intracellular bacteria

细胞内细菌
  • 文章类型: Journal Article
    细胞内细菌可以在宿主细胞内繁殖并操纵其生物学特性,传统抗生素药物治疗胞内细菌的疗效受到药物积累不足的限制。与这些隐形细菌的斗争一直是一个长期的挑战。这里,使用蛋白质自组装技术制备刺激响应性乳铁蛋白(Lf)纳米颗粒系统,以递送广谱抗生素利福平(Rif)(Rif@LfNP),用于通过靶向消除细胞内细菌来增强感染治疗。与Rif@BSANP相比,Rif@LfNPs可以特异性地靶向被细菌感染的巨噬细胞,从而增加巨噬细胞内Rif的积累。随后,具有正表面电荷的Rif@LfNP进一步显示对巨噬细胞内的细菌的靶向粘附并以氧化还原响应方式快速释放Rif。结合Lf和Rif的抗菌活性,Rif@LfNPs对细胞内细菌和生物膜显示出广谱抗生素能力。因此,具有高安全性的Rif@LfNP在皮下感染的疾病模型中表现出优异的治疗效果,脓毒症,和细菌性角膜炎。一起来看,负载抗生素的Lf纳米颗粒提供了一个有希望的平台,通过靶向消除细胞内细菌来对抗病原体感染.
    Intracellular bacteria can multiply inside host cells and manipulate their biology, and the efficacy of traditional antibiotic drug therapy for intracellular bacteria is limited by inadequate drug accumulation. Fighting against these stealthy bacteria has been a long-standing challenge. Here, a system of stimuli-responsive lactoferrin (Lf) nanoparticles is prepared using protein self-assembly technology to deliver broad-spectrum antibiotic rifampicin (Rif) (Rif@Lf NPs) for enhanced infection therapy through targeted elimination of intracellular bacteria. Compared to Rif@BSA NPs, the Rif@Lf NPs can specifically target macrophages infected by bacteria, thus increasing the accumulation of Rif within macrophages. Subsequently, Rif@Lf NPs with positive surface charge further displayed targeted adherence to the bacteria within macrophages and released Rif rapidly in a redox-responsive manner. Combined with the antibacterial activities of Lf and Rif, the Rif@Lf NPs showed broad-spectrum antibiotic abilities to intracellular bacteria and biofilms. As a result, the Rif@Lf NPs with high safety exhibited excellent therapeutic efficacy in the disease models of subcutaneous infection, sepsis, and bacterial keratitis. Taken together, the antibiotic-loaded Lf nanoparticles present a promising platform to combat pathogen infections through targeted elimination of intracellular bacteria.
    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
    衣原体是同时有趣和重要的细菌。衣原体属包括许多专性细胞内生物体:它们只能在其宿主生物体的细胞内繁殖。许多,也许大多数动物都有自己特别适应的衣原体物种。在人类中,临床上最相关的物种是沙眼衣原体,作为性传播疾病的代理人特别重要。猪是猪衣原体的天然宿主,但也可能携带流产衣原体和衣原体。C.abortus和可能的C.Suis具有人畜共患病的潜力,这让它们对人类医学很有趣,但是这三个物种都给猪带来了巨大的疾病负担。基因组序列比较的最新可用性表明衣原体物种适应其各自的宿主。在细胞生物学方面,所有物种的许多方面似乎相似但不相同:细菌大多在上皮细胞内复制;它们被宿主细胞在内体中吸收,它们定制以产生胞质液泡;他们必须逃避细胞防御,必须组织营养运输到液泡;最后,它们必须组织它们的释放,以便能够感染下一个细胞或下一个宿主。似乎非常困难和具有挑战性的事情,实际上是一种非常成功的寄生方式。我将在这里尝试介绍衣原体感染生物学的一些方面,从细胞生物学到免疫防御,流行病学和预防的可能性。我将讨论猪作为宿主物种和已知感染猪的物种,但将特别利用我们对感染特别是人类的物种的更详细的知识。
    Chlamydiae are bacteria that are intriguing and important at the same time. The genus Chlamydia encompasses many species of obligate intracellular organisms: they can multiply only inside the cells of their host organism. Many, perhaps most animals have their own specifically adapted chlamydial species. In humans, the clinically most relevant species is Chlamydia trachomatis, which has particular importance as an agent of sexually transmitted disease. Pigs are the natural host of Chlamydia suis but may also carry Chlamydia abortus and Chlamydia pecorum. C. abortus and possibly C. suis have anthropozoonotic potential, which makes them interesting to human medicine, but all three species bring a substantial burden of disease to pigs. The recent availability of genomic sequence comparisons suggests adaptation of chlamydial species to their respective hosts. In cell biological terms, many aspects of all the species seem similar but non-identical: the bacteria mostly replicate within epithelial cells; they are taken up by the host cell in an endosome that they customize to generate a cytosolic vacuole; they have to evade cellular defences and have to organize nutrient transport to the vacuole; finally, they have to organize their release to be able to infect the next cell or the next host. What appears to be very difficult and challenging to achieve, is in fact a greatly successful style of parasitism. I will here attempt to cover some of the aspects of the infection biology of Chlamydia, from cell biology to immune defence, epidemiology and possibilities of prevention. I will discuss the pig as a host species and the species known to infect pigs but will in particular draw on the more detailed knowledge that we have on species that infect especially humans.
    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
    沙眼衣原体,细菌性传播感染的主要原因,通过将多种效应子(Incs[包涵膜蛋白])易位并插入包涵膜中,可以创建专门的细胞内复制性小生境。这里,我们描述IncE,一种多功能公司,在其短胞质C末端编码两个不重叠的短线性基序(SLiMs)。近端SLiM,通过模仿R-N-乙基马来酰亚胺敏感因子衔接蛋白受体(SNARE)基序的一小部分,结合并募集含突触素(STX)7-和STX12的囊泡。远端SLiM模拟分选nexin(SNX)5和SNX6货物结合位点,以招募包含SNX6的囊泡。通过同时结合两种不同的囊泡类型,IncE使这些囊泡在包涵体处彼此紧密并置,以促进沙眼衣原体细胞内发育。我们的工作表明,Incs可能已经进化出SLiM,使其能够在有限的蛋白质空间中快速进化,从而破坏宿主细胞过程。
    Chlamydia trachomatis, a leading cause of bacterial sexually transmitted infections, creates a specialized intracellular replicative niche by translocation and insertion of a diverse array of effectors (Incs [inclusion membrane proteins]) into the inclusion membrane. Here, we characterize IncE, a multifunctional Inc that encodes two non-overlapping short linear motifs (SLiMs) within its short cytosolic C terminus. The proximal SLiM, by mimicking just a small portion of an R-N-ethylmaleimide-sensitive factor adaptor protein receptor (SNARE) motif, binds and recruits syntaxin (STX)7- and STX12-containing vesicles to the inclusion. The distal SLiM mimics the sorting nexin (SNX)5 and SNX6 cargo binding site to recruit SNX6-containing vesicles to the inclusion. By simultaneously binding two distinct vesicle classes, IncE brings these vesicles in close apposition with each other at the inclusion to facilitate C. trachomatis intracellular development. Our work suggests that Incs may have evolved SLiMs to enable rapid evolution in a limited protein space to disrupt host cell processes.
    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
    Wolbachia属的特定细胞内细菌广泛存在于节肢动物和几种丝虫线虫中。媒介传播疾病的控制程序(登革热,Zika,疟疾)和抗生素的抗丝虫疗法是基于这种重要的内共生体。调查Wolbachia,然而,受到宿主细胞需求的阻碍。在这项研究中,通过qPCR表征了WolbachiawAlbB在无宿主细胞体外培养系统中生长的需求。来自白纹伊蚊C6/36昆虫细胞的细胞裂解物级分含有细胞膜和含有胎牛血清的培养基,被鉴定为Wolbachia无细胞复制所必需的。补充昆虫细胞裂解物的膜部分可使细胞外Wolbachia复制增加4.2倍。与在昆虫细胞内生长的Wolbachia相比,无昆虫细胞培养物中的复制速率较低。然而,内细菌能够复制长达12天,并感染未感染的C6/36细胞。用脂质II生物合成抑制剂磷霉素处理的无细胞Wolbachia具有扩大的表型,先前在C6/36细胞中观察到的细胞内沃尔巴克氏菌,表明细菌无法分裂。总之,我们开发了一种无细胞培养系统,其中Wolbachia复制长达12天,提供了一种体外工具来阐明这些内细菌的生物学,例如,通过使用可能不会进入C6/36细胞的化合物进行细胞分裂。更好地理解Wolbachia生物学,特别是宿主-共生体的相互作用,是在病媒控制计划中使用Wolbachia以及未来针对丝虫病的药物开发的关键。
    Obligate intracellular endobacteria of the genus Wolbachia are widespread in arthropods and several filarial nematodes. Control programs for vector-borne diseases (dengue, Zika, malaria) and anti-filarial therapy with antibiotics are based on this important endosymbiont. Investigating Wolbachia, however, is impeded by the need for host cells. In this study, the requirements for Wolbachia wAlbB growth in a host cell-free in vitro culture system were characterized via qPCRs. A cell lysate fraction from Aedes albopictus C6/36 insect cells containing cell membranes and medium with fetal bovine serum were identified as requisite for cell-free replication of Wolbachia. Supplementation with the membrane fraction of insect cell lysate increased extracellular Wolbachia replication by 4.2-fold. Replication rates in the insect cell-free culture were lower compared to Wolbachia grown inside insect cells. However, the endobacteria were able to replicate for up to 12 days and to infect uninfected C6/36 cells. Cell-free Wolbachia treated with the lipid II biosynthesis inhibitor fosfomycin had an enlarged phenotype, seen previously for intracellular Wolbachia in C6/36 cells, indicating that the bacteria were unable to divide. In conclusion, we have developed a cell-free culture system in which Wolbachia replicate for up to 12 days, providing an in vitro tool to elucidate the biology of these endobacteria, e.g., cell division by using compounds that may not enter the C6/36 cells. A better understanding of Wolbachia biology, and in particular host-symbiont interactions, is key to the use of Wolbachia in vector control programs and to future drug development against filarial diseases.
    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
    需要有效的分子策略来靶向在哺乳动物细胞内茁壮成长和增殖的致病细菌。许多治疗方法无法进入的避难所。在这里,我们提出了一类阳离子两亲性聚脯氨酸螺旋(CAPHs),其中阳离子部分在聚脯氨酸螺旋上有刚性放置,并评估了构成CAPHs的非天然脯氨酸残基的构型作用。通过缩短胍基侧链和药剂的脯氨酸骨架之间的距离,观察到细胞摄取和抗菌活性显着增加,而将吡咯烷环上部分的构型从顺式改变为反式导致更适度的增加。当评估这两种活动的组合时,更严格的CAPHs在根除巨噬细胞内的细胞内耐甲氧西林金黄色葡萄球菌(MRSA)和沙门氏菌感染方面异常有效,显著超过与父CAPH的间隙。
    Effective molecular strategies are needed to target pathogenic bacteria that thrive and proliferate within mammalian cells, a sanctuary inaccessible to many therapeutics. Herein, we present a class of cationic amphiphilic polyproline helices (CAPHs) with a rigid placement of the cationic moiety on the polyproline helix and assess the role of configuration of the unnatural proline residues making up the CAPHs. By shortening the distance between the guanidinium side chain and the proline backbone of the agents, a notable increase in cellular uptake and antibacterial activity was observed, whereas changing the configuration of the moieties on the pyrrolidine ring from cis to trans resulted in more modest increases. When the combination of these two activities was evaluated, the more rigid CAPHs were exceptionally effective at eradicating intracellular methicillin-resistant Staphylococcus aureus (MRSA) and Salmonella infections within macrophages, significantly exceeding the clearance with the parent CAPH.
    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
    伯氏柯希菌是一种专性细胞内细菌,可引起全球人畜共患疾病Q热。慢性感染的治疗选择有限,而新的治疗策略的发展需要更深入地了解伯氏梭菌是如何与免疫信号相互作用的。已知细胞死亡反应由C.burnetii操纵,但是caspase-8的作用,caspase-8是多种细胞死亡途径的核心调节因子,没有被调查。在这项研究中,我们研究了caspase-8信号传导的细菌操作和caspase-8对伯氏梭菌感染的意义,检查细菌复制,细胞死亡诱导,和细胞因子信号。我们测量了胱天蛋白酶,RIPK,和MLKL在布氏梭菌感染的肿瘤坏死因子α(TNFα)/环己酰亚胺处理的THP-1巨噬细胞样细胞和TNFα/ZVAD处理的L929细胞中的激活,以评估细胞凋亡和坏死性凋亡信号传导。此外,我们测量了C.Burnetii复制,细胞死亡,和TNFα诱导超过12天的RIPK1激酶死亡,RIPK3-激酶死亡,或RIPK3-激酶-死亡-caspase-8-/-骨髓源性巨噬细胞(BMDMs),以了解caspase-8和RIPK1/3在感染过程中的重要性。我们发现caspase-8被伯氏梭菌抑制,同时抑制细胞凋亡和增加对坏死的易感性。此外,在缺乏caspase-8的BMDMs中,芽孢杆菌的复制增加,但在缺乏RIPK1/3激酶活性的BMDMs中没有,与TNFα产生减少和细胞死亡减少相对应。由于TNFα与伯氏梭菌的控制有关,ThislackofaTNFaresponsemayallowfortheuncheckedbacterygrowthwesawincaspase-8-/-BMDMs.Thisresearchidentifiesandexpectscaspase-8asakeyregulatorofC.burnetiiinfection,打开新颖的治疗之门。
    Coxiella burnetii is an obligate intracellular bacteria that causes the global zoonotic disease Q Fever. Treatment options for chronic infection are limited, and the development of novel therapeutic strategies requires a greater understanding of how C. burnetii interacts with immune signaling. Cell death responses are known to be manipulated by C. burnetii, but the role of caspase-8, a central regulator of multiple cell death pathways, has not been investigated. In this research, we studied bacterial manipulation of caspase-8 signaling and the significance of caspase-8 to C. burnetii infection, examining bacterial replication, cell death induction, and cytokine signaling. We measured caspase, RIPK, and MLKL activation in C. burnetii-infected tumor necrosis factor alpha (TNFα)/cycloheximide-treated THP-1 macrophage-like cells and TNFα/ZVAD-treated L929 cells to assess apoptosis and necroptosis signaling. Additionally, we measured C. burnetii replication, cell death, and TNFα induction over 12 days in RIPK1-kinase-dead, RIPK3-kinase-dead, or RIPK3-kinase-dead-caspase-8-/- bone marrow-derived macrophages (BMDMs) to understand the significance of caspase-8 and RIPK1/3 during infection. We found that caspase-8 is inhibited by C. burnetii, coinciding with inhibition of apoptosis and increased susceptibility to necroptosis. Furthermore, C. burnetii replication was increased in BMDMs lacking caspase-8, but not in those lacking RIPK1/3 kinase activity, corresponding with decreased TNFα production and reduced cell death. As TNFα is associated with the control of C. burnetii, this lack of a TNFα response may allow for the unchecked bacterial growth we saw in caspase-8-/- BMDMs. This research identifies and explores caspase-8 as a key regulator of C. burnetii infection, opening novel therapeutic doors.
    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
    许多细菌病原体已经进化出有效的策略来干扰泛素化网络以逃避先天免疫系统的清除。这里,我们报道了OTUB1,哺乳动物细胞中最丰富的去泛素酶(DUB)之一,在嗜肺军团菌感染期间同时进行规范和非规范的泛素化。效应子SidC和SdcA在多个赖氨酸残基上催化OTUB1泛素化,导致其与含军团菌的液泡相关联。通过SidC和SdcA的赖氨酸泛素化促进OTUB1和DEPTOR之间的相互作用,MTORC1通路的抑制剂,从而抑制MTORC1信号。MTORC1的抑制导致宿主蛋白合成的抑制和宿主巨自噬/自噬在肺炎杆菌感染期间的促进。此外,SidE家族效应子(SidEs)的成员在Ser16和Ser18处诱导OTUB1的磷酸核糖(PR)连接的泛素化,并阻断其DUB活性。OTUB1的赖氨酸和丝氨酸泛素化的水平进一步受到具有拮抗SidC活性的效应子的调节,SdcA和侧面,包括Lem27Dupa,Dupb,SidJ和SdjA.我们的研究揭示了调节宿主DUB活性的效应子介导的复杂机制。缩写:BafA1:bafilomycinA1;BMDMs:骨髓衍生的巨噬细胞;DUB:去泛素酶;Dot/Icm:细胞器运输/细胞内繁殖缺陷;DEPTOR:含有MTOR相互作用蛋白的DEP结构域;GAPDH:甘油醛-3-磷酸脱氢酶;嗜肺军团菌:嗜肺军团菌;LCV:含军团菌复合酶1的MTUO泛素醛结合1;PR-Ub:磷酸核糖(PR)连接的泛素;PTM:翻译后修饰;SDS-PAGE:十二烷基硫酸钠-聚丙烯酰胺凝胶电泳;SidEs:SidE家族效应子;Ub:泛素。
    Many bacterial pathogens have evolved effective strategies to interfere with the ubiquitination network to evade clearance by the innate immune system. Here, we report that OTUB1, one of the most abundant deubiquitinases (DUBs) in mammalian cells, is subjected to both canonical and noncanonical ubiquitination during Legionella pneumophila infection. The effectors SidC and SdcA catalyze OTUB1 ubiquitination at multiple lysine residues, resulting in its association with a Legionella-containing vacuole. Lysine ubiquitination by SidC and SdcA promotes interactions between OTUB1 and DEPTOR, an inhibitor of the MTORC1 pathway, thus suppressing MTORC1 signaling. The inhibition of MTORC1 leads to suppression of host protein synthesis and promotion of host macroautophagy/autophagy during L. pneumophila infection. In addition, members of the SidE family effectors (SidEs) induce phosphoribosyl (PR)-linked ubiquitination of OTUB1 at Ser16 and Ser18 and block its DUB activity. The levels of the lysine and serine ubiquitination of OTUB1 are further regulated by effectors that function to antagonize the activities of SidC, SdcA and SidEs, including Lem27, DupA, DupB, SidJ and SdjA. Our study reveals an effectors-mediated complicated mechanism in regulating the activity of a host DUB.Abbreviations: BafA1: bafilomycin A1; BMDMs: bone marrow-derived macrophages; DUB: deubiquitinase; Dot/Icm: defective for organelle trafficking/intracellular multiplication; DEPTOR: DEP domain containing MTOR interacting protein; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; L. pneumophila: Legionella pneumophila; LCV: Legionella-containing vacuole; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; MTORC1: mechanistic target of rapamycin kinase complex 1; OTUB1: OTU deubiquitinase, ubiquitin aldehyde binding 1; PR-Ub: phosphoribosyl (PR)-linked ubiquitin; PTM: posttranslational modification; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; SidEs: SidE family effectors; Ub: ubiquitin.
    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
    衣原体感染和丝虫线虫引起的疾病是全球健康问题。然而,由于持续存在的衣原体和针对丝虫感染的长期治疗方案伴随着低依从性,可能导致治疗失败,因此治疗面临挑战.一种新的治疗策略可能是靶向专性细胞内细菌衣原体和Wolbachia中参与细胞分裂的减少的肽聚糖结构,后者是支持丝虫发育的义务内共生体,增长,和生存。这里,沙眼衣原体和Wolbachia的细胞培养实验表明,核苷类抗生素鼠霉素和卡巴霉素干扰细菌细胞分裂,衣原体中类似青霉素诱导的持久性表型的异常细胞。用纯化的肺炎克雷伯菌MraY进行的酶抑制实验表明,穆雷霉素衍生物消除了肽聚糖前体脂质的合成I.衣原体和wolbachialMraY与Aquifexaeolicus中相应的特征良好的酶的计算机模拟分析显示出高度的保守性,提供类似抑制模式的证据。MuraymycinD2治疗从已建立的青霉素诱导的持续性感染中根除了持续的非分裂沙眼衣原体细胞。这一发现表明核苷抗生素可能具有可以破坏细菌持久性的其他特性。
    Chlamydial infections and diseases caused by filarial nematodes are global health concerns. However, treatment presents challenges due to treatment failures potentially caused by persisting Chlamydia and long regimens against filarial infections accompanied by low compliance. A new treatment strategy could be the targeting of the reduced peptidoglycan structures involved in cell division in the obligate intracellular bacteria Chlamydia and Wolbachia, the latter being obligate endosymbionts supporting filarial development, growth, and survival. Here, cell culture experiments with C. trachomatis and Wolbachia showed that the nucleoside antibiotics muraymycin and carbacaprazamycin interfere with bacterial cell division and induce enlarged, aberrant cells resembling the penicillin-induced persistence phenotype in Chlamydia. Enzymatic inhibition experiments with purified C. pneumoniae MraY revealed that muraymycin derivatives abolish the synthesis of the peptidoglycan precursor lipid I. Comparative in silico analyses of chlamydial and wolbachial MraY with the corresponding well-characterized enzyme in Aquifex aeolicus revealed a high degree of conservation, providing evidence for a similar mode of inhibition. Muraymycin D2 treatment eradicated persisting non-dividing C. trachomatis cells from an established penicillin-induced persistent infection. This finding indicates that nucleoside antibiotics may have additional properties that can break bacterial persistence.
    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
    尽管已经探索了各种类型的基于mRNA的疫苗,诱导体液和细胞免疫的最佳条件仍然未知。在这项研究中,在小鼠中通过不同途径给药后,评估脂质复合物(LPX)或脂质纳米颗粒(LNP)中核苷修饰的mRNA疫苗,评估mRNA递送,耐受性和免疫原性。此外,我们研究了mRNA疫苗是否可以受益于包含佐剂α-半乳糖神经酰胺(αGC),一种不变的自然杀伤T(NKT)细胞配体。用αGC佐剂化的LNP中包封的编码卵清蛋白(OVA)的mRNA的肌内(IM)疫苗接种显示出最高的抗体和CD8T细胞应答。此外,我们观察到,在编码OVA的mRNA序列中添加信号肽和LAMP1或HLA-B7的内吞分选信号,虽然减少了IgG抗体应答的诱导,但进一步增强了CD8+T细胞的活化.此外,与具有SM-102的mRNALNP相比,具有可电离类脂质C12-200的mRNALNP表现出更高的促炎和反应原活性,与T细胞活化和抗肿瘤潜力增加相关。我们还观察到αGC可以进一步增强临床相关mRNALNP疫苗的细胞免疫。从而促进治疗性抗肿瘤潜力。最后,补充有αGC的单核细胞增生李斯特菌mRNALNP疫苗对李斯特菌病显示出协同保护作用,强调了在抗菌mRNA疫苗中共同激活iNKT细胞的关键优势。一起来看,我们的研究为优化针对疾病应用的mRNA疫苗的设计提供了多种见解,如癌症和细胞内细菌感染。
    Although various types of mRNA-based vaccines have been explored, the optimal conditions for induction of both humoral and cellular immunity remain rather unknown. In this study, mRNA vaccines of nucleoside-modified mRNA in lipoplexes (LPXs) or lipid nanoparticles (LNPs) were evaluated after administration in mice through different routes, assessing mRNA delivery, tolerability and immunogenicity. In addition, we investigated whether mRNA vaccines could benefit from the inclusion of the adjuvant alpha-galactosylceramide (αGC), an invariant Natural Killer T (iNKT) cell ligand. Intramuscular (IM) vaccination with ovalbumin (OVA)-encoding mRNA encapsulated in LNPs adjuvanted with αGC showed the highest antibody- and CD8+ T cell responses. Furthermore, we observed that addition of signal peptides and endocytic sorting signals of either LAMP1 or HLA-B7 in the OVA-encoding mRNA sequence further enhanced CD8+ T cell activation although reducing the induction of IgG antibody responses. Moreover, mRNA LNPs with the ionizable lipidoid C12-200 exhibited higher pro-inflammatory- and reactogenic activity compared to mRNA LNPs with SM-102, correlating with increased T cell activation and antitumor potential. We also observed that αGC could further enhance the cellular immunity of clinically relevant mRNA LNP vaccines, thereby promoting therapeutic antitumor potential. Finally, a Listeria monocytogenes mRNA LNP vaccine supplemented with αGC showed synergistic protective effects against listeriosis, highlighting a key advantage of co-activating iNKT cells in antibacterial mRNA vaccines. Taken together, our study offers multiple insights for optimizing the design of mRNA vaccines for disease applications, such as cancer and intracellular bacterial 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
    支原体是细胞内致病菌,感染多种植物物种,包括重要农业作物和观赏树。然而,我们对症状严重程度之间关系的理解,疾病进展,由于无法将植物质机械接种到新的植物宿主中,因此植物质浓度仍然有限。本研究调查了长春花和番茄的植物血浆滴度动态和症状发展,两者都使用小型幼苗嫁接方法感染了相同的马铃薯紫顶(PPT)植浆菌株。绿化,Phyllody,和巫婆扫帚(WB)症状依次出现在长春花,而在番茄植物中,大芽(BB,一种叶状),花椰菜状花序(CLI),WB出现的顺序。从定量聚合酶链反应(qPCR)靶向PPT植原体的16SrRNA基因显示,在两个宿主物种,在不同的感染阶段,植物质滴度差异显着。值得注意的是,在显示叶状症状的样本中,观察到了最高的植物血浆浓度,而在西红柿中,滴度在BB阶段达到峰值。蛋白质印迹分析,利用对PPT植物浆特异的抗体,证实在显示叶状和BB症状的样品中存在大量的植原体,与qPCR结果一致。这些发现挑战了传统的理解,即植原体感染动力学导致后期更高的滴度,如WB(过度营养生长),而不是在早期阶段,如叶形(生殖生长异常)。此外,附属物中的PPT植物浆滴度明显高于番茄植物,表明宿主之间不同的敏感性。这项研究揭示了不同的宿主反应,对PPT植原体感染,提供对植物血浆滴度动态和症状发展的有价值的见解,对未来农业疾病的管理具有重要意义。
    Phytoplasmas are intracellular pathogenic bacteria that infect a wide range of plant species, including agriculturally important crops and ornamental trees. However, our understanding of the relationship between symptom severity, disease progression, and phytoplasma concentration remains limited due to the inability to inoculate phytoplasmas mechanically into new plant hosts. The present study investigated phytoplasma titer dynamics and symptom development in periwinkle and tomato, both infected with the same potato purple top (PPT) phytoplasma strain using a small seedling grafting approach. Virescence, phyllody, and witches\'-broom (WB) symptoms sequentially developed in periwinkle, while in tomato plants, big bud (BB, a form of phyllody), cauliflower-like inflorescence (CLI), and WB appeared in order. Results from quantitative polymerase chain reaction (qPCR) targeting the PPT phytoplasma\'s 16S rRNA gene revealed that in both host species, phytoplasma titers differed significantly at different infection stages. Notably, the highest phytoplasma concentration in periwinkles was observed in samples displaying phyllody symptoms, whereas in tomatoes, the titer peaked at the BB stage. Western blot analysis, utilizing an antibody specific to PPT phytoplasma, confirmed substantial phytoplasma presence in samples displaying phyllody and BB symptoms, consistent with the qPCR results. These findings challenge the conventional understanding that phytoplasma infection dynamics result in a higher titer at later stages, such as WB (excessive vegetative growth), rather than in the early stage, such as phyllody (abnormal reproductive growth). Furthermore, the PPT phytoplasma titer was markedly higher in periwinkles than in tomato plants, indicating differing susceptibilities between the hosts. This study reveals distinct host responses to PPT phytoplasma infection, providing valuable insights into phytoplasma titer dynamics and symptom development, with implications for the future management of agricultural disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号