我们的工作表明,在ECTV感染中幸存下来的受感染动物中,iMO和B细胞的内在和旁观者感染差异控制对免疫细胞功能重要的基因的转录,也许,细胞身份。旁观者细胞上调新陈代谢,抗原呈递,和干扰素刺激的基因。受感染的细胞下调许多细胞类型特异性基因,并上调非免疫细胞典型的转录本。旁观者(Bys)和感染的(Inf)iMO非冗余地有助于细胞因子环境和干扰素反应。此外,我们揭示了I型干扰素(IFN-I)或IFN-γ信号传导如何差异调节Inf和BysiMO的免疫途径,在稳定状态下,IFN-I启动iMO用于快速IFN-I产生和抗原呈递。
    Inflammatory monocytes (iMOs) and B cells are the main targets of the poxvirus ectromelia virus (ECTV) in the lymph nodes of mice and play distinct roles in surviving the infection. Infected and bystander iMOs control ECTV\'s systemic spread, preventing early death, while B cells make antibodies that eliminate ECTV. Our work demonstrates that within an infected animal that survives ECTV infection, intrinsic and bystander infection of iMOs and B cells differentially control the transcription of genes important for immune cell function and, perhaps, cell identity. Bystander cells upregulate metabolism, antigen presentation, and interferon-stimulated genes. Infected cells downregulate many cell-type-specific genes and upregulate transcripts typical of non-immune cells. Bystander (Bys) and infected (Inf) iMOs non-redundantly contribute to the cytokine milieu and the interferon response. Furthermore, we uncover how type I interferon (IFN-I) or IFN-γ signaling differentially regulates immune pathways in Inf and Bys iMOs and that, at steady state, IFN-I primes iMOs for rapid IFN-I production and antigen presentation.
    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
    炎性单核细胞是急性排斥人肾脏同种异体移植物中细胞浸润的主要成分。由于免疫修饰纳米颗粒(IMP)通过特异性清道夫受体MARCO与循环炎性单核细胞结合,导致转移到脾脏和随后的细胞凋亡,我们研究了带负电荷的治疗潜力,500纳米直径的聚苯乙烯IMP,以防止肾脏同种异体移植排斥。从BALB/c(H2d)向C57BL/6(H2b)小鼠进行肾脏移植,分为两组:对照(allo)和输注IMP的allo小鼠。研究组14(急性排斥)或100(慢性排斥)天。接受IMP的同种异体小鼠表现出优越的存活率和明显较少急性排斥反应,有更好的肾功能,少了根瘤炎,炎症细胞密度降低,与同种异体小鼠相比,同种异体移植物中的细胞因子和细胞毒性分子表达以及第14天血清中供体特异性IgG2c抗体的滴度较低。从接受IMPs的allo小鼠的肾脏中分离的细胞显示出减少的Ly6Chi单核细胞,CD11b+细胞和NKT+细胞与同种异体小鼠比较。IMP主要结合血流中的CD11b+细胞和脾脏中的CD11b+和CD11c-B220+边缘区B细胞。在脾脏里,IMP主要在红色纸浆中发现,与MARCO共定位并表达裂解的caspase-3。在第100天,接受IMP的同种异体小鼠表现出降低的巨噬细胞M1应答,但未被保护免于慢性排斥。IMP对急性排斥反应有显著的保护作用,抑制先天和适应性同种免疫。因此,我们目前的实验发现,再加上我们早期证明IMP在肾脏缺血再灌注损伤中的保护作用,确定IMP是肾移植的潜在诱导剂。
    Inflammatory monocytes are a major component of the cellular infiltrate in acutely rejecting human kidney allografts. Since immune-modifying nanoparticles (IMPs) bind to circulating inflammatory monocytes via the specific scavenger receptor MARCO, causing diversion to the spleen and subsequent apoptosis, we investigated the therapeutic potential of negatively charged, 500-nm diameter polystyrene IMPs to prevent kidney allograft rejection. Kidney transplants were performed from BALB/c (H2d) to C57BL/6 (H2b) mice in two groups: controls (allo) and allo mice infused with IMPs. Groups were studied for 14 (acute rejection) or 100 (chronic rejection) days. Allo mice receiving IMPs exhibited superior survival and markedly less acute rejection, with better kidney function, less tubulitis, and diminished inflammatory cell density, cytokine and cytotoxic molecule expression in the allograft and lower titers of donor-specific IgG2c antibody in serum at day 14, as compared to allo mice. Cells isolated from kidneys from allo mice receiving IMPs showed reduced Ly6Chi monocytes, CD11b+ cells and NKT+ cells compared to allo mice. IMPs predominantly bound CD11b+ cells in the bloodstream and CD11b+ and CD11c-B220+ marginal zone B cells in the spleen. In the spleen, IMPs were found predominantly in red pulp, colocalized with MARCO and expression of cleaved caspase-3. At day 100, allo mice receiving IMPs exhibited reduced macrophage M1 responses but were not protected from chronic rejection. IMPs afforded significant protection from acute rejection, inhibiting both innate and adaptive alloimmunity. Thus, our current experimental findings, coupled with our earlier demonstration of IMP-induced protection in kidney ischemia-reperfusion injury, identify IMPs as a potential induction agent in kidney transplantation.
    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
    在炎症和免疫的背景下,有零散的和观察性的研究,有关的药理活性的芒果及其主要活性成分,芒果苷.因此,我们旨在分析这种植物提取物的潜在有益作用(MIE,芒果苷中的90%)在痛风性关节炎的小鼠模型中,允许评估细胞免疫表型和MIE活性以外的生化机制。痛风性关节炎由MSU晶体(200μg20μl-1)的关节内给药诱导,而MIE(0.1-10mgkg-1)或相应的媒介物(DMSO/盐水1:3)与MSU同时口服(时间0),刺激后6和12小时。此后,除了对COX-2/mPGES-1轴进行westernblot分析外,还评估了膝关节评分和水肿.此外,对促炎/抗炎细胞趋化因子进行分析,并对细胞浸润物进行表型分析.用MIE治疗显示出关节炎症评分的剂量依赖性降低,在10mgkg-1时观察到最大抑制作用。MIE显着降低了炎症组织中白细胞的浸润和活化以及不同促炎细胞趋化因子的表达。此外,生化分析表明,MIE调节COX-2/mPGES-1和mPGDS-1/PPARγ途径。流式细胞术分析还强调了炎性单核细胞(CD11b/CD115/LY6Chi)的显着调节,MIE处理后的Treg细胞(CD4+/CD25+/FOXP3+)。总的来说,这项研究的结果证明了MIE在痛风性关节炎的发病和进展中积极影响局部和全身炎症/免疫紊乱的新功能.
    In the context of inflammation and immunity, there are fragmented and observational studies relating to the pharmacological activity of Mangifera indica L. and its main active component, mangiferin. Therefore, we aimed to analyze the potential beneficial effects of this plant extract (MIE, 90 % in mangiferin) in a mouse model of gouty arthritis, to allow the evaluation of cellular immune phenotypes and the biochemical mechanism/s beyond MIE activity. Gouty arthritis was induced by the intra-articular administration of MSU crystals (200 μg 20 µl-1), whereas MIE (0.1-10 mg kg-1) or corresponding vehicle (DMSO/saline 1:3) were orally administrated concomitantly with MSU (time 0), 6 and 12 h after the stimulus. Thereafter, knee joint score and oedema were evaluated in addition to western blot analysis for COX-2/mPGES-1 axis. Moreover, the analysis of pro/anti-inflammatory cyto-chemokines coupled with the phenotyping of the cellular infiltrate was performed. Treatment with MIE revealed a dose-dependent reduction in joint inflammatory scores with maximal inhibition observed at 10 mg kg-1. MIE significantly reduced leukocyte infiltration and activation and the expression of different pro-inflammatory cyto-chemokines in inflamed tissues. Furthermore, biochemical analysis revealed that MIE modulated COX-2/mPGES-1 and mPGDS-1/PPARγ pathways. Flow cytometry analysis also highlighted a prominent modulation of inflammatory monocytes (CD11b+/CD115+/LY6Chi), and Treg cells (CD4+/CD25+/FOXP3+) after MIE treatment. Collectively, the results of this study demonstrate a novel function of MIE to positively affect the local and systemic inflammatory/immunological perturbance in the onset and progression of gouty arthritis.
    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
    病原真菌的清除,荚膜组织胞浆,需要先天免疫和适应性免疫之间的合作。由于这种生物被吸入,肺巨噬细胞和树突状细胞(DC)是第一道防线。此外,DC作为APC驱动1型Th细胞产生IFNγ的教育,这有助于最终消除荚膜H.在这项研究中,我们使用肺组织胞浆菌病小鼠模型探讨了Notch信号在宿主防御中的重要性.我们发现在肺部和淋巴结感染后,吞噬细胞和IFNγCD4T细胞上的Notch配体(NL)和Notch受体(NRs)上调。为了确定Notch对感染过程的影响,我们使用了γ-分泌酶抑制剂(GSI),LY-411,575,其抑制NR下游信号传导。当在感染时或感染后7天给予时,该化合物损害真菌清除。然而,GSI不影响具有预先存在的免疫力的小鼠的真菌清除。宿主防御减弱与单核细胞衍生的DC和单核细胞衍生的巨噬细胞和肺泡巨噬细胞向M2的分化和成熟降低有关。我们的研究揭示了Notch信号在维持这种感染因子控制方面的关键性质。
    The clearance of the pathogenic fungus, Histoplasma capsulatum, requires cooperation between innate and adaptive immunity. Since this organism is inhaled, lung macrophages and dendritic cells (DCs) are the first lines of defense. Moreover, DCs act as APCs to drive the education of type 1 Th cells to produce IFNγ, which contributes to the final elimination of H. capsulatum. In this study, we explored the importance of Notch signaling in host defenses using a mouse model of pulmonary histoplasmosis. We found up-regulation of Notch ligands (NLs) and Notch receptors (NRs) on phagocytes and IFNγ+ CD4+ T cells upon infection in lungs and lymph nodes. To ascertain the influence of Notch on the course of infection, we used a gamma-secretase inhibitor (GSI), LY-411,575, which inhibits NR downstream signaling. This compound impaired fungal clearance when given at the time of infection or 7 days after infection. However, GSI did not impact fungal clearance in mice with preexisting immunity. The dampened host defenses were associated with reduced differentiation and maturation of monocyte-derived DCs and elevatmonocyte-derived macrophage and alveolar macrophage polarization to M2. Our study reveals the critical nature of Notch signaling in maintaining control of this infectious agent.
    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
    新出现和重新出现的人类冠状病毒(hCoV)会导致人类严重的呼吸道疾病,但是这些疾病中致命性肺炎的基础还不清楚。肺泡巨噬细胞(AMs)是宿主抗病毒防御和组织耐受性在各种呼吸道感染的关键协调,AM功能障碍与严重COVID-19相关。在这项研究中,使用中东呼吸综合征冠状病毒(MERS-CoV)感染的小鼠模型,我们研究了AMs在MERS发病机制中的作用.我们的结果表明,使用氯膦酸盐(CL)脂质体消耗AMs可显着增加人二肽基肽酶4敲入(hDPP4-KI)小鼠的发病率和死亡率。在感染后不同天,对对照和AM耗尽的肺的详细检查显示,中性粒细胞活性增加,但在感染后期,AM缺乏的肺中MERS-CoV特异性CD4T细胞反应显着降低。此外,AM耗竭小鼠的MERS严重程度增强与肺部炎症和病变相关。总的来说,这些数据表明,AMs对于形成最佳病毒特异性T细胞应答和控制MERS-CoV感染期间的过度炎症至关重要.
    Emerging and re-emerging human coronaviruses (hCoVs) cause severe respiratory illness in humans, but the basis for lethal pneumonia in these diseases is not well understood. Alveolar macrophages (AMs) are key orchestrators of host antiviral defense and tissue tolerance during a variety of respiratory infections, and AM dysfunction is associated with severe COVID-19. In this study, using a mouse model of Middle East respiratory syndrome coronavirus (MERS-CoV) infection, we examined the role of AMs in MERS pathogenesis. Our results show that depletion of AMs using clodronate (CL) liposomes significantly increased morbidity and mortality in human dipeptidyl peptidase 4 knock-in (hDPP4-KI) mice. Detailed examination of control and AM-depleted lungs at different days postinfection revealed increased neutrophil activity but a significantly reduced MERS-CoV-specific CD4 T-cell response in AM-deficient lungs during later stages of infection. Furthermore, enhanced MERS severity in AM-depleted mice correlated with lung inflammation and lesions. Collectively, these data demonstrate that AMs are critical for the development of an optimal virus-specific T-cell response and controlling excessive inflammation during MERS-CoV 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
    尽管进行了广泛的分析,仍然迫切需要描述导致重症冠状病毒病2019(COVID-19)患者死亡的免疫细胞状态.这里,我们对重症COVID-19患者的血液和呼吸道样本进行了高维分析,以检查细胞连锁分子特征与死亡结局之间的关联.基于单细胞RNAseq的免疫状态去卷积的外周转录谱与COVID-19死亡率相关。Further,随着时间的推移,单核细胞中持续高水平的干扰素信号传导模块导致随后的炎性细胞因子的协同上调.SARS-CoV-2感染的下呼吸道骨髓细胞上调CXCL10,导致更高的死亡风险。我们的分析表明,在严重的COVID-19中,病毒感染的骨髓细胞和延长的干扰素信号传导具有关键作用。
    Despite extensive analyses, there remains an urgent need to delineate immune cell states that contribute to mortality in people critically ill with COVID-19. Here, we present high-dimensional profiling of blood and respiratory samples from people with severe COVID-19 to examine the association between cell-linked molecular features and mortality outcomes. Peripheral transcriptional profiles by single-cell RNA sequencing (RNA-seq)-based deconvolution of immune states are associated with COVID-19 mortality. Further, persistently high levels of an interferon signaling module in monocytes over time lead to subsequent concerted upregulation of inflammatory cytokines. SARS-CoV-2-infected myeloid cells in the lower respiratory tract upregulate CXCL10, leading to a higher risk of death. Our analysis suggests a pivotal role for viral-infected myeloid cells and protracted interferon signaling in severe COVID-19.
    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
    肿瘤进展基因座2(Tpl2)是一种丝氨酸-苏氨酸激酶,已知可促进各种病原体相关分子模式(PAMPs)的炎症反应。炎性细胞因子和G蛋白偶联受体,因此有助于宿主对病原体的抗性。我们最近表明,Tpl2-/-小鼠以未知的机制死于低致病性流感株(x31,H3N2)的感染。在这项研究中,我们试图表征流感感染的Tpl2-/-小鼠的细胞因子和免疫细胞谱,以深入了解其宿主保护作用.虽然Tpl2-/-小鼠表现出适度受损的病毒控制,在Tpl2-/-小鼠的肺中,在发病和死亡高峰当天未观察到病毒,提示发病不是由于病毒致细胞病变作用,而是由于过度活跃的抗病毒免疫应答.的确,干扰素-β(IFN-β)水平升高,IFN诱导的单核细胞趋化蛋白-1(MCP-1,CCL2),巨噬细胞炎性蛋白1α(MIP-1α;CCL3),MIP-1β(CCL4),RANTES(CCL5),在感染后7天(dpi),在流感感染的Tpl2-/-小鼠的肺中观察到IP-10(CXCL10)和干扰素-γ(IFN-γ)。升高的细胞因子和趋化因子伴随着炎性单核细胞和嗜中性粒细胞对肺的浸润增加。此外,我们注意到IFN-β的增加与肺中CCL2,CXCL1和一氧化氮合酶(NOS2)表达的增加相关,这与严重的流感感染有关。骨髓嵌合体与Tpl2消融定位于放射抗性细胞证实Tpl2功能,至少在某种程度上,在放射抗性细胞内限制对病毒感染的促炎反应。总的来说,这项研究表明,Tpl2通过限制干扰素和趋化因子的产生来缓和流感感染期间的炎症,已知干扰素和趋化因子可促进有害的炎性单核细胞和中性粒细胞的募集.
    Tumor progression locus 2 (Tpl2) is a serine-threonine kinase known to promote inflammation in response to various pathogen-associated molecular patterns (PAMPs), inflammatory cytokines and G-protein-coupled receptors and consequently aids in host resistance to pathogens. We have recently shown that Tpl2-/- mice succumb to infection with a low-pathogenicity strain of influenza (x31, H3N2) by an unknown mechanism. In this study, we sought to characterize the cytokine and immune cell profile of influenza-infected Tpl2-/- mice to gain insight into its host protective effects. Although Tpl2-/- mice display modestly impaired viral control, no virus was observed in the lungs of Tpl2-/- mice on the day of peak morbidity and mortality suggesting that morbidity is not due to virus cytopathic effects but rather to an overactive antiviral immune response. Indeed, increased levels of interferon-β (IFN-β), the IFN-inducible monocyte chemoattractant protein-1 (MCP-1, CCL2), Macrophage inflammatory protein 1 alpha (MIP-1α; CCL3), MIP-1β (CCL4), RANTES (CCL5), IP-10 (CXCL10) and Interferon-γ (IFN-γ) was observed in the lungs of influenza-infected Tpl2-/- mice at 7 days post infection (dpi). Elevated cytokine and chemokines were accompanied by increased infiltration of the lungs with inflammatory monocytes and neutrophils. Additionally, we noted that increased IFN-β correlated with increased CCL2, CXCL1 and nitric oxide synthase (NOS2) expression in the lungs, which has been associated with severe influenza infections. Bone marrow chimeras with Tpl2 ablation localized to radioresistant cells confirmed that Tpl2 functions, at least in part, within radioresistant cells to limit pro-inflammatory response to viral infection. Collectively, this study suggests that Tpl2 tempers inflammation during influenza infection by constraining the production of interferons and chemokines which are known to promote the recruitment of detrimental inflammatory monocytes and neutrophils.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号