Cytokine storm

细胞因子风暴
  • 文章类型: Journal Article
    SARS-CoV-2感染可在某些患者中引发细胞因子风暴,其特征在于细胞因子和化学介质的过量产生。这种过度活跃的免疫应答可能导致显著的组织损伤和多器官衰竭(MOF)。COVID-19的严重程度与细胞因子风暴的强度相关,涉及IFN等元素,NF-κB,IL-6、HMGB1等。必须迅速进行适应性免疫以有效控制疾病进展。CD4+T细胞通过改善B细胞产生中和抗体和激活CD8+T细胞来促进免疫反应。有助于根除病毒感染的细胞。同时,来自B细胞的抗体可以中和病毒,阻碍宿主细胞的进一步感染。在从疾病中康复的个体中,观察到病毒特异性抗体和记忆T细胞,可以提供一定程度的保护,降低再感染的可能性或减轻严重程度。本文讨论了巨噬细胞的作用,IFN,细胞因子释放综合征(CRS)中的IL-6和HMGB1,适应性免疫的复杂性,免疫记忆的持久性,所有这些对于COVID-19的预防和治疗策略都至关重要。
    SARS-CoV-2 infection can trigger cytokine storm in some patients, which characterized by an excessive production of cytokines and chemical mediators. This hyperactive immune response may cause significant tissue damage and multiple organ failure (MOF). The severity of COVID-19 correlates with the intensity of cytokine storm, involving elements such as IFN, NF-κB, IL-6, HMGB1, etc. It is imperative to rapidly engage adaptive immunity to effectively control the disease progression. CD4+ T cells facilitate an immune response by improving B cells in the production of neutralizing antibodies and activating CD8+ T cells, which are instrumental in eradicating virus-infected cells. Meanwhile, antibodies from B cells can neutralize virus, obstructing further infection of host cells. In individuals who have recovered from the disease, virus-specific antibodies and memory T cells were observed, which could confer a level of protection, reducing the likelihood of re-infection or attenuating severity. This paper discussed the roles of macrophages, IFN, IL-6 and HMGB1 in cytokine release syndrome (CRS), the intricacies of adaptive immunity, and the persistence of immune memory, all of which are critical for the prevention and therapeutic strategies against 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
    布鲁氏菌感染的综合免疫景观对于开发布鲁氏菌病的新疗法至关重要。这里,我们利用来自35个个体的290,369个细胞的单细胞RNA测序(scRNA-seq),包括29例急性布鲁氏菌病患者(n=10),亚急性(n=9),和慢性(n=10)阶段以及六个健康供体。将酶联免疫吸附测定应用于该群组内的验证。布鲁氏菌感染引起外周免疫细胞组成的显著改变,炎症是布鲁氏菌病的关键特征。急性患者的特征在于由S100A8/A9的全身上调引起的潜在细胞因子风暴,主要是由于经典的单核细胞。细胞因子风暴可能通过激活S100A8/A9-TLR4-MyD88信号通路介导。此外,单核细胞髓源性抑制细胞是急性患者免疫麻痹的可能原因.慢性患者的特征是Th1反应失调,以IFN-γ和Th1特征的表达减少以及高耗尽状态为标志。此外,布鲁氏菌感染可以抑制骨髓细胞凋亡(例如,mDC,经典单核细胞),抑制专职抗原呈递细胞中的抗原呈递(APC;例如,mDC)和非专业APC(例如,单核细胞),并诱导CD8+T/NK细胞的耗竭,可能导致慢性感染的建立。总的来说,我们的研究系统地破译了布鲁氏菌在感染不同阶段的协调免疫反应,这有助于全面了解布鲁氏菌病的免疫发病机制,并可能有助于开发新的有效治疗策略,尤其是那些有慢性感染的人。
    A comprehensive immune landscape for Brucella infection is crucial for developing new treatments for brucellosis. Here, we utilized single-cell RNA sequencing (scRNA-seq) of 290,369 cells from 35 individuals, including 29 brucellosis patients from acute (n = 10), sub-acute (n = 9), and chronic (n = 10) phases as well as six healthy donors. Enzyme-linked immunosorbent assays were applied for validation within this cohort. Brucella infection caused a significant change in the composition of peripheral immune cells and inflammation was a key feature of brucellosis. Acute patients are characterized by potential cytokine storms resulting from systemic upregulation of S100A8/A9, primarily due to classical monocytes. Cytokine storm may be mediated by activating S100A8/A9-TLR4-MyD88 signaling pathway. Moreover, monocytic myeloid-derived suppressor cells were the probable contributors to immune paralysis in acute patients. Chronic patients are characterized by a dysregulated Th1 response, marked by reduced expression of IFN-γ and Th1 signatures as well as a high exhausted state. Additionally, Brucella infection can suppress apoptosis in myeloid cells (e.g., mDCs, classical monocytes), inhibit antigen presentation in professional antigen-presenting cells (APCs; e.g., mDC) and nonprofessional APCs (e.g., monocytes), and induce exhaustion in CD8+ T/NK cells, potentially resulting in the establishment of chronic infection. Overall, our study systemically deciphered the coordinated immune responses of Brucella at different phases of the infection, which facilitated a full understanding of the immunopathogenesis of brucellosis and may aid the development of new effective therapeutic strategies, especially for those with chronic infection.
    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
    大鼠有丝分裂后间隔(SEP)神经元,被设计为在33℃有条件地增殖,当在37.5℃下被捕时可以分化,并且可以维持数周而没有细胞毒性作用。制备9个独立的cDNA文库以跟踪正常(N1)未感染和CJ剂感染的SEP细胞中的阻滞诱导的神经分化和先天性免疫应答。增殖的Nl与潜伏感染的(CJ-)细胞显示很少的RNA-seq差异。然而,逮捕引发了重大变化。正常细胞显示过多的抗增殖转录物。此外,已知的神经元分化转录本,例如,Agtr2、神经调节蛋白-1、GDF6、SFRP4和Prnp上调。这些Nl神经元还展示了许多激活的IFN先天免疫基因,例如,OAS1,RTP4,ISG20,GTB4,CD80和细胞因子,补语,和与错误折叠的蛋白质结合的簇蛋白(CLU)。相比之下,被捕的高传染性CJ细胞(10log/gm)下调了许多复制控制。此外,被捕的CJ+细胞抑制了神经元分化转录本,包括对CJ病原体感染至关重要的Prnp。CJ+细胞也增强了IFN刺激途径,对342个CJ+独特转录本的分析揭示了额外的先天免疫和抗病毒相关转录本,例如,Il17、ISG15和RSAD2(viperin)。这些数据显示:1)在分化期间由正常神经元产生先天免疫转录物;2)CJ感染可以增强和扩大抗病毒应答;3)潜伏CJ感染表观遗传印记许多增殖途径以阻止完全停滞。CJ+脑小胶质细胞,具有共享转录本的白细胞和肠道骨髓细胞可能被刺激以产生潜伏的CJD感染,这种感染在临床上可以沉默超过30年。
    Rat post-mitotic septal (SEP) neurons, engineered to conditionally proliferate at 33°C, differentiate when arrested at 37.5°C and can be maintained for weeks without cytotoxic effects. Nine independent cDNA libraries were made to follow arrest-induced neural differentiation and innate immune responses in normal (Nl) uninfected and CJ agent infected SEP cells. Proliferating Nl versus latently infected (CJ-) cells showed few RNA-seq differences. However arrest induced major changes. Normal cells displayed a plethora of anti-proliferative transcripts. Additionally, known neuron differentiation transcripts, e.g., Agtr2, Neuregulin-1, GDF6, SFRP4 and Prnp were upregulated. These Nl neurons also displayed many activated IFN innate immune genes, e.g., OAS1, RTP4, ISG20, GTB4, CD80 and cytokines, complement, and clusterin (CLU) that binds to misfolded proteins. In contrast, arrested highly infectious CJ+ cells (10 logs/gm) downregulated many replication controls. Furthermore, arrested CJ+ cells suppressed neuronal differentiation transcripts, including Prnp which is essential for CJ agent infection. CJ+ cells also enhanced IFN stimulated pathways, and analysis of the 342 CJ+ unique transcripts revealed additional innate immune and anti-viral-linked transcripts, e.g., Il17, ISG15, and RSAD2 (viperin). These data show: 1) innate immune transcripts are produced by normal neurons during differentiation; 2) CJ infection can enhance and expand anti-viral responses; 3) latent CJ infection epigenetically imprints many proliferative pathways to thwart complete arrest. CJ+ brain microglia, white blood cells and intestinal myeloid cells with shared transcripts may be stimulated to educe latent CJD infections that can be clinically silent for >30 years.
    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
    严重的COVID-19病例通常进展为危及生命的疾病,如急性呼吸窘迫综合征(ARDS),脓毒症,多器官功能障碍综合征(MODS)。Gelsolin(GSN),一种具有抗炎和免疫调节特性的肌动蛋白结合蛋白,是严重COVID-19的有希望的治疗靶标。危重病患者血浆GSN水平显著下降,包括COVID-19,与免疫反应失调和不良预后相关。补充GSN可以减轻急性肺损伤,ARDS,和败血症,通过清除肌动蛋白,与严重的COVID-19具有共同的病理生理特征,调节细胞因子的产生,增强巨噬细胞吞噬,并稳定肺泡毛细血管屏障。初步数据表明,重组人血浆GSN可改善重症COVID-19ARDS患者的氧合和肺功能。尽管需要进一步的研究来优化GSN治疗,目前的证据支持其减轻COVID-19严重后果并改善患者预后的潜力。这篇综述提供了一个全面的生物学特性分析,机制,GSN在重症COVID-19中的治疗价值。
    Severe COVID-19 cases often progress to life-threatening conditions such as acute respiratory distress syndrome (ARDS), sepsis, and multiple organ dysfunction syndrome (MODS). Gelsolin (GSN), an actin-binding protein with anti-inflammatory and immunomodulatory properties, is a promising therapeutic target for severe COVID-19. Plasma GSN levels are significantly decreased in critical illnesses, including COVID-19, correlating with dysregulated immune responses and poor outcomes. GSN supplementation may mitigate acute lung injury, ARDS, and sepsis, which share pathophysiological features with severe COVID-19, by scavenging actin, modulating cytokine production, enhancing macrophage phagocytosis, and stabilizing the alveolar-capillary barrier. Preliminary data indicate that recombinant human plasma GSN improves oxygenation and lung function in severe COVID-19 patients with ARDS. Although further research is needed to optimize GSN therapy, current evidence supports its potential to mitigate severe consequences of COVID-19 and improve patient outcomes. This review provides a comprehensive analysis of the biological characteristics, mechanisms, and therapeutic value of GSN 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
    噬血细胞淋巴组织细胞增生症(HLH)包括广泛的威胁生命的细胞因子风暴综合征,分为原发性(遗传)或继发性(获得性)HLH。后者发生在各种医疗条件下,包括感染,恶性肿瘤,自身免疫性和自身炎症性疾病,获得性免疫缺陷,和代谢紊乱。尽管该领域最近取得了进展,继发性HLH的发病机制尚不完全清楚。考虑到继发性HLH的触发因素和基础疾病的异质性,已经开发了大量的动物模型来探索关键的疾病机制。迄今为止,已经描述了超过20个动物模型,每个都概括了继发性HLH的某些方面。这篇综述全面概述了现有的模型,强调相关发现,讨论不同细胞类型和细胞因子在疾病发展和进展中的参与,并考虑未来治疗策略的兴趣点。
    Hemophagocytic lymphohistiocytosis (HLH) comprises a broad spectrum of life-threatening cytokine storm syndromes, classified into primary (genetic) or secondary (acquired) HLH. The latter occurs in a variety of medical conditions, including infections, malignancies, autoimmune and autoinflammatory diseases, acquired immunodeficiency, and metabolic disorders. Despite recent advances in the field, the pathogenesis of secondary HLH remains incompletely understood. Considering the heterogeneity of triggering factors and underlying diseases in secondary HLH, a large diversity of animal models has been developed to explore pivotal disease mechanisms. To date, over 20 animal models have been described that each recapitulates certain aspects of secondary HLH. This review provides a comprehensive overview of the existing models, highlighting relevant findings, discussing the involvement of different cell types and cytokines in disease development and progression, and considering points of interest toward future therapeutic strategies.
    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
    HIV感染引发炎症反应,在大多数HIV感染的个体中表现为急性逆转录病毒综合征(ARS)。虽然这种综合征通常是自我限制的,原发性HIV感染有时会引发与细胞因子风暴综合征(CSS)一致的暴发性炎症过程。CSS的许多关键发现,包括发烧,脾肿大,在ARS中经常观察到血细胞减少,提示CSS在急性HIV感染的背景下可能未得到认可。与其他CSS场景不同,ARS相关的CSS通常对HIV靶向治疗反应良好。晚期HIV感染也与CSS有关,尽管这通常涉及额外的感染性侮辱。偶尔,HIV治疗导致进化为CSS的免疫应答的快速恢复。
    HIV infection triggers an inflammatory response that manifests as acute retroviral syndrome (ARS) in most individuals infected by HIV. While this syndrome is usually self-limited, primary HIV infection sometimes triggers a fulminant inflammatory process consistent with cytokine storm syndrome (CSS). Many of the key findings of CSS including fever, splenomegaly, and cytopenias are routinely observed in ARS, suggesting CSS may be under recognized in the setting of acute HIV infection. Unlike other CSS scenarios, ARS-associated CSS generally responds well to HIV-targeted therapies. Advanced HIV infection is also associated with CSS, although typically this involves additional infectious insults. Occasionally, HIV therapy results in rapid recovery of the immune response that evolves into CSS.
    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
    白细胞介素-6(IL-6),高炎性免疫反应的生物标志物,可用于确定需要重症监护的多器官受累患者2019年冠状病毒病(COVID-19)的严重程度。我们研究的目的是了解血液透析的效用,不仅在减轻肾脏负担方面,而且还可以通过解决COVID细胞因子风暴综合征来改善结果。
    在这个前景中,观察性研究,入住COVID重症监护病房(ICU)病房的126例患者因急性肾损伤(AKI)接受了血液透析治疗。评估患者的常规基线血液参数。在所有患者的透析前和存活患者的出院当天测量IL-6。
    在总共126名患者中,79人是幸存者,47人是非幸存者。在非幸存者中,大多数年龄较大(P=0.009)。两组的男性比例均较高(幸存者和非幸存者分别为78.72%和55.69%,分别)。与幸存者相比,非幸存者的平均中性粒细胞淋巴细胞比率(NLR)和D-二聚体水平显着升高(P<0.001)。平均血清尿素,肌酐,非存活者中IL-6水平显著升高(P<0.001)。幸存者接受的平均血液透析次数较高。存活者的deltaIL-6和delta血清肌酐曲线呈显著正相关(r=0.819,P<0.001)。
    我们的研究将IL-6确定为COVIDICUAKI患者的不良预后预测因子。它还强调使用血液透析作为一种具有成本效益的挽救生命的治疗介入方式,不仅可以改善肾脏预后,而且还通过降低IL-6水平来抑制细胞因子风暴。
    UNASSIGNED: Interleukin-6 (IL-6), a biomarker of hyperinflammatory immune response, can be used to determine the severity of coronavirus disease 2019 (COVID-19) in patients with multi-organ involvement requiring critical care. The aim of our study is to understand the utility of hemodialysis, not only in terms of reducing renal burden, but also improving the outcome by tackling the COVID cytokine storm syndrome.
    UNASSIGNED: In this prospective, observational study, 126 patients admitted to the COVID intensive care unit (ICU) wards were treated with hemodialysis for acute kidney injury (AKI). Patients\' routine baseline blood parameters were evaluated. IL-6 was measured predialysis in all patients and on the day of discharge in the patients who survived.
    UNASSIGNED: Out of total 126 patients, 79 were survivors and 47 were nonsurvivors. Among nonsurvivors, majority were older (P = 0.009). Both the groups had a higher percentage of males (78.72% and 55.69% in survivors and nonsurvivors, respectively). Mean neutrophil lymphocyte ratio (NLR) and D-dimer level were significantly higher in nonsurvivors compared to survivors (P < 0.001). Mean serum urea, creatinine, and IL-6 levels were significantly greater in nonsurvivors (P < 0.001). Mean number of hemodialysis sessions received by survivors was higher. The curve between delta IL-6 and delta serum creatinine for survivors showed a significant positive association (r = 0.819, P < 0.001).
    UNASSIGNED: Our study establishes IL-6 as a poor outcome predictor in COVID ICU patients with AKI. It also emphasizes the use of hemodialysis as a cost-effective lifesaving therapeutic interventional modality to not only improve the renal outcome, but also curb the cytokine storm by reducing IL-6 levels.
    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
    实时监测汗液中的低浓度细胞因子如TNF-α可以帮助临床医生评估炎症的严重程度。与收集和杂质的存在相关的挑战可以显著阻碍汗液中蛋白质的检测。这个问题是通过结合纳米球阵列设计的自动汗液运输,再加上一个可重复使用的传感器,采用Nafion/适体修改的MoS2场效应晶体管。具有逐步润湿性的纳米球阵列能够自动收集汗液并阻止杂质污染检测区。该装置能够直接检测未稀释汗液中的TNF-α蛋白,在10fM至1nM的检测范围内。超薄的使用,超柔性基板确保稳定的电气性能,甚至在30次极端变形之后。研究结果表明,在临床情况下,该设备可能通过汗液检测提供对患者免疫状态的实时评估和管理。
    The real-time monitoring of low-concentration cytokines such as TNF-α in sweat can aid clinical physicians in assessing the severity of inflammation. The challenges associated with the collection and the presence of impurities can significantly impede the detection of proteins in sweat. This issue is addressed by incorporating a nanosphere array designed for automatic sweat transportation, coupled with a reusable sensor that employs a Nafion/aptamer-modified MoS2 field-effect transistor. The nanosphere array with stepwise wettability enables automatic collection of sweat and blocks impurities from contaminating the detection zone. This device enables direct detection of TNF-α proteins in undiluted sweat, within a detection range of 10 fM to 1 nM. The use of an ultrathin, ultraflexible substrate ensures stable electrical performance, even after up to 30 extreme deformations. The findings indicate that in clinical scenarios, this device could potentially provide real-time evaluation and management of patients\' immune status via sweat testing.
    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
    感染相关的脂多糖(LPS)释放引起细胞因子风暴和急性肺损伤。新的数据表明,白介素6(IL-6)抑制剂托珠单抗可以改善脓毒症患者的肺损伤。本研究旨在探讨托珠单抗对肝硬化大鼠急性肺损伤的治疗作用。在胆总管结扎(BDL)的Sprague-Dawley大鼠中诱发胆汁性肝硬化。假手术大鼠作为手术对照。在术后第21天施用Tocilizumab,并且在第29天腹膜内注射LPS。注射LPS后三小时,血液动力学参数,生物化学数据,和动脉血气分析进行了评估,以及IL-6和肿瘤坏死因子-α(TNF-α)的测量。检查肝和肺组织学,和蛋白质水平进行了分析。LPS给药降低门静脉压力,BDL大鼠的门静脉流量和心脏指数。此外,LPS诱导急性肺损伤,缺氧和升高的TNF-α和IL-6水平。托珠单抗治疗前不影响血液动力学和生物化学数据,但它改善了肺损伤,降低了TNF-α,IL-6和CD68阳性巨噬细胞浸润。此外,托珠单抗给药改善了BDL大鼠的缺氧和气体交换,并下调肝脏和肺部炎症蛋白的表达。总之,LPS诱导胆汁性肝硬化大鼠急性肺损伤。托珠单抗预处理可减少肺损伤和缺氧,可能通过下调炎症蛋白和减少IL-6,TNF-α和CD68阳性巨噬细胞在肺中的募集。
    Infection-related lipopolysaccharide (LPS) release causes cytokine storm and acute lung injury. Emerging data show that the interleukin 6 (IL-6) inhibitor tocilizumab can improve lung damage in patients with sepsis. This study aimed to investigate the therapeutic effect of tocilizumab on acute lung injury in cirrhotic rats. Biliary cirrhosis was induced in Sprague-Dawley rats with common bile duct ligation (BDL). Sham-operated rats served as surgical controls. Tocilizumab was administered on post-operative day 21, and LPS was injected intraperitoneally on day 29. Three hours after LPS injection, hemodynamic parameters, biochemistry data, and arterial blood gas analysis were evaluated, along with measurements of IL-6 and tumor necrosis factor-α (TNF-α). Liver and lung histology was examined, and protein levels were analyzed. LPS administration reduced portal pressure, portal venous flow and cardiac index in the BDL rats. In addition, LPS administration induced acute lung injury, hypoxia and elevated TNF-α and IL-6 levels. Pre-treatment with tocilizumab did not affect hemodynamic and biochemistry data, but it ameliorated lung injury and decreased TNF-α, IL-6, and CD68-positive macrophage infiltration. Moreover, tocilizumab administration improved hypoxia and gas exchange in the BDL rats, and downregulated hepatic and pulmonary inflammatory protein expression. In conclusion, LPS administration induced acute lung injury in biliary cirrhotic rats. Pre-treatment with tocilizumab reduces lung damage and hypoxia, possibly by downregulating inflammatory proteins and reducing IL-6, TNF-α and CD68-positive macrophage recruitment in the lung.
    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
    2019年末出现了严重急性呼吸系统综合症冠状病毒2(SARS-CoV-2),COVID-19的致病因素对全球社会经济结构造成了深刻和前所未有的破坏,对全世界数百万人的生活产生负面影响。严重COVID-19的典型标志是由于先天免疫细胞的异常细胞因子释放(细胞因子风暴)引起的过度炎症。最近的研究表明,SARS-CoV-2通过其尖峰(S)蛋白,可以通过Toll样受体(TLRs)激活身体的先天免疫细胞,特别是TLR4。计算机模拟研究表明,S蛋白以高亲和力与TLR4结合,触发下游信号传导过程,导致促炎细胞因子释放。与其他TLR相比,如TLR2,TLR4在启动和维持与严重COVID-19相关的炎症反应中起着更重要的作用。此外,病毒与靶细胞之间的相互作用可以增强TLR4的细胞表达,使细胞对病毒相互作用和随后的炎症更敏感。这种增加的TLR4的表达在病毒进入创建一个反馈回路,TLR4水平升高导致炎症反应放大,导致疾病的严重程度。此外,TLR4对炎症途径的有效激活使其与其他TLRs区分开来,强调其在COVID-19发病机制中的关键作用。在这次审查中,我们彻底探索了SARS-CoV-2用于诱发炎症的多种调节信号通路.与其他TLRs相比,我们特别关注TLR4激活的关键影响,强调TLR4与病毒S蛋白的相互作用如何加剧COVID-19的严重程度。通过深入研究TLR4介导的炎症机制,我们的目标是阐明可能减轻严重COVID-19引起的炎症损伤的潜在治疗靶点。了解TLR4在SARS-CoV-2感染背景下的独特作用可以为特异性抑制该受体活性的新型治疗策略铺平道路。从而降低总体疾病负担并改善患者预后。
    The late 2019 emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, caused profound and unprecedented disruption to the global socio-economic structure, negatively affecting millions of lives worldwide. A typical hallmark of severe COVID-19 is hyper inflammation due to aberrant cytokine release (cytokine storm) by innate immune cells. Recent studies have revealed that SARS-CoV-2, through its spike (S) protein, can activate the body\'s innate immune cells via Toll-Like Receptors (TLRs), particularly TLR4. In silico studies have demonstrated that the S protein binds with high affinity to TLR4, triggering downstream signaling processes that result in pro-inflammatory cytokine release. Compared to other TLRs, such as TLR2, TLR4 plays a more significant role in initiating and sustaining the inflammatory response associated with severe COVID-19. Furthermore, interactions between the virus and target cells can enhance the cellular expression of TLR4, making cells more susceptible to viral interactions and subsequent inflammation. This increased expression of TLR4 upon viral entry creates a feedback loop, where heightened TLR4 levels lead to amplified inflammatory responses, contributing to the severity of the disease. Additionally, TLR4\'s potent activation of inflammatory pathways sets it apart from other TLRs, underscoring its pivotal role in the pathogenesis of COVID-19. In this review, we thoroughly explore the multitude of regulatory signaling pathways that SARS-CoV-2 employs to incite inflammation. We specifically focus on the critical impact of TLR4 activation compared to other TLRs, highlighting how TLR4\'s interactions with the viral S protein can exacerbate the severity of COVID-19. By delving into the mechanisms of TLR4-mediated inflammation, we aim to shed light on potential therapeutic targets that could mitigate the inflammatory damage caused by severe COVID-19. Understanding the unique role of TLR4 in the context of SARS-CoV-2 infection could pave the way for novel treatment strategies that specifically inhibit this receptor\'s activity, thereby reducing the overall disease burden and improving patient outcomes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号