Host-pathogen interactions

宿主 - 病原体相互作用
  • 文章类型: Journal Article
    要对复杂系统进行建模,基于个人的模型(IBM),有时称为“基于代理的模型”(ABM),通过元素的适当表示来描述系统的简化。IBM模拟系统中离散个体/主体的行为和交互,以发现来自这些交互的行为模式。生物系统中的个体/试剂的实例是个体免疫细胞和细菌,其独立地具有由行为规则定义的自身独特属性。在IBM中,这些代理中的每一个都驻留在空间环境中,并且交互由预定义的规则指导。这些规则通常很简单,可以很容易地实现。预计在这些规则的指导下进行交互之后,我们将对代理-代理交互以及代理-环境交互有更好的了解。必须考虑由概率分布描述的随机性。很少发生的事件,如罕见突变的积累,可以很容易地建模。因此,IBM能够跟踪模型中每个个人/代理的行为,同时还可以获取有关其集体行为结果的信息。可以捕获一个代理对另一个代理的影响,从而允许在总体结果上充分表示直接和间接因果关系。这意味着可以获得重要的新见解并测试假设。
    To model complex systems, individual-based models (IBMs), sometimes called \"agent-based models\" (ABMs), describe a simplification of the system through an adequate representation of the elements. IBMs simulate the actions and interaction of discrete individuals/agents within a system in order to discover the pattern of behavior that comes from these interactions. Examples of individuals/agents in biological systems are individual immune cells and bacteria that act independently with their own unique attributes defined by behavioral rules. In IBMs, each of these agents resides in a spatial environment and interactions are guided by predefined rules. These rules are often simple and can be easily implemented. It is expected that following the interaction guided by these rules we will have a better understanding of agent-agent interaction as well as agent-environment interaction. Stochasticity described by probability distributions must be accounted for. Events that seldom occur such as the accumulation of rare mutations can be easily modeled.Thus, IBMs are able to track the behavior of each individual/agent within the model while also obtaining information on the results of their collective behaviors. The influence of impact of one agent with another can be captured, thus allowing a full representation of both direct and indirect causation on the aggregate results. This means that important new insights can be gained and hypotheses tested.
    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
    数学模型已用于研究传染病在人与人之间的传播。最近的研究正在开发宿主内建模,该模型提供了对病原体细菌,真菌,寄生虫,或病毒-发展,传播,并在单个体内进化,以及它们与宿主免疫系统的相互作用。这样的模型有可能提供对宿主内疾病的发病机理的更详细和完整的描述,并鉴定可能无法检测到的其他影响因素。数学模型可用于帮助理解全球抗生素耐药性(ABR)危机,并确定应对这种威胁的新方法。当细菌响应随机或选择性压力并通过获得新的遗传性状来适应新的环境时,就会发生ABR。这通常是通过从其他细菌中获取DNA片段,一个叫做水平基因转移(HGT)的过程,对细菌中的一段DNA的修饰,或通过。细菌已经进化出机制,使它们能够通过突变来应对环境威胁,和水平基因转移(HGT):接合;转导;和转化。HGT在全球范围内传播抗生素耐药性的常见机制是共轭,因为它允许移动遗传元件(MGEs)的直接转移。虽然有几个MGE,质粒和转座子是促进细菌群体中抗菌药物抗性基因发展和快速传播的最重要的MGE。可以对上面提到的每个抗性扩散机制进行建模,从而使我们能够更好地理解过程并定义减少抗性的策略。
    Mathematical models have been used to study the spread of infectious diseases from person to person. More recently studies are developing within-host modeling which provides an understanding of how pathogens-bacteria, fungi, parasites, or viruses-develop, spread, and evolve inside a single individual and their interaction with the host\'s immune system.Such models have the potential to provide a more detailed and complete description of the pathogenesis of diseases within-host and identify other influencing factors that may not be detected otherwise. Mathematical models can be used to aid understanding of the global antibiotic resistance (ABR) crisis and identify new ways of combating this threat.ABR occurs when bacteria respond to random or selective pressures and adapt to new environments through the acquisition of new genetic traits. This is usually through the acquisition of a piece of DNA from other bacteria, a process called horizontal gene transfer (HGT), the modification of a piece of DNA within a bacterium, or through. Bacteria have evolved mechanisms that enable them to respond to environmental threats by mutation, and horizontal gene transfer (HGT): conjugation; transduction; and transformation. A frequent mechanism of HGT responsible for spreading antibiotic resistance on the global scale is conjugation, as it allows the direct transfer of mobile genetic elements (MGEs). Although there are several MGEs, the most important MGEs which promote the development and rapid spread of antimicrobial resistance genes in bacterial populations are plasmids and transposons. Each of the resistance-spread-mechanisms mentioned above can be modeled allowing us to understand the process better and to define strategies to reduce resistance.
    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
    越来越需要新的治疗方案来对抗抗生素抗性细菌菌株。金黄色葡萄球菌是一种临床上重要的、对一系列抗生素产生抗药性的机会主义病原体。系统疾病的斑马鱼幼虫模型已越来越多地用于阐明金黄色葡萄球菌的毒力机制和宿主-病原体相互作用。这里,我们概述了该模型如何用于研究不同抗生素单独和联合使用对金黄色葡萄球菌的作用.
    There is an increasing need for new treatment regimens to combat antibiotic-resistant strains of bacteria. Staphylococcus aureus is a clinically important, opportunist pathogen that has developed resistance to a range of antibiotics. The zebrafish larval model of systemic disease has been increasingly utilized to elucidate S. aureus virulence mechanisms and host-pathogen interactions. Here, we outline how this model can be used to investigate the effects of different antibiotics alone and in combination against S. aureus.
    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
    COVID-19大流行期间的研究表明,与成年人相比,儿童的鼻先天免疫反应增强。为了评估鼻腔病毒和细菌在驱动这些反应中的作用,我们进行了细胞因子分析和全面,在2021-22年接受SARS-CoV-2检测的儿童鼻咽样本中,呼吸道病毒和细菌性病原体的症状无关性检测(n=467).呼吸道病毒和/或病原体非常普遍(82%的有症状儿童和30%的无症状儿童;90%和49%的<5岁儿童)。病毒检测和载量与鼻干扰素反应生物标志物CXCL10相关,先前报道的SARS-CoV-2病毒载量与鼻干扰素反应之间的差异可通过病毒共感染来解释。细菌病原体与IL-1β和TNF升高的明显促炎反应相关,但与CXCL10无关。此外,分开1-2周收集的健康1岁儿童的配对样本显示呼吸道病毒频繁获取或清除,与粘膜免疫表型平行变化。这些发现表明,动态的宿主-病原体相互作用驱动儿童鼻先天免疫激活.
    Studies during the COVID-19 pandemic showed that children had heightened nasal innate immune responses compared with adults. To evaluate the role of nasal viruses and bacteria in driving these responses, we performed cytokine profiling and comprehensive, symptom-agnostic testing for respiratory viruses and bacterial pathobionts in nasopharyngeal samples from children tested for SARS-CoV-2 in 2021-22 (n = 467). Respiratory viruses and/or pathobionts were highly prevalent (82% of symptomatic and 30% asymptomatic children; 90 and 49% for children <5 years). Virus detection and load correlated with the nasal interferon response biomarker CXCL10, and the previously reported discrepancy between SARS-CoV-2 viral load and nasal interferon response was explained by viral coinfections. Bacterial pathobionts correlated with a distinct proinflammatory response with elevated IL-1β and TNF but not CXCL10. Furthermore, paired samples from healthy 1-year-olds collected 1-2 wk apart revealed frequent respiratory virus acquisition or clearance, with mucosal immunophenotype changing in parallel. These findings reveal that frequent, dynamic host-pathogen interactions drive nasal innate immune activation in children.
    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: Letter
    暂无摘要。
    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
    痘病毒已经进化出一系列逃避免疫反应的机制,我们提供了不同免疫调节策略的概述。痘病毒会阻止病毒DNA的识别,从而触发免疫反应并抑制受感染细胞内的信号传导途径。痘病毒的一个独特特征是产生模拟细胞因子和细胞因子受体的分泌蛋白,充当诱饵受体以中和细胞因子和趋化因子的活性。这些蛋白质通过抑制细胞因子激活逃避细胞免疫反应的能力得到痘病毒阻断自然杀伤细胞和细胞毒性T细胞的策略的补充。通常通过干扰抗原呈递途径。靶向补体激活的机制也由痘病毒编码。靶向免疫分子和途径的病毒编码蛋白在免疫调节中起主要作用,以及它们对病毒发病机理的贡献,促进病毒复制或预防免疫病理学,正在讨论。
    Poxviruses have evolved a wide array of mechanisms to evade the immune response, and we provide an overview of the different immunomodulatory strategies. Poxviruses prevent the recognition of viral DNA that triggers the immune responses and inhibit signaling pathways within the infected cell. A unique feature of poxviruses is the production of secreted proteins that mimic cytokines and cytokine receptors, acting as decoy receptors to neutralize the activity of cytokines and chemokines. The capacity of these proteins to evade cellular immune responses by inhibiting cytokine activation is complemented by poxviruses\' strategies to block natural killer cells and cytotoxic T cells, often through interfering with antigen presentation pathways. Mechanisms that target complement activation are also encoded by poxviruses. Virus-encoded proteins that target immune molecules and pathways play a major role in immune modulation, and their contribution to viral pathogenesis, facilitating virus replication or preventing immunopathology, is discussed.
    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
    手,脚,和口蹄疫(HFMD)是由肠道病毒71(EV71)引起的常见传染病,经常影响儿童,在某些情况下导致严重感染。总的来说,当感染发生时,身体上调炎症反应以消除病原微生物,保护宿主免受感染。然而,EV71可能抑制宿主的先天免疫以促进病毒感染。目前,尚不完全了解EV71如何劫持宿主细胞进行自身复制。Toll样受体4(TLR4),天然免疫受体,历史上与细菌内毒素诱导的炎症反应相关。然而,目前尚不清楚在EV71感染期间TLR4是否以及如何改变.在这项研究中,我们在RD中观察到TLR4蛋白和基因转录水平的降低,EV71感染后的GES-1和Vero细胞,通过RT-qPCR检测,免疫荧光染色和蛋白质印迹。此外,我们观察到MYD88的TLR4下游分子、p-NF-κBp65、p-TBK1和相关炎性细胞因子也减少,提示抗病毒先天性免疫和炎症反应被抑制。为了确定TLR4变化对EV71感染的影响,我们用TLR4激动剂或抑制剂干扰EV71感染的RD细胞,结果表明TLR4的激活抑制了EV71的复制,而抑制TLR4促进EV71复制。此外,在TLR4siRNA转染和EV71感染的RD细胞中也促进了EV71的复制。这表明EV71下调TLR4的表达可以抑制宿主的免疫防御以促进EV71的自我复制。这种新机制可能是EV71逃避宿主免疫的一种策略。
    Hand, foot, and mouth disease (HFMD) is a common infectious disease caused by enterovirus 71 (EV71) that frequently affects children, leading to severe infections in some cases. In general, when infection occurs, the body upregulates inflammatory responses to eliminate pathogenic microorganisms to protect the host from infection. However, EV71 may inhibit host\'s innate immunity to promote virus infection. At present, it is not fully understood how EV71 hijack the host cells for its own replication. Toll-like receptor 4 (TLR4), a natural immune receptor, historically associated with bacterial endotoxin-induced inflammatory responses. However, it is still unclear whether and how TLR4 is altered during EV71 infection. In this study, we observed a reduction in both TLR4 protein and gene transcript levels in RD, GES-1, and Vero cells following EV71 infection, as detected by RT-qPCR, immunofluorescence staining and western blot. Furthermore, we observed that the TLR4 downstream molecules of MYD88, p-NF-κB p65, p-TBK1 and related inflammatory cytokines were also reduced, suggesting that antiviral innate immune and inflammatory response were suppressed. To determine the impact of TLR4 changes on EV71 infection, we interfered EV71-infected RD cells with TLR4 agonist or inhibitor and the results showed that activation of TLR4 inhibited EV71 replication, while inhibition of TLR4 promote EV71 replication. Besides, EV71 replication was also promoted in TLR4 siRNA-transfected and EV71-infected RD cells. This suggests that down-regulation the expression of TLR4 by EV71 can inhibit host immune defense to promote EV71 self-replication. This novel mechanism may be a strategy for EV71 to evade host immunity.
    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
    人类嗜T淋巴细胞病毒1型(HTLV-1)是第一个发现的人类致癌逆转录病毒,两种严重疾病的病因已被确定为成人T细胞白血病/淋巴瘤恶性肿瘤和HTLV-1相关性脊髓病/热带痉挛性轻瘫(HAM/TSP),使人衰弱的慢性神经脊髓病。尽管有40多年的分子,HTLV-1相关疾病的组织病理学和免疫学研究,该病毒的毒力和致病性尚待阐明。为什么大多数HTLV-1感染的个体(95%)仍然是无症状携带者的原因尚不清楚。免疫系统对致癌和自身免疫的恶化使HTLV-1成为研究恶性肿瘤和神经炎性疾病的天然探针。此外,其缓慢的全球传播促使公共卫生当局和研究人员,正如世界卫生组织所敦促的那样,专注于根除HTLV-1。相比之下,既没有引入有效的疗法,也没有引入保护性疫苗。这篇综合综述集中于HTLV-1诱导的HAM/TSP的神经炎症倾向的最相关研究。将在表观遗传学上严格讨论HAM/TSP发病机理中对病毒-宿主相互作用的这种强调。这些发现可能为设计和开发适当的HTLV-1疗法的未来研究场所提供启示。
    Human T-lymphotropic virus type-1 (HTLV-1) was the first discovered human oncogenic retrovirus, the etiological agent of two serious diseases have been identified as adult T-cell leukaemia/lymphoma malignancy and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP), a debilitating chronic neuro-myelopathy. Despite more than 40 years of molecular, histopathological and immunological studies on HTLV-1-associated diseases, the virulence and pathogenicity of this virus are yet to be clarified. The reason why the majority of HTLV-1-infected individuals (∼95%) remain asymptomatic carriers is still unclear. The deterioration of the immune system towards oncogenicity and autoimmunity makes HTLV-1 a natural probe for the study of malignancy and neuro-inflammatory diseases. Additionally, its slow worldwide spreading has prompted public health authorities and researchers, as urged by the WHO, to focus on eradicating HTLV-1. In contrast, neither an effective therapy nor a protective vaccine has been introduced. This comprehensive review focused on the most relevant studies of the neuro-inflammatory propensity of HTLV-1-induced HAM/TSP. Such an emphasis on the virus-host interactions in the HAM/TSP pathogenesis will be critically discussed epigenetically. The findings may shed light on future research venues in designing and developing proper HTLV-1 therapeutics.
    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
    众所周知,气道微生物群有助于肺部疾病,如囊性纤维化(CF),但它们对发病机制的贡献仍不清楚。为了提高我们对宿主-微生物相互作用的理解,我们开发了一种基于分析和生物信息学质谱(MS)的综合元蛋白质组学工作流程,用于分析气道疾病患者的临床支气管肺泡灌洗(BAL)样本.将来自BAL细胞颗粒的蛋白质加工并合并在按疾病状态分类的组中(CF与非CF)和细菌多样性,基于先前进行的小亚基rRNA测序数据。将来自每个汇集的样品组的蛋白质消化并进行液相色谱串联质谱(MS/MS)。使用宏基因组学指导的蛋白质序列数据库和严格的评估,利用生物信息学工作流程将MS/MS光谱与人和细菌肽序列进行匹配。无标记定量显示,在CF中具有已知作用的蛋白质中,人类肽的丰度不同。比如中性粒细胞弹性蛋白酶和胶原酶,和在CF中具有鲜为人知作用的蛋白质,包括载脂蛋白.从已知的CF病原体中鉴定出差异丰富的细菌肽(例如,假单胞菌),以及其他在CF中具有潜在新作用的分类单元。我们使用这个宿主微生物肽组进行靶向平行反应监测验证,首次证明基于MS的测定可有效定量来自单个CF患者的BAL细胞内的宿主微生物蛋白质动力学。我们集成的生物信息学和分析工作流程结合了发现,验证,和验证应该被证明对于表征气道疾病中微生物贡献者的不同研究是有用的。此外,我们描述了一个有希望的差异丰富的微生物和宿主肽序列的初步小组,用于进一步研究,作为CF疾病发病机制中宿主-微生物关系的潜在标志物。重要意义识别气道疾病中的微生物致病因素和人类反应失调,如CF,对于了解疾病进展和开发更有效的治疗方法至关重要。为此,表征疾病进展过程中从细菌微生物和人类宿主细胞表达的蛋白质可以提供有价值的新见解。我们在这里描述了一种新的方法来自信地检测和监测来自通常从CF患者收集的具有挑战性的BAL样品的微生物和宿主蛋白的丰度变化。我们的方法使用最先进的基于质谱的仪器来检测这些样品中存在的蛋白质,并使用定制的生物信息学软件工具来分析数据并表征检测到的蛋白质及其与CF的关联。我们证明了使用这种方法来表征单个BAL样品中的微生物和宿主蛋白,为了解CF和其他气道疾病的分子贡献者的新方法铺平了道路。
    Airway microbiota are known to contribute to lung diseases, such as cystic fibrosis (CF), but their contributions to pathogenesis are still unclear. To improve our understanding of host-microbe interactions, we have developed an integrated analytical and bioinformatic mass spectrometry (MS)-based metaproteomics workflow to analyze clinical bronchoalveolar lavage (BAL) samples from people with airway disease. Proteins from BAL cellular pellets were processed and pooled together in groups categorized by disease status (CF vs. non-CF) and bacterial diversity, based on previously performed small subunit rRNA sequencing data. Proteins from each pooled sample group were digested and subjected to liquid chromatography tandem mass spectrometry (MS/MS). MS/MS spectra were matched to human and bacterial peptide sequences leveraging a bioinformatic workflow using a metagenomics-guided protein sequence database and rigorous evaluation. Label-free quantification revealed differentially abundant human peptides from proteins with known roles in CF, like neutrophil elastase and collagenase, and proteins with lesser-known roles in CF, including apolipoproteins. Differentially abundant bacterial peptides were identified from known CF pathogens (e.g., Pseudomonas), as well as other taxa with potentially novel roles in CF. We used this host-microbe peptide panel for targeted parallel-reaction monitoring validation, demonstrating for the first time an MS-based assay effective for quantifying host-microbe protein dynamics within BAL cells from individual CF patients. Our integrated bioinformatic and analytical workflow combining discovery, verification, and validation should prove useful for diverse studies to characterize microbial contributors in airway diseases. Furthermore, we describe a promising preliminary panel of differentially abundant microbe and host peptide sequences for further study as potential markers of host-microbe relationships in CF disease pathogenesis.IMPORTANCEIdentifying microbial pathogenic contributors and dysregulated human responses in airway disease, such as CF, is critical to understanding disease progression and developing more effective treatments. To this end, characterizing the proteins expressed from bacterial microbes and human host cells during disease progression can provide valuable new insights. We describe here a new method to confidently detect and monitor abundance changes of both microbe and host proteins from challenging BAL samples commonly collected from CF patients. Our method uses both state-of-the art mass spectrometry-based instrumentation to detect proteins present in these samples and customized bioinformatic software tools to analyze the data and characterize detected proteins and their association with CF. We demonstrate the use of this method to characterize microbe and host proteins from individual BAL samples, paving the way for a new approach to understand molecular contributors to CF and other diseases of the airway.
    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: 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)

公众号