intracellular infections

  • 文章类型: Journal Article
    基于RNA的疫苗引发了核酸药物治疗和预防疾病的范式转变。在两种基于信使RNA的COVID-19疫苗获得全球批准后,核酸疗法和疫苗的开发出现了显著的激增。这种增长是由临床前阶段对许多RNA产物的探索推动的,提供了优于传统方法的几个优点,即,安全,功效,可扩展性,和成本效益。在这一章中,我们概述了各种类型的RNA及其刺激免疫反应和诱导治疗效果的作用机制。此外,本章深入研究了不同的交付系统,特别强调使用纳米颗粒递送RNA。递送系统的选择是一个复杂的过程,涉及开发核酸药物,显着增强其稳定性,生物相容性,和位点特异性。此外,这一章揭示了针对细胞内病原体的RNA疗法和疫苗的临床试验的现状。
    RNA-based vaccines have sparked a paradigm shift in the treatment and prevention of diseases by nucleic acid medicines. There has been a notable surge in the development of nucleic acid therapeutics and vaccines following the global approval of the two messenger RNA-based COVID-19 vaccines. This growth is fueled by the exploration of numerous RNA products in preclinical stages, offering several advantages over conventional methods, i.e., safety, efficacy, scalability, and cost-effectiveness. In this chapter, we provide an overview of various types of RNA and their mechanisms of action for stimulating immune responses and inducing therapeutic effects. Furthermore, this chapter delves into the varying delivery systems, particularly emphasizing the use of nanoparticles to deliver RNA. The choice of delivery system is an intricate process involved in developing nucleic acid medicines that significantly enhances their stability, biocompatibility, and site-specificity. Additionally, this chapter sheds light on the current landscape of clinical trials of RNA therapeutics and vaccines against intracellular pathogens.
    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
    内源性大麻素系统(ECS),最初确定它在维持体内平衡方面的作用,特别是在调节大脑功能方面,已经演变成一个复杂的协调器,影响各种生理过程,超出其与神经系统的原始关联。值得注意的是,越来越多的证据强调了ECS在调节免疫反应中的关键作用。虽然ECS在细菌感染中的具体作用仍在研究中,令人信服的迹象表明其积极参与宿主-病原体相互作用。将ECS纳入细菌病原体感染的框架为我们对其功能的理解引入了一层复杂性。虽然一些研究提出了大麻素调节细菌功能和免疫反应的潜力,结果本身取决于所考虑的特定感染和大麻素.此外,ECS和肠道微生物群之间的双向关系强调了不同生理过程之间复杂的相互作用。ECS的影响力远远超出了它最初的发现,在一系列医疗条件下成为有希望的治疗目标,包括细菌感染,生态失调,还有败血症.这篇综述全面探讨了ECS在细菌调节中的复杂作用,宿主对细菌感染的反应,和微生物组的动态。特别强调大麻素受体类型1和2的作用,其信号传导复杂地影响微生物-宿主相互作用中的免疫细胞功能。
    The endocannabinoid system (ECS), initially identified for its role in maintaining homeostasis, particularly in regulating brain function, has evolved into a complex orchestrator influencing various physiological processes beyond its original association with the nervous system. Notably, an expanding body of evidence emphasizes the ECS\'s crucial involvement in regulating immune responses. While the specific role of the ECS in bacterial infections remains under ongoing investigation, compelling indications suggest its active participation in host-pathogen interactions. Incorporating the ECS into the framework of bacterial pathogen infections introduces a layer of complexity to our understanding of its functions. While some studies propose the potential of cannabinoids to modulate bacterial function and immune responses, the outcomes inherently hinge on the specific infection and cannabinoid under consideration. Moreover, the bidirectional relationship between the ECS and the gut microbiota underscores the intricate interplay among diverse physiological processes. The ECS extends its influence far beyond its initial discovery, emerging as a promising therapeutic target across a spectrum of medical conditions, encompassing bacterial infections, dysbiosis, and sepsis. This review comprehensively explores the complex roles of the ECS in the modulation of bacteria, the host\'s response to bacterial infections, and the dynamics of the microbiome. Special emphasis is placed on the roles of cannabinoid receptor types 1 and 2, whose signaling intricately influences immune cell function in microbe-host interactions.
    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
    卡介苗(BCG)是一种减毒细菌,源自牛分枝杆菌。它是用于预防儿童严重结核病的唯一许可疫苗。除了它对结核病的具体影响,在人和小鼠中异源刺激后,BCG施用还与有益的非特异性效应(NSE)相关。来自BCG的NSE可能与适应性和先天免疫反应有关。后者也被称为训练免疫(TI)。最近描述的先天细胞的生物学特征,能够基于代谢和表观遗传重编程来改善功能。目前,与BCG介导的TI相关的机制是紧张研究的焦点,但是许多差距仍然需要阐明。这篇综述讨论了目前对BCG诱导TI的理解,探索对造血干细胞和单核细胞/巨噬细胞谱系的训练表型至关重要的信号通路。它侧重于BCG介导的TI机制,包括代谢-表观遗传轴和这些细胞中对抗细胞内病原体的炎性小体途径。此外,这项研究探讨了不同免疫细胞类型中的TI,它具有防止各种细胞内感染的能力,以及将经过训练的先天记忆与适应性记忆相结合,以塑造下一代疫苗。
    The bacillus Calmette-Guérin (BCG) is an attenuated bacterium derived from virulent Mycobacterium bovis. It is the only licensed vaccine used for preventing severe forms of tuberculosis in children. Besides its specific effects against tuberculosis, BCG administration is also associated with beneficial non-specific effects (NSEs) following heterologous stimuli in humans and mice. The NSEs from BCG could be related to both adaptive and innate immune responses. The latter is also known as trained immunity (TI), a recently described biological feature of innate cells that enables functional improvement based on metabolic and epigenetic reprogramming. Currently, the mechanisms related to BCG-mediated TI are the focus of intense research, but many gaps are still in need of elucidation. This review discusses the present understanding of TI induced by BCG, exploring signaling pathways that are crucial to a trained phenotype in hematopoietic stem cells and monocytes/macrophages lineage. It focuses on BCG-mediated TI mechanisms, including the metabolic-epigenetic axis and the inflammasome pathway in these cells against intracellular pathogens. Moreover, this study explores the TI in different immune cell types, its ability to protect against various intracellular infections, and the integration of trained innate memory with adaptive memory to shape next-generation vaccines.
    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
    隐藏在宿主细胞中的细胞内细菌病原体耐受先天免疫系统和高剂量抗生素,导致难以治疗的复发性感染。在这里,一种归巢式导弹状纳米治疗剂(FeSAs@Sa。M)由单原子铁纳米酶(FeSAs)核心组成,该核心包被有感染的巨噬细胞膜(Sa。M)被开发用于原位消除细胞内耐甲氧西林金黄色葡萄球菌(MRSA)。机械上,FeSAs@Sa。M最初通过Sa的细菌识别能力与细胞外MRSA结合。M分量。随后,FeSAs@Sa。M可以在其附着的细胞外MRSA的引导下,像归巢导弹一样转运到宿主细胞内MRSA定位区域,通过FeSAs核心的酶活性产生高毒性的活性氧(ROS)用于杀死细胞内MRSA。TheFeSAs@Sa.M在杀死细胞内MRSA方面远远优于FeSA,提出了一种通过在细菌驻留区域原位产生ROS来治疗细胞内感染的可行策略。
    Intracellular bacterial pathogens hiding in host cells tolerate the innate immune system and high-dose antibiotics, resulting in recurrent infections that are difficult to treat. Herein, a homing missile-like nanotherapeutic (FeSAs@Sa.M) composed of a single-atom iron nanozyme (FeSAs) core coated with infected macrophage membrane (Sa.M) is developed for in situ elimination of intracellular methicillin-resistant S. aureus (MRSA). Mechanically, the FeSAs@Sa.M initially binds to the extracellular MRSA via the bacterial recognition ability of the Sa.M component. Subsequently, the FeSAs@Sa.M can be transported to the intracellular MRSA-located regions in the host cell like a homing missile under the guidance of the extracellular MRSA to which it is attached, generating highly toxic reactive oxygen species (ROS) for intracellular MRSA killing via the enzymatic activities of the FeSAs core. The FeSAs@Sa.M is far superior to FeSAs in killing intracellular MRSA, proposing a feasible strategy for treating intracellular infections by in situ generating ROS in bacterial residing regions.
    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
    旨在使用先天免疫细胞的吞噬溶酶体作为细胞内复制和存活位点来预防由病原体引起的疾病的预防性疫苗接种策略在很大程度上是无效的。这些包括结核分枝杆菌(Mtb),利什曼原虫。,和隐球菌属。这些失败的策略传统上靶向CD4+T辅助(Th)1细胞介导的免疫记忆,认为它对疫苗效力至关重要。这种失败需要对保护的替代调解人进行调查。这里,我们提出了三种在感染前或感染时激活吞噬细胞的新方法。我们假设,防止在吞噬溶酶体内形成病原体生态位对于预防疾病至关重要,更加强调吞噬细胞激活的时机应该会产生更有效的预防性治疗选择。
    Prophylactic vaccination strategies designed to prevent diseases caused by pathogens using the phagolysosome of innate immune cells as a site of intracellular replication and survival have been largely ineffective. These include Mycobacterium tuberculosis (Mtb), Leishmania spp., and Cryptococcus spp. These failed strategies have traditionally targeted CD4+ T helper (Th) 1 cell-mediated immune memory, deeming it crucial for vaccine efficacy. This failure warrants an investigation of alternative mediators of protection. Here, we suggest three novel approaches to activate phagocytic cells prior to or at the time of infection. We hypothesize that preventing the formation of the pathogen niche within the phagolysosome is essential for preventing disease, and a greater emphasis on the timing of phagocyte activation should generate more effective prophylactic treatment options.
    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
    在这项工作中,左氧氟沙星(LVX),第三代氟喹诺酮类抗生素,被封装在壳聚糖-g-聚(甲基丙烯酸甲酯)的两亲性聚合物纳米颗粒中,该壳聚糖-g-聚(甲基丙烯酸甲酯)通过自组装产生并通过与三聚磷酸钠的离子交联而物理稳定。非交联纳米颗粒的尺寸为29nm,ζ电位为+36mV,而交联对应物显示45nm和+24mV,分别。细胞的兼容性,摄取,和细胞内运输的特征在于体外在鼠肺泡巨噬细胞系MH-S和人支气管上皮细胞系BEAS-2B中。10分钟后检测到内化事件,摄取被几种内吞抑制剂抑制,表明复杂的内吞途径的参与。此外,在溶酶体区室中检测到纳米颗粒。然后,在感染金黄色葡萄球菌的MH-S和BEAS-2B细胞中评估了负载LVX的纳米制剂(50%w/w药物含量)的抗菌功效,细菌负荷降低了49%和46%,分别。相比之下,游离LVX导致8%和5%的下降,分别,在相同的感染细胞系中。最后,静脉注射斑马鱼幼虫模型表明,纳米颗粒在巨噬细胞和内皮中积累,并证明这些两亲性纳米颗粒有望靶向细胞内感染。
    In this work, levofloxacin (LVX), a third-generation fluoroquinolone antibiotic, is encapsulated within amphiphilic polymeric nanoparticles of a chitosan-g-poly(methyl methacrylate) produced by self-assembly and physically stabilized by ionotropic crosslinking with sodium tripolyphosphate. Non-crosslinked nanoparticles display a size of 29 nm and a zeta-potential of +36 mV, while the crosslinked counterparts display 45 nm and +24 mV, respectively. The cell compatibility, uptake, and intracellular trafficking are characterized in the murine alveolar macrophage cell line MH-S and the human bronchial epithelial cell line BEAS-2B in vitro. Internalization events are detected after 10 min and the uptake is inhibited by several endocytosis inhibitors, indicating the involvement of complex endocytic pathways. In addition, the nanoparticles are detected in the lysosomal compartment. Then, the antibacterial efficacy of LVX-loaded nanoformulations (50% w/w drug content) is assessed in MH-S and BEAS-2B cells infected with Staphylococcus aureus and the bacterial burden is decreased by 49% and 46%, respectively. In contrast, free LVX leads to a decrease of 8% and 5%, respectively, in the same infected cell lines. Finally, intravenous injection to a zebrafish larval model shows that the nanoparticles accumulate in macrophages and endothelium and demonstrate the promise of these amphiphilic nanoparticles to target intracellular infections.
    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
    病原体如金黄色葡萄球菌能够在许多类型的宿主细胞中存活,包括吞噬细胞如嗜中性粒细胞和巨噬细胞,从而导致细胞内感染。通过常规抗菌剂治疗细胞内感染(例如,抗生素)通常由于药物的细胞内功效低而无效。因此,非常需要能够增强细胞内抗菌剂活性的新技术。我们最近的研究表明,光化学内化(PCI)是一种有前途的方法,可以提高庆大霉素等抗生素对细胞内葡萄球菌感染的疗效。在这一章中,我们描述了旨在使用RAW264.7细胞感染模型和斑马鱼胚胎感染模型在体外和体内研究PCI-抗生素治疗对细胞内感染的潜力的方案.证明了这种方法的概念。预期该方案将促进用于与病原体的细胞内存活相关的具有临床挑战性的感染性疾病的基于PCI抗微生物的新疗法的进一步开发。
    Pathogens such as Staphylococcus aureus are able to survive in many types of host cells including phagocytes such as neutrophils and macrophages, thereby resulting in intracellular infections. Treatment of intracellular infections by conventional antimicrobials (e.g., antibiotics) is often ineffective due to low intracellular efficacy of the drugs. Thus, novel techniques which can enhance the activity of antimicrobials within cells are highly demanded. Our recent studies have shown that photochemical internalization (PCI) is a promising approach for improving the efficacy of antibiotics such as gentamicin against intracellular staphylococcal infection. In this chapter, we describe the protocols aiming to study the potential of PCI-antibiotic treatment for intracellular infections in vitro and in vivo using a RAW 264.7 cell infection model and a zebrafish embryo infection model. Proof of concept of this approach is demonstrated. The protocols are expected to prompt further development of PCI-antimicrobial based novel therapies for clinically challenging infectious diseases associated with intracellular survival of pathogens.
    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
    Recalcitrant respiratory tract infections caused by bacteria have emerged as one of the greatest health challenges worldwide. Aerosolized antimicrobial therapy is becoming increasingly attractive to combat such infections, as it allows targeted delivery of high drug concentrations to the infected organ while limiting systemic exposure. However, successful aerosolized antimicrobial therapy is still challenged by the diverse biological barriers in infected lungs. Nanoparticle-mediated pulmonary drug delivery is gaining increasing attention as a means to overcome the biological barriers and accomplish site-specific drug delivery by controlling release of the loaded drug(s) at the target site. With the aim to summarize emerging efforts in combating respiratory tract infections by using nanoparticle-mediated pulmonary delivery strategies, this review provides a brief introduction to the bacterial infection-related pulmonary diseases and the biological barriers for effective treatment of recalcitrant respiratory tract infections. This is followed by a summary of recent advances in design of inhalable nanoparticle-based drug delivery systems that overcome the biological barriers and increase drug bioavailability. Finally, challenges for the translation from exploratory laboratory research to clinical application are also discussed and potential solutions proposed.
    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
    Bacterial infections are an imminent global healthcare threat evolving from rapidly advancing bacterial defence mechanisms that antibiotics fail to overcome. Antibiotics have been designed for systemic administration to target planktonic bacteria, leading to difficulties in reaching the site of localized bacterial infection and an inability to overcome the biological, chemical and physical barriers of bacteria, including biofilms, intracellular infections and antimicrobial resistance. The amphiphilic, biomimetic and antimicrobial properties of lipids provide a promising toolbox to innovate and advance antimicrobial therapies, overcoming the barriers presented by bacteria in order to directly and effectively treat recalcitrant infections. Nanoparticulate lipid-based drug delivery systems can enhance antibiotic permeation through the chemical and physical barriers of bacterial infections, as well as fuse with bacterial cell membranes, release antibiotics in response to bacteria and act synergistically with loaded antibiotics to enhance the total antimicrobial efficacy. This review explores the barriers presented by bacterial infections that pose bio-pharmaceutical challenges to antibiotics and how different structural and functional mechanisms of lipids can enhance antimicrobial therapies. Different nanoparticulate lipid-based systems are presented as valuable drug delivery systems to advance the efficacy of antibiotics, including liposomes, liquid crystalline nanoparticles, solid lipid nanoparticles, nanostructured lipid carriers and lipid nanocarriers. In summary, liquid crystalline nanoparticles are emerging with the greatest potential for clinical applications and commercial success as an \"all-rounder\" advanced lipid-based antimicrobial therapy that overcomes the multiple biological, chemical and physical barriers of bacteria.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Exploring the structure-activity relationship (SAR) at the cationic part of arylthiazole antibiotics revealed hydrazine as an active moiety. The main objective of the study is to overcome the inherited toxicity associated with the free hydrazine. A series of hydrocarbon bridges was inserted in between the groups, to separate the two amino groups. Hence, the aminomethylpiperidine-containing analog 16 was identified as a new promising antibacterial agent with efficient antibacterial and pharmacokinetic profiles. Briefly, compound 16 outperformed vancomycin in terms of the antibacterial spectrum against vancomycin-resistant staphylococcal and enterococcal strains with minimum inhibitory concentrations (MICs) ranging from 2 to 4 μg/mL, which is a faster bactericidal mode of action, completely eradicating the high staphylococcal burden within 6-8 h, and it has a unique ability to completely clear intracellular staphylococci. In addition, the initial pharmacokinetic assessment confirmed the high metabolic stability of compound 16 (biological half-life >4 h); it had a good extravascular distribution and maintained a plasma concentration higher than the average MIC value for over 12 h. Moreover, compound 16 significantly reduced MRSA burden in an in vivo MRSA skin infection mouse experiment. These attributes collectively suggest that compound 16 is a good therapeutic candidate for invasive staphylococcal and enterococcal infections. From a mechanistic point of view, compound 16 inhibited undecaprenyl diphosphate phosphatase (UppP) with an IC50 value of 29 μM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

公众号