host-pathogen interactions

宿主 - 病原体相互作用
  • 文章类型: Journal Article
    程序性细胞死亡(PCD)在维持人体消化道的正常结构和功能中起着至关重要的作用。幽门螺杆菌感染(H.幽门螺杆菌)是导致胃损伤的重要因素,促进Correa级联反应并加速从胃炎到胃癌的过渡。最近的研究表明,在幽门螺杆菌感染过程中,一些PCD信号通路异常激活,PCD的功能障碍被认为是胃癌发生发展的原因,并干扰治疗。随着H.pylori感染PCD研究的不断深入,探索幽门螺杆菌与机体在不同PCD通路中的相互作用机制,可能成为未来治疗幽门螺杆菌感染和幽门螺杆菌相关性胃癌的重要研究方向。此外,可以抑制或诱导PCD的生物活性化合物可以作为治疗这种疾病的关键要素。在这次审查中,我们简要描述了PCD的过程,探讨PCD信号通路与H.pylori感染或H.pylori相关性胃癌的作用机制,并总结了在此过程中可能在每个PCD途径中起治疗作用的活性分子,以期更全面地了解PCD在幽门螺杆菌感染中的作用。
    Programmed cell death (PCD) plays a crucial role in maintaining the normal structure and function of the digestive tract in the body. Infection with Helicobacter pylori (H. pylori) is an important factor leading to gastric damage, promoting the Correa cascade and accelerating the transition from gastritis to gastric cancer. Recent research has shown that several PCD signaling pathways are abnormally activated during H. pylori infection, and the dysfunction of PCD is thought to contribute to the development of gastric cancer and interfere with treatment. With the deepening of studies on H. pylori infection in terms of PCD, exploring the interaction mechanisms between H. pylori and the body in different PCD pathways may become an important research direction for the future treatment of H. pylori infection and H. pylori-related gastric cancer. In addition, biologically active compounds that can inhibit or induce PCD may serve as key elements for the treatment of this disease. In this review, we briefly describe the process of PCD, discuss the interaction between different PCD signaling pathways and the mechanisms of H. pylori infection or H. pylori-related gastric cancer, and summarize the active molecules that may play a therapeutic role in each PCD pathway during this process, with the expectation of providing a more comprehensive understanding of the role of PCD in H. pylori 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
    微孢子虫是细胞内真核病原体,对免疫受损的宿主构成重大威胁。这些病原体在感染期间操纵宿主细胞的方式仍然知之甚少。使用邻近生物素化策略,我们确定微孢子虫EnP1是修饰宿主细胞环境的核靶向效应物。EnP1向宿主细胞核的易位通过核定位信号(NLS)进行冥想。在细胞核中,EnP1与宿主组蛋白H2B相互作用。这种相互作用破坏了H2B单氮化(H2Bub),随后影响p53表达。至关重要的是,这种对p53的抑制作用减弱了其对下游靶基因SLC7A11的控制,增强了宿主细胞在微孢子虫感染期间抵抗铁凋亡的能力。这种有利条件促进了宿主细胞内微孢子虫的增殖。这些发现揭示了微孢子虫修饰其宿主细胞以促进其存活的分子机制。
    Microsporidia are intracellular eukaryotic pathogens that pose a substantial threat to immunocompromised hosts. The way these pathogens manipulate host cells during infection remains poorly understood. Using a proximity biotinylation strategy we established that microsporidian EnP1 is a nucleus-targeted effector that modifies the host cell environment. EnP1\'s translocation to the host nucleus is meditated by nuclear localization signals (NLSs). In the nucleus, EnP1 interacts with host histone H2B. This interaction disrupts H2B monoubiquitination (H2Bub), subsequently impacting p53 expression. Crucially, this inhibition of p53 weakens its control over the downstream target gene SLC7A11, enhancing the host cell\'s resilience against ferroptosis during microsporidian infection. This favorable condition promotes the proliferation of microsporidia within the host cell. These findings shed light on the molecular mechanisms by which microsporidia modify their host cells to facilitate their survival.
    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
    枯萎病是一种真菌病原体,在农业生产中造成重大损失。由于其传输迅速,主机范围广,探索与感染索兰尼的防御反应有关的基因已成为一项重要任务。这里,我们进行了时程RNA-Seq实验,以探索在接种后6,12,24,36,48和72小时(hpi)参与宿主对枯萎病菌AG3-TB感染应答的关键基因或途径.GO和KEGG富集分析显示,大多数DEGs富集在基础代谢途径中,包括碳水化合物代谢过程和氨基酸的生物合成。此外,过氧化氢酶(CAT)和超氧化物歧化酶(SOD)表达上调,和转录因子(TFs),如WRKY,与对照(0hpi)相比,AP2和MYB显著增加。WRKY70和过氧化氢酶-3的沉默表现出对真菌感染的敏感性升高。总结一下,TFsWRKY70和WRKY75,参与茉莉酸(JA)的基因,水杨酸(SA),和油菜素类固醇(BR)信号通路,防御相关酶可能在宿主对枯萎病菌AG3-TB感染的反应中起关键作用。
    Rhizoctonia solani is a fungal pathogen that causes significant losses in agricultural production. Because of its rapid transmission and broad host range, the exploration of genes involved in defense responses to the infection of R. solani has become an important task. Here, we performed a time-course RNA-Seq experiment to explore crucial genes or pathways involved in host responses to R. solani AG3-TB infection at 6, 12, 24, 36, 48, and 72 hours post inoculation (hpi). GO and KEGG enrichment analysis revealed that most DEGs were enriched in the basal metabolism pathways, including carbohydrate metabolic processes and the biosynthesis of amino acids. Moreover, catalase (CAT) and superoxide dismutase (SOD) were up-regulated, and transcription factors (TFs) such as WRKY, AP2, and MYB were increased significantly compared to the control (0 hpi). Silencing of WRKY70 and catalase-3 exhibited elevated susceptibility to the fungal infection. To summarize, the TFs WRKY70 and WRKY75, genes involved in jasmonic acid (JA), salicylic acid (SA), and brassinosteroids (BR) signaling pathways, and defense-related enzymes may play crucial roles in the host responses to R. solani AG3-TB 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
    几丁质酶基因,作为一类细胞壁水解酶,对尖孢镰刀菌的发育和发病机理至关重要。血管感染(F.ox)在棉花中,但相关研究集中在几丁质酶基因是有限的。本研究从高抗性品种新海41和敏感品种新海14中探索了两个海岛棉根系分泌物,通过加权相关网络分析(WGCNA)研究了它们与F.ox的相互作用。因此,出现了两个与真菌致病性相关的模块。此外,共鉴定出25个几丁质酶基因。最后,进行FoChi20的宿主诱导基因沉默(HIGS),棉花植株表现出明显温和的病害,病害指数明显低于对照。本研究阐明几丁质酶基因在棉花枯萎病菌的致病性中起着至关重要的作用。FoChi20基因可能参与F.ox的发病机制和宿主-病原体相互作用,建立了海岛棉病害防治的理论框架。
    Chitinase genes, as a class of cell wall hydrolases, are essential for the development and pathogenesis of Fusarium oxysporum f.sp. vasinfectum (F. ox) in cotton, but related research focused on chitinase genes are limited. This study explored two island cotton root secretions from the highly resistant cultivar Xinhai 41 and sensitive cultivar Xinhai 14 to investigate their interaction with F. ox by a weighted correlation network analysis (WGCNA). As a result, two modules that related to the fungal pathogenicity emerged. Additionally, a total of twenty-five chitinase genes were identified. Finally, host-induced gene silencing (HIGS) of FoChi20 was conducted, and the cotton plants showed noticeably milder disease with a significantly lower disease index than the control. This study illuminated that chitinase genes play crucial roles in the pathogenicity of cotton wilt fungi, and the FoChi20 gene could participate in the pathogenesis of F. ox and host-pathogen interactions, which establishes a theoretical framework for disease control in Sea Island cotton.
    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
    巨细胞病毒(CMV)由于其抵消宿主抗病毒先天性免疫反应的能力,已成功在人类中建立了持久的潜伏感染。在与宿主的协同进化过程中,该病毒已经进化出各种逃避技术来逃避宿主的先天免疫监视。目前,目前还没有疫苗可用于预防和治疗CMV感染,CMV感染与宿主抗病毒先天性免疫之间的相互作用尚不清楚。然而,正在进行的研究将为如何治疗和预防CMV感染及其相关疾病提供新的见解。这里,我们更新了最近关于CMV如何逃避抗病毒先天免疫的研究,重点关注CMV蛋白如何靶向和破坏抗病毒先天性免疫信号通路的关键衔接子。这篇综述还讨论了一些经典的内在细胞防御,这些防御对于抵抗病毒入侵至关重要。全面审查CMV抗病毒先天免疫的逃避机制将有助于研究人员确定新的治疗靶标并开发针对CMV感染的疫苗。
    Cytomegalovirus (CMV) has successfully established a long-lasting latent infection in humans due to its ability to counteract the host antiviral innate immune response. During coevolution with the host, the virus has evolved various evasion techniques to evade the host\'s innate immune surveillance. At present, there is still no vaccine available for the prevention and treatment of CMV infection, and the interaction between CMV infection and host antiviral innate immunity is still not well understood. However, ongoing studies will offer new insights into how to treat and prevent CMV infection and its related diseases. Here, we update recent studies on how CMV evades antiviral innate immunity, with a focus on how CMV proteins target and disrupt critical adaptors of antiviral innate immune signaling pathways. This review also discusses some classic intrinsic cellular defences that are crucial to the fight against viral invasion. A comprehensive review of the evasion mechanisms of antiviral innate immunity by CMV will help investigators identify new therapeutic targets and develop vaccines against CMV 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
    orf病毒(ORFV)对家庭小反刍动物的健康构成严重威胁(即,绵羊和山羊)和全球范围内的人类,每年给畜牧业造成约1.5亿美元的损失。然而,涉及ORFV感染和复制的宿主因素仍然难以捉摸。在这项研究中,我们比较了ORFV感染或未感染的绵羊睾丸间质细胞(STIC)的RNA-seq谱,并鉴定了一个新的宿主基因,钾电压门控通道亚家族E成员4(KCNE4),作为参与ORFV感染的关键宿主因子。RNA-seq数据和RT-qPCR测定均显示,在感染后(hpi)9至48小时,感染的STIC中KCNE4的表达显著增加。另一方面,RT-qPCR检测到ORFV感染后KCNE4siRNA和KCNE4敲除(KO)HeLa细胞转染的STIC中ORFV拷贝数减少,在24hpi时,KOHeLa细胞中ORFV-GFP的荧光比率降低,表明KCNE4对ORFV感染至关重要。此外,附着和内化分析显示ORFV附着减少,内化,复制,并由KOHeLa细胞释放,证明了KCNE4对ORFV进入细胞的潜在抑制作用。用KCNE4抑制剂如奎尼丁和氟西汀预处理显著抑制ORFV感染。我们所有的发现都揭示了KCNE4作为ORFV进入和复制的新型宿主调节因子,为ORFV感染的相互作用机制提供了新的见解。该研究还强调了K+通道可能是阻止病毒感染和疾病的药物靶标。
    The orf virus (ORFV) poses a serious threat to the health of domestic small ruminants (i.e., sheep and goats) and humans on a global scale, causing around $150 million in annual losses to livestock industry. However, the host factors involved in ORFV infection and replication are still elusive. In this study, we compared the RNA-seq profiles of ORFV-infected or non-infected sheep testicular interstitial cells (STICs) and identified a novel host gene, potassium voltage-gated channel subfamily E member 4 (KCNE4), as a key host factor involved in the ORFV infection. Both RNA-seq data and RT-qPCR assay revealed a significant increase in the expression of KCNE4 in the infected STICs from 9 to 48 h post infection (hpi). On the other hand, the RT-qPCR assay detected a decrease in ORFV copy number in both the STICs transfected by KCNE4 siRNA and the KCNE4 knockout (KO) HeLa cells after the ORFV infection, together with a reduced fluorescence ratio of ORFV-GFP in the KO HeLa cells at 24 hpi, indicating KCNE4 to be critical for the ORFV infection. Furthermore, the attachment and internalization assays showed decreased ORFV attachment, internalization, replication, and release by the KO HeLa cells, demonstrating a potential inhibition of ORFV entry into the cells by KCNE4. Pretreatment with the KCNE4 inhibitors such as quinidine and fluoxetine significantly repressed the ORFV infection. All our findings reveal KCNE4 as a novel host regulator of the ORFV entry and replication, shedding new insight into the interactive mechanism of ORFV infection. The study also highlights the K+ channels as possible druggable targets to impede viral infection and disease.
    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
    结核病(TB)仍然是全球最严重的健康挑战之一。结核分枝杆菌(M.结核病),病原体,采用复杂的免疫逃避和发病策略。其在免疫细胞内茁壮成长并激发强烈的炎症反应的能力延长了感染和传播。分枝杆菌的高级适应促进了通过人类免疫系统的导航,并在整个不同的感染阶段呈现可变的抗原谱。研究这些策略揭示了针对结核病的有效疫苗开发的针对性方法。这篇综述深入研究了结核分枝杆菌在各个感染阶段的免疫逃避策略和致病过程的最先进和详尽的见解。从该分析中提取的知识有望指导创新的结核病疫苗的创建,并将理论基础转化为实际的免疫防御。
    Tuberculosis (TB) remains one of the gravest global health challenges. Mycobacterium tuberculosis (M. tuberculosis), the causative agent, employs sophisticated immune evasion and pathogenesis strategies. Its capability to thrive within immune cells and incite robust inflammatory responses prolongs infection and dissemination. Mycobacterial advanced adaptations facilitate navigation through the human immune system and present a variable antigenic profile throughout different infection stages. Investigating these strategies unfolds targeted approaches to effective vaccine development against TB. This review delves into the most advanced and exhaustive insights into the immune evasion tactics and pathogenic processes of M. tuberculosis across various infection stages. The knowledge distilled from this analysis holds the promise of guiding the creation of innovative TB vaccines and translating theoretical groundwork into practical immunological defenses.
    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
    蝙蝠携带可以感染其他哺乳动物的高毒力病毒,包括人类,对他们的免疫耐受机制提出质疑。蝙蝠细胞采用多种策略来限制病毒复制和病毒诱导的免疫病理学,但是蝙蝠和致命病毒的共存仍然知之甚少。这里,我们研究了蝙蝠细胞中的抗病毒RNA干扰途径,发现它们具有增强的抗病毒RNAi应答,在人类细胞中缺失的辛德毕斯病毒感染后产生典型的病毒小干扰RNA。Dicer功能的破坏导致蝙蝠细胞中三种不同RNA病毒的病毒载量增加,表明干扰素非依赖性抗病毒途径。此外,我们的发现揭示了Dicer和模式识别受体的同时参与,如维甲酸诱导基因I,双链RNA,这表明Dicer减弱了蝙蝠细胞的干扰素反应。这些见解促进了我们对蝙蝠与病毒共存的独特策略的理解。
    Bats harbor highly virulent viruses that can infect other mammals, including humans, posing questions about their immune tolerance mechanisms. Bat cells employ multiple strategies to limit virus replication and virus-induced immunopathology, but the coexistence of bats and fatal viruses remains poorly understood. Here, we investigate the antiviral RNA interference pathway in bat cells and discover that they have an enhanced antiviral RNAi response, producing canonical viral small interfering RNAs upon Sindbis virus infection that are missing in human cells. Disruption of Dicer function results in increased viral load for three different RNA viruses in bat cells, indicating an interferon-independent antiviral pathway. Furthermore, our findings reveal the simultaneous engagement of Dicer and pattern-recognition receptors, such as retinoic acid-inducible gene I, with double-stranded RNA, suggesting that Dicer attenuates the interferon response initiation in bat cells. These insights advance our comprehension of the distinctive strategies bats employ to coexist with viruses.
    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
    人乳头瘤病毒(HPV)11/16E6/E7蛋白已被认为在病毒发病机理中至关重要。这项研究试图揭示HPV11/16E6/E7转染的角质形成细胞如何抑制外周血单核细胞(PBMC)中细胞因子分泌的潜在机制。在以非接触方式共培养HPV11/16E6/E7转染的角质形成细胞与PBMC时,我们观察到PBMC分泌的各种细胞因子明显减少。为了确定这种抑制是否由特定的常见分泌因子介导,我们对这些转染细胞进行了转录组测序。该分析在所有四种HPV转染的细胞中鉴定了53个常见的差异分泌基因。生物信息学分析表明这些基因主要参与免疫调节。定量PCR(qPCR)和大量文献综述的结果表明12个基因(ACE2,BMP3,BPIFB1,CLU,CST6,CTF1,HMGB2,MMP12,PDGFA,RNASE7,SULF2,TGM2),和7个基因的上调(CCL17,CCL22,FBLN1,PLAU,S100A7,S100A8,S100A9),可能在调节肿瘤免疫和对抗病原体感染方面至关重要,基因S100A8和S100A9,IL-17信号通路尤其值得注意。因此,HPV11/16E6/E7蛋白可能通过改变宿主分泌基因的表达来抑制免疫细胞的细胞因子分泌。对这些基因的进一步探索可能会对HPV感染的复杂动力学产生新的见解。
    Human papillomavirus (HPV) 11/16 E6/E7 proteins have been recognized to be pivotal in viral pathogenesis. This study sought to uncover the potential mechanisms of how HPV11/16 E6/E7-transfected keratinocytes inhibit cytokine secretion in peripheral blood mononuclear cells (PBMC). Upon co-culturing HPV11/16 E6/E7-transfected keratinocytes with PBMC in a non-contact manner, we observed a marked decrease in various cytokines secreted by PBMC. To determine if this suppression was mediated by specific common secreted factors, we conducted transcriptomic sequencing on these transfected cells. This analysis identified 53 common differentially secreted genes in all four HPV-transfected cells. Bioinformatics analysis demonstrated these genes were predominantly involved in immune regulation. Results from quantitative PCR (qPCR) and an extensive literature review suggested the downregulation of 12 genes (ACE2, BMP3, BPIFB1, CLU, CST6, CTF1, HMGB2, MMP12, PDGFA, RNASE7, SULF2, TGM2), and upregulation of 7 genes (CCL17, CCL22, FBLN1, PLAU, S100A7, S100A8, S100A9), may be crucial in modulating tumor immunity and combating pathogenic infections, with genes S100A8 and S100A9, and IL-17 signaling pathway being particularly noteworthy. Thus, HPV11/16 E6/E7 proteins may inhibit cytokine secretion of immune cells by altering the expression of host-secreted genes. Further exploration of these genes may yield new insights into the complex dynamics of HPV 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
    探讨气道微生物组对慢性阻塞性肺疾病急性加重(AECOPD)的作用机制。
    我们招募了31名AECOPD患者和26名稳定期COPD患者,他们的痰样本被收集用于宏基因组和RNA测序,然后进行生物信息学分析。使用同一批样品通过定量实时PCR(qPCR)验证宿主基因的表达。
    我们的结果表明,在AECOPD组(p=0.015)和流感嗜血杆菌(p=0.005)的表达较高。检测到的不同表达基因(DEGs)在基因功能注释中显著富集于“Ⅰ型干扰素信号通路”(p<0.001,q=0.001),和“细胞溶质DNA传感途径”(p=0.002,q=0.024),“Toll样受体信号通路”(p=0.006,q=0.045),和“TNF信号通路”(p=0.006,q=0.045)在KEGG富集分析中。qPCR扩增实验证实AECOPD组OASL和IL6的表达显著增高。
    肺细菌菌群失调可能通过I型干扰素信号通路和Toll样受体信号通路等固有免疫系统通路调节AECOPD的发病机制。
    UNASSIGNED: To explore the underlying mechanisms the airway microbiome contributes to Acute Exacerbation of Chronic Obstructive Pulmonary Disease(AECOPD).
    UNASSIGNED: We enrolled 31 AECOPD patients and 26 stable COPD patients, their sputum samples were collected for metagenomic and RNA sequencing, and then subjected to bioinformatic analyses. The expression of host genes was validated by Quantitative Real-time PCR(qPCR) using the same batch of specimens.
    UNASSIGNED: Our results indicated a higher expression of Rothia mucilaginosa(p=0.015) in the AECOPD group and Haemophilus influenzae(p=0.005) in the COPD group. The Different expressed genes(DEGs) detected were significantly enriched in \"type I interferon signaling pathway\"(p<0.001, q=0.001) in gene function annotation, and \"Cytosolic DNA-sensing pathway\"(p=0.002, q=0.024), \"Toll-like receptor signaling pathway\"(p=0.006, q=0.045), and \"TNF signaling pathway\"(p=0.006, q=0.045) in KEGG enrichment analysis. qPCR amplification experiment verified that the expression of OASL and IL6 increased significantly in the AECOPD group.
    UNASSIGNED: Pulmonary bacteria dysbiosis may regulate the pathogenesis of AECOPD through innate immune system pathways like type I interferon signaling pathway and Toll-like receptor signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号