Cytokine storm

细胞因子风暴
  • 文章类型: Journal Article
    噬血细胞性淋巴组织细胞增生症(HLH)是一种常见且高度致命的高炎症综合征,其特征是巨噬细胞的异常激活。迄今为止,缺乏针对HLH的靶向治疗。在先前的研究中,验证了HLH中的巨噬细胞有效吞噬来自免疫性血小板减少症(ITP)患者的抗CD41血小板(抗CD41-PLT)。因此,模拟ITP的病理机制,并利用抗CD41-PLT负载巨噬细胞毒性药物VP16,构建巨噬细胞可靶向的工程血小板抗CD41-PLT-VP16,这是一种针对HLH的新型靶向疗法。体外和体内研究均表明抗CD41-PLT-VP16具有优异的靶向和促巨噬细胞凋亡作用。在HLH模型小鼠中,抗CD41-PLT-VP16可预防吞噬作用并抑制细胞因子风暴。机制研究表明,抗CD41-PLT-VP16增加VP16的细胞毒性,促进对巨噬细胞的精确干预。此外,它作为减少过度炎症综合征的战略“守望者”,能间接防止T细胞和NK细胞的异常活化,降低Ab依赖性细胞介导的细胞毒性效应。第一个基于血小板的临床试验正在进行中。结果表明,与接受常规化疗的患者相比,用抗CD41-PLT-VP16治疗后,HLH患者的总体反应率增加了三倍。总之,抗CD41-PLT-VP16提供了对高炎症综合征的总体认识,并为HLH提供了一种新的临床治疗策略.
    Hemophagocytic lymphohistiocytosis (HLH) is a common and highly fatal hyperinflammatory syndrome characterized by the aberrant activation of macrophages. To date, there is a lack of targeted therapies for HLH. It is validated that macrophages in HLH efficiently phagocytose anti-CD41-platelets (anti-CD41-PLTs) from immune thrombocytopenia (ITP) patients in previous research. Hence, the pathological mechanisms of ITP are mimicked and anti-CD41-PLTs are utilized to load the macrophage-toxic drug VP16 to construct macrophage-targetable engineered platelets anti-CD41-PLT-VP16, which is a novel targeted therapy against HLH. Both in vitro and in vivo studies demonstrate that anti-CD41-PLT-VP16 has excellent targeting and pro-macrophage apoptotic effects. In HLH model mice, anti-CD41-PLT-VP16 prevents hemophagocytosis and inhibits the cytokine storm. Mechanistic studies reveal that anti-CD41-PLT-VP16 increases the cytotoxicity of VP16, facilitating precise intervention in macrophages. Furthermore, it operates as a strategic \"besieger\" in diminishing hyperinflammation syndrome, which can indirectly prevent the abnormal activation of T cells and NK cells and reduce the Ab-dependent cell-mediated cytotoxicity effect. The first platelet-based clinical trial is ongoing. The results show that after treatment with anti-CD41-PLT-VP16, HLH patients have a threefold increase in the overall response rate compared to patients receiving conventional chemotherapy. In conclusion, anti-CD41-PLT-VP16 provides a general insight into hyperinflammation syndrome and offers a novel clinical therapeutic strategy for HLH.
    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
    细胞因子风暴(CS)是由病原体和过度免疫疗法等各种因素引发的加剧的炎症反应。如果不加以控制,对生命构成重大威胁。槲皮素,在中药中发现的单体,表现出显着的抗炎和抗病毒特性。本研究试图通过网络药理学分析和实验验证相结合来探讨槲皮素干预是否可以减轻CS。首先,通过网络药理学鉴定了槲皮素和CS影响的常见靶基因和潜在机制,和分子对接实验证实槲皮素和核心靶标。随后,脂多糖(LPS)刺激Raw264.7细胞的体外实验表明,槲皮素能有效抑制促炎介质的过表达,调节AKT1-FoxO1信号通路。同时,槲皮素可通过Keap1-Nrf2信号通路减少活性氧。此外,注射LPS的C57BL/6小鼠的体内研究进一步证实了槲皮素对CS的抑制作用。总之,这项研究阐明了与槲皮素对CS的治疗作用有关的新的靶基因和信号通路。此外,它提供了令人信服的证据支持槲皮素逆转LPS诱导的CS的功效,主要通过调节AKT1-FoxO1和Keap1-Nrf2信号通路。
    Cytokine storm (CS) emerges as an exacerbated inflammatory response triggered by various factors such as pathogens and excessive immunotherapy, posing a significant threat to life if left unchecked. Quercetin, a monomer found in traditional Chinese medicine, exhibits notable anti-inflammatory and antiviral properties. This study endeavors to explore whether quercetin intervention could mitigate CS through a combination of network pharmacology analysis and experimental validation. First, common target genes and potential mechanisms affected by quercetin and CS were identified through network pharmacology, and molecular docking experiments confirmed quercetin and core targets. Subsequently, in vitro experiments of Raw264.7 cells stimulated by lipopolysaccharide (LPS) showed that quercetin could effectively inhibit the overexpression of pro-inflammatory mediators and regulate the AKT1-FoxO1 signaling pathway. At the same time, quercetin can reduce ROS through the Keap1-Nrf2 signaling pathway. In addition, in vivo studies of C57BL/6 mice injected with LPS further confirmed quercetin\'s inhibitory effect on CS. In conclusion, this investigation elucidated novel target genes and signaling pathways implicated in the therapeutic effects of quercetin on CS. Moreover, it provided compelling evidence supporting the efficacy of quercetin in reversing LPS-induced CS, primarily through the regulation of the AKT1-FoxO1 and Keap1-Nrf2 signaling pathways.
    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
    自身免疫性风湿性疾病包括一组以非器官特异性炎症为特征的免疫相关病症。这些疾病包括系统性红斑狼疮(SLE),类风湿性关节炎(RA),强直性脊柱炎(AS),痛风,在其他人中。通常涉及血液系统,这些疾病也可能影响多个器官和系统。自身免疫性风湿免疫性疾病的发病机制复杂,不同的病因,都与免疫功能障碍有关。目前这种类型疾病的治疗选择相对有限,并伴有某些副作用。因此,目前的紧迫挑战仍然是确定这些疾病的新治疗靶点。甾醇调节元件结合蛋白(SREBP)是基本的螺旋-环-螺旋-亮氨酸拉链转录因子,可调节脂质和胆固醇生物合成相关基因的表达。SREBPs的表达和转录活性可以通过细胞外刺激如多不饱和脂肪酸来调节,氨基酸,葡萄糖,和能量途径,包括AKT-mTORC和AMP激活的蛋白激酶(AMPK)。研究表明,SREBPs在调节脂质代谢、细胞因子产生,炎症,和生发中心B(GCB)细胞的增殖。这些功能在风湿病和免疫性疾病的发病机理中具有重要意义(图形摘要)。因此,本文综述了SREBPs在SLE发病中的潜在机制,RA,和痛风,基于对其功能的探索。
    Autoimmune rheumatic diseases comprise a group of immune-related disorders characterized by non-organ-specific inflammation. These diseases include systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), ankylosing spondylitis (AS), gout, among others. Typically involving the hematologic system, these diseases may also affect multiple organs and systems. The pathogenesis of autoimmune rheumatic immune diseases is complex, with diverse etiologies, all associated with immune dysfunction. The current treatment options for this type of disease are relatively limited and come with certain side effects. Therefore, the urgent challenge remains to identify novel therapeutic targets for these diseases. Sterol regulatory element-binding proteins (SREBPs) are basic helix-loop-helix-leucine zipper transcription factors that regulate the expression of genes involved in lipid and cholesterol biosynthesis. The expression and transcriptional activity of SREBPs can be modulated by extracellular stimuli such as polyunsaturated fatty acids, amino acids, glucose, and energy pathways including AKT-mTORC and AMP-activated protein kinase (AMPK). Studies have shown that SREBPs play roles in regulating lipid metabolism, cytokine production, inflammation, and the proliferation of germinal center B (GCB) cells. These functions are significant in the pathogenesis of rheumatic and immune diseases (Graphical abstract). Therefore, this paper reviews the potential mechanisms of SREBPs in the development of SLE, RA, and gout, based on an exploration of their functions.
    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
    巨噬细胞的持续激活在细胞因子风暴(CS)的发病机理中起着至关重要的作用。考虑到CS是多种细胞因子参与的结果,单一细胞因子或其受体靶向阻断治疗的疗效尚不确定.蜂毒素,通过抑制活化巨噬细胞的丝裂原活化蛋白激酶和核因子κB途径,系统地抑制促炎介质的过度表达,在缓解CS和急性炎症损伤(AII)方面显示出巨大的潜力。然而,其临床应用受到其溶血活性的限制,非特异性细胞毒性和缺乏靶向性。在这项研究中,开发了具有核壳结构的叶酸修饰和蜂毒素稳定负载的固体脂质纳米颗粒(Fa-MpG@LNP),用于CS控制,通过靶向抑制活化巨噬细胞中促炎介质的过度产生,并具有叶酸受体-β的特异性表达。所得Fa-MpG@LNP具有理想的理化性质和稳定性,低溶血活性和非特异性细胞毒性,它可以特异性结合脂多糖(LPS)刺激的巨噬细胞,并有效降低促炎介质的升高水平。静脉给药后,Fa-MpG@LNP在发炎组织中积累,并显着下调组织浸润的巨噬细胞中促炎细胞因子的过度产生,导致LPS诱导的急性肺炎小鼠炎症组织和血清中细胞因子浓度显着降低,最终缓解AII,并产生无法检测到的毒副作用。这些结果表明Fa-MpG@LNP在缓解CS及其相关症状方面的临床应用潜力。
    The continuous activation of macrophages play a critical role in the pathogenesis of cytokine storm (CS). Considering that CS results from the participation of multiple cytokines, the therapeutic effect of a single cytokine or its receptor-targeted blockade therapy remains uncertain. Melittin, which can systematically suppress the overexpression of proinflammatory mediators via inhibiting the mitogen-activated protein kinase and nuclear factor kappa-B pathways in activated macrophages, shows great potential in alleviating CS and acute inflammatory injury (AII). However, its clinical application is limited by its hemolytic activity, non-specific cytotoxicity and lack of targeting. In this study, a folic acid-modified and melittin stable-loaded solid lipid nanoparticle (Fa-MpG@LNP) with a core-shell structure was developed for CS control via targeted inhibition of the overproduction of proinflammatory mediators in activated macrophages with specific expression of folate receptor-β. The resultant Fa-MpG@LNP showed ideal physicochemical properties and stability, low hemolytic activity and non-specific cytotoxicity, and it can specifically bind to lipopolysaccharide (LPS)-stimulated macrophages and effectively reduce the elevated levels of proinflammatory mediators. After intravenous administration, the Fa-MpG@LNP accumulated at inflamed tissue and significantly downregulate the overproduction of proinflammatory cytokines in tissue-infiltrated macrophages, resulting in a significant decrease of cytokine concentration in inflamed tissue and serum in LPS-induced acute pneumonia mice, and finally alleviate AII with undetectable toxic side effects. These results indicate the clinical application potential of Fa-MpG@LNP in alleviating CS and its related symptoms.
    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
    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
    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
    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
    脓毒症是由宿主对感染的反应失调引起的危及生命的器官功能障碍。炎性细胞因子风暴导致全身器官损伤,尤其是脓毒症患者的急性肺损伤。在这项研究中,我们发现S期激酶相关蛋白2(Skp2)的表达在脓毒症诱导的急性肺损伤(ALI)中显著降低.脓毒症激活MEK/ERK通路,抑制肺上皮Skp2表达,导致溶质载体家族3成员2(SLC3A2)的K48泛素化减少,从而损害其膜定位和胱氨酸/谷氨酸交换功能。因此,细胞内氧化还原反应失调诱导肺上皮细胞铁凋亡,导致肺损伤。最后,我们证明,Skp2mRNA封装的脂质纳米粒(LNPs)的静脉给药抑制了脓毒症小鼠的肺上皮铁凋亡并减轻了肺损伤.一起来看,这些数据提供了对脓毒症诱导的ALI的潜在机制的创新性理解,并为脓毒症提供了有前景的治疗策略.
    Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. The inflammatory cytokine storm causes systemic organ damage, especially acute lung injury in sepsis. In this study, we found that the expression of S-phase kinase-associated protein 2 (Skp2) was significantly decreased in sepsis-induced acute lung injury (ALI). Sepsis activated the MEK/ERK pathway and inhibited Skp2 expression in the pulmonary epithelium, resulting in a reduction of K48 ubiquitination of solute carrier family 3 member 2 (SLC3A2), thereby impairing its membrane localization and cystine/glutamate exchange function. Consequently, the dysregulated intracellular redox reactions induced ferroptosis in pulmonary epithelial cells, leading to lung injury. Finally, we demonstrated that intravenous administration of Skp2 mRNA-encapsulating lipid nanoparticles (LNPs) inhibited ferroptosis in the pulmonary epithelium and alleviated lung injury in septic mice. Taken together, these data provide an innovative understanding of the underlying mechanisms of sepsis-induced ALI and a promising therapeutic strategy for sepsis.
    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
    如金解毒汤(RJJDD)是中药的经典方剂,长期以来一直用于治疗由外部感染引起的肺炎,但它是否以及如何对流感病毒治疗有益仍不清楚。本研究旨在研究RJJDD对小鼠流感模型的抗炎作用,并探讨其可能的作用机制。
    将小鼠用PBS模拟感染或用PR8病毒感染,然后用RJJDD或抗病毒奥司他韦处理。每天监测小鼠的体重减轻和发病率。网络药理学用于探索RJJDD可能调节的潜在途径。进行qRT-PCR和ELISA以评估炎症细胞因子在肺组织和巨噬细胞中的表达。收集肠道粪便进行16SrDNA测序以评估肠道微生物群的变化。
    我们证明RJJDD可预防IAV引起的肺炎。MassSpec鉴定的RJJDD成分的综合网络药理学分析表明,RJJDD可能通过下调关键信号通路产生炎症细胞因子,IAV感染的小鼠肺组织和IAV单链RNA模拟物R837诱导的巨噬细胞中的细胞因子表达分析证实了这一点。此外,肠道菌群分析表明,RJJDD可预防IAV诱导的宿主肠道菌群失调,从而为RJJDD在流感肺炎中的疗效提供了一个机制解释。
    这项研究定义了RJJDD在预防流感中以前未表征的作用,可能是通过维持肠道微生物群的稳态,并为严重流感提供了新的治疗选择。
    UNASSIGNED: Rujin Jiedu decoction (RJJDD) is a classical prescription of Traditional Chinese Medicine that has long been applied to treat pneumonia caused by external infection, but whether and how it benefits influenza virus therapy remains largely unclear. The aim of this study was to investigate the anti-inflammatory effect of RJJDD on the mouse model of influenza and to explore its potential mechanism.
    UNASSIGNED: The mice were mock-infected with PBS or infected with PR8 virus followed by treatment with RJJDD or antiviral oseltamivir. The weight loss and morbidity of mice were monitored daily. Network pharmacology is used to explore the potential pathways that RJJDD may modulate. qRT-PCR and ELISA were performed to assess the expression of inflammatory cytokines in the lung tissue and macrophages. The intestinal feces were collected for 16S rDNA sequencing to assess the changes in gut microbiota.
    UNASSIGNED: We demonstrate that RJJDD protects against IAV-induced pneumonia. Comprehensive network pharmacology analyses of the Mass Spec-identified components of RJJDD suggest that RJJDD may act through down-regulating key signaling pathways producing inflammatory cytokines, which was experimentally confirmed by cytokine expression analysis in IAV-infected mouse lung tissues and IAV single-strand RNA mimic R837-induced macrophages. Furthermore, gut microbiota analysis indicates that RJJDD prevented IAV-induced dysbiosis of host intestinal flora, thereby offering a mechanistic explanation for RJJDD\'s efficacy in influenza pneumonia.
    UNASSIGNED: This study defines a previously uncharacterized role for RJJDD in protecting against influenza likely by maintaining homeostasis of gut microbiota, and provides a new therapeutic option for severe influenza.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号