Chlamydia muridarum

鼠衣原体
  • 文章类型: Journal Article
    呼吸道感染是全球新生儿疾病和发病的最常见原因之一。在急性期,已知感染引起广泛的外周炎症。然而,尚未研究炎症对关键神经呼吸控制中心的影响。利用新生儿呼吸道感染的特征良好的模型,我们调查了延髓内的急性反应,延髓内包含关键的呼吸区域。新生小鼠在出生后24小时内鼻内接种,无论是鼠衣原体还是假感染,和组织收集在出生后第15天,周围炎症的高峰。这项研究的一个关键发现是,虽然外围似乎没有显示新生儿呼吸道感染的性别特异性影响,性别对延髓的炎症反应有显著影响。髓质有明显的性别特异性反应,与周围炎症的高峰一致,女性表现出抗炎细胞因子的上调,男性表现出很少的变化。小胶质细胞还表现出性别特异性,雌性和雄性的形态因细胞核而异。星形胶质细胞在对新生儿感染的急性反应中表现出有限的变化。这些数据强调了呼吸道感染在急性炎症期对髓质的强烈的性别特异性影响。
    Respiratory infections are one of the most common causes of illness and morbidity in neonates worldwide. In the acute phase infections are known to cause wide-spread peripheral inflammation. However, the inflammatory consequences to the critical neural control centres for respiration have not been explored. Utilising a well characterised model of neonatal respiratory infection, we investigated acute responses within the medulla oblongata which contains key respiratory regions. Neonatal mice were intranasally inoculated within 24 h of birth, with either Chlamydia muridarum or sham-infected, and tissue collected on postnatal day 15, the peak of peripheral inflammation. A key finding of this study is that, while the periphery appeared to show no sex-specific effects of a neonatal respiratory infection, sex had a significant impact on the inflammatory response of the medulla oblongata. There was a distinct sex-specific response in the medulla coincident with peak of peripheral inflammation, with females demonstrating an upregulation of anti-inflammatory cytokines and males showing very few changes. Microglia also demonstrated sex-specificity with the morphology of females and males differing based upon the nuclei. Astrocytes showed limited changes during the acute response to neonatal infection. These data highlight the strong sex-specific impact of a respiratory infection can have on the medulla in the acute inflammatory phase.
    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
    为了开始优化我们报道的PLGA-rMOMP纳米疫苗[PLGA包裹的衣原体(Cm)重组主要外膜蛋白(rMOMP)]的免疫途径,我们比较了两种初免-加强免疫策略(皮下(SC)和肌内(IM-p)初免途径,随后进行两次SC加强免疫),以评估纳米疫苗在雌性BALB/c小鼠中诱导的保护功效和免疫原性.我们的结果表明,通过SC和IM-p途径免疫的小鼠通过减少细菌负担和SC小鼠中的细菌减少而免受Cm生殖器攻击。与rMOMP特异性Th1(IL-2,IFN-γ)而不是Th2(IL-4,IL-9,IL-13)细胞因子相关的小鼠的保护,和CD4+记忆(CD44highCD62Lhigh)T细胞,特别是在SC小鼠中。我们还观察到更高水平的IL-1α,SC免疫小鼠中的IL-6、IL-17、CCL-2和G-CSF。值得注意的是,在SC中的攻击后观察到细胞因子/趋化因子的增加,IM-p,和对照小鼠(rMOMP和PBS),暗示厘米刺激。并行,rMOMP特异性Th1(IgG2a,IgG2b)和Th2(IgG1)血清,粘膜,血清亲和力,和中和抗体在SC比IM-p小鼠中升高。总的来说,与异源IM-p相比,小鼠的同源SC初免-加强免疫诱导增强的细胞和抗体应答,具有更好的针对生殖器攻击的保护作用。
    To begin to optimize the immunization routes for our reported PLGA-rMOMP nanovaccine [PLGA-encapsulated Chlamydia muridarum (Cm) recombinant major outer membrane protein (rMOMP)], we compared two prime-boost immunization strategies [subcutaneous (SC) and intramuscular (IM-p) prime routes followed by two SC-boosts)] to evaluate the nanovaccine-induced protective efficacy and immunogenicity in female BALB/c mice. Our results showed that mice immunized via the SC and IM-p routes were protected against a Cm genital challenge by a reduction in bacterial burden and with fewer bacteria in the SC mice. Protection of mice correlated with rMOMP-specific Th1 (IL-2 and IFN-γ) and not Th2 (IL-4, IL-9, and IL-13) cytokines, and CD4+ memory (CD44highCD62Lhigh) T-cells, especially in the SC mice. We also observed higher levels of IL-1α, IL-6, IL-17, CCL-2, and G-CSF in SC-immunized mice. Notably, an increase of cytokines/chemokines was seen after the challenge in the SC, IM-p, and control mice (rMOMP and PBS), suggesting a Cm stimulation. In parallel, rMOMP-specific Th1 (IgG2a and IgG2b) and Th2 (IgG1) serum, mucosal, serum avidity, and neutralizing antibodies were more elevated in SC than in IM-p mice. Overall, the homologous SC prime-boost immunization of mice induced enhanced cellular and antibody responses with better protection against a genital challenge compared to the heterologous IM-p.
    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
    我们以前已经证明,产生TNF-α的CD8+T细胞介导衣原体发病机制,可能以抗原(Ag)特异性方式。在这里,我们假设免疫和/或攻击后Ag特异性CD8+T细胞应答的抑制将与保护性疫苗方案诱导的针对输卵管病理的保护相关。鼻内(i.n.)活衣原体基本体(EB),肌内(i.m.)活EB,或i.n.无关抗原,牛血清白蛋白(BSA),免疫动物诱导几乎完全保护,50%保护,或者没有保护,分别针对i.vag后的输卵管病理学。c.muridarum挑战。在这些模型中,我们评估了免疫或攻击后不同时间段的Ag特异性CD8+T细胞细胞因子应答.结果显示疫苗方案的保护效力与在阴道后Ag特异性CD8+T细胞TNF-α应答的减少相关。衣原体挑战,免疫接种后没有。CD4+T细胞的耗竭被废除,而在衣原体攻击后,Ag特异性CD4+T细胞的过继转移诱导了Ag特异性CD8+T细胞TNF-α反应的显着降低。总之,保护性抗衣原体疫苗方案诱导Ag特异性CD4+T细胞应答,其在攻击后介导致病性CD8+T细胞应答的早期抑制,并且可以作为针对衣原体诱导的慢性病理的保护的预测性生物标志物。
    We have demonstrated previously that TNF-α-producing CD8+ T cells mediate chlamydial pathogenesis, likely in an antigen (Ag)-specific fashion. Here we hypothesize that inhibition of Ag-specific CD8+ T cell response after immunization and/or challenge would correlate with protection against oviduct pathology induced by a protective vaccine regimen. Intranasal (i.n.) live chlamydial elementary body (EB), intramuscular (i.m.) live EB, or i.n. irrelevant antigen, bovine serum albumin (BSA), immunized animals induced near-total protection, 50% protection, or no protection, respectively against oviduct pathology following i.vag. C. muridarum challenge. In these models, we evaluated Ag-specific CD8+ T cell cytokine response at various time-periods after immunization or challenge. The results show protective efficacy of vaccine regimens correlated with reduction of Ag-specific CD8+ T cell TNF-α responses following i.vag. chlamydial challenge, not after immunization. Depletion of CD4+ T cells abrogated, whereas adoptive transfer of Ag-specific CD4+ T cells induced the significant reduction of Ag-specific CD8+ T cell TNF-α response after chlamydial challenge. In conclusion, protective anti-chlamydial vaccine regimens induce Ag-specific CD4+ T cell response that mediate early inhibition of pathogenic CD8+ T cell response following challenge and may serve as a predictive biomarker of protection against Chlamydia -induced chronic pathologies.
    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
    鼠衣原体(Cm),一种具有历史重要性的胞内细菌,最近被重新发现在研究小鼠殖民地中中等流行。大约80年前,Cm首次被报道为小鼠重症肺炎的病原体,虽然它已被实验用于模拟人类沙眼衣原体感染,目前尚无与自然感染相关的临床疾病的进一步报道.我们观察了2个基因工程小鼠(GEM)品系的临床疾病和病理,Il12rb2KO和STAT1KO,在已知被Cm定植和脱落的各种GEM菌株的集落中,干扰素-γ信号传导和Th1CD4T细胞反应受损。临床症状包括病情不佳,驼背的姿势,和可怜的繁殖力。组织病理学显示播散的Cm,肺部有病变,胃肠,和泌尿生殖组织。使用针对Cm主要外膜蛋白-1抗原的免疫组织化学和使用针对Cm菌株Nigg的选择区域的靶探针的原位杂交来确认Cm的存在。还发现Cm与一只小鼠的尿路上皮乳头状瘤有关。这些病例为从研究小鼠菌落中排除Cm提供了额外的支持。
    Chlamydia muridarum (Cm), an intracellular bacterium of historical importance, was recently rediscovered as moderately prevalent in research mouse colonies. Cm was first reported as a causative agent of severe pneumonia in mice about 80 y ago, and while it has been used experimentally to model Chlamydia trachomatis infection of humans, there have been no further reports of clinical disease associated with natural infection. We observed clinical disease and pathology in 2 genetically engi- neered mouse (GEM) strains, Il12rb2 KO and STAT1 KO, with impaired interferon-γ signaling and Th1 CD4+ T cell responses in a colony of various GEM strains known to be colonized with and shedding Cm. Clinical signs included poor condition, hunched posture, and poor fecundity. Histopathology revealed disseminated Cm with lesions in pulmonary, gastrointestinal, and urogenital tissues. The presence of Cm was confirmed using both immunohistochemistry for Cm major outer membrane protein-1 antigen and in situ hybridization using a target probe directed against select regions of Cm strain Nigg. Cm was also found in association with a urothelial papilloma in one mouse. These cases provide additional support for excluding Cm from research mouse colonies.
    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
    衣原体是专性的细胞内细菌病原体,其可通过诱导破坏性宿主免疫应答而引起生殖器病理学。人类适应沙眼衣原体在人类宿主中引起炎症性疾病,但在小鼠中很容易被清除,和小鼠适应的鼠衣原体在鼠宿主中建立了生产性和致病性感染。虽然已经在小鼠和人类中发现了许多抗衣原体宿主抗性因子,对于独立于宿主抵抗力促进宿主适应性的宿主因素知之甚少。这里,我们表明,干扰素诱导的免疫相关的GTP酶M(Irgm)蛋白的功能作为这样的宿主因子改善感染相关的后遗症在小鼠女性生殖道,从而表征Irgm蛋白作为疾病耐受性的介质。具体来说,我们证明了所有三种鼠Irgm同源物(pan-Irgm-/-)缺陷的小鼠对沙眼衣原体的细胞自主免疫缺陷,这与细菌负荷的早期和短暂增加以及体内持续的炎症过度相关。相比之下,在用C.muridarum感染pan-Irgm-/-小鼠后,细菌负担不受影响,然而,生殖器炎症和疤痕病理仍在增加,证明Irgm蛋白可以在不改变细菌负担的情况下促进宿主的健康。此外,泛Irgm-/-小鼠在生殖器衣原体感染中显示肉芽肿性炎症增加,提示Irgm蛋白在肉芽肿形成和维持的调节中。这些发现表明,Irgm蛋白在体内调节对衣原体感染的致病性免疫反应,建立有效的感染模型来检测Irgm蛋白的免疫调节功能和机制。
    目的:针对生殖器衣原体感染,免疫系统启动促炎反应来抵抗病原体,然而,必须严格控制炎症,以避免对宿主生殖器组织的附带损害和疤痕。人IRGM基因的变异与自身炎性疾病的易感性有关,但其在改善由感染引起的炎性疾病中的作用尚不清楚。这里,我们使用缺乏所有三种鼠Irgm同源物的小鼠来证明Irgm蛋白不仅提供宿主对衣原体感染的抗性,而且限制女性生殖道的相关炎症。特别是,我们发现鼠Irgm表达可以预防肉芽肿性炎症,与人IRGM变异相关的炎性疾病平行。因此,我们的发现将生殖器衣原体感染确立为研究Irgm蛋白在促进保护性免疫和限制致病性炎症中的作用的有用模型。
    Chlamydiae are obligate intracellular bacterial pathogens that may cause genital pathology via induction of destructive host immune responses. Human-adapted Chlamydia trachomatis causes inflammatory disease in human hosts but is easily cleared in mice, and mouse-adapted Chlamydia muridarum establishes a productive and pathogenic infection in murine hosts. While numerous anti-chlamydial host resistance factors have been discovered in mice and humans alike, little is known about host factors promoting host fitness independent of host resistance. Here, we show that interferon-inducible immunity-related GTPase M (Irgm) proteins function as such host factors ameliorating infection-associated sequalae in the murine female genital tract, thus characterizing Irgm proteins as mediators of disease tolerance. Specifically, we demonstrate that mice deficient for all three murine Irgm paralogs (pan-Irgm-/-) are defective for cell-autonomous immunity to C. trachomatis, which correlates with an early and transient increase in bacterial burden and sustained hyperinflammation in vivo. In contrast, upon infection of pan-Irgm-/- mice with C. muridarum, bacterial burden is unaffected, yet genital inflammation and scarring pathology are nonetheless increased, demonstrating that Irgm proteins can promote host fitness without altering bacterial burden. Additionally, pan-Irgm-/- mice display increased granulomatous inflammation in genital Chlamydia infection, implicating Irgm proteins in the regulation of granuloma formation and maintenance. These findings demonstrate that Irgm proteins regulate pathogenic immune responses to Chlamydia infection in vivo, establishing an effective infection model to examine the immunoregulatory functions and mechanisms of Irgm proteins.
    OBJECTIVE: In response to genital Chlamydia infection, the immune system mounts a proinflammatory response to resist the pathogen, yet inflammation must be tightly controlled to avoid collateral damage and scarring to host genital tissue. Variation in the human IRGM gene is associated with susceptibility to autoinflammatory diseases but its role in ameliorating inflammatory diseases caused by infections is poorly defined. Here, we use mice deficient for all three murine Irgm paralogs to demonstrate that Irgm proteins not only provide host resistance to Chlamydia infections but also limit associated inflammation in the female genital tract. In particular, we find that murine Irgm expression prevents granulomatous inflammation, which parallels inflammatory diseases associated with variants in human IRGM. Our findings therefore establish genital Chlamydia infection as a useful model to study the roles for Irgm proteins in both promoting protective immunity and limiting pathogenic inflammation.
    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
    衣原体疫苗方法渴望诱导Th1细胞以获得最佳保护,尽管没有直接证据表明Th1介导的衣原体从女性生殖道(FRT)中清除。我们最近报道,T-bet缺陷小鼠可以正常解决原发性衣原体感染,破坏Th1细胞在衣原体免疫中的潜在保护作用。这里,我们表明,T-bet缺陷小鼠发展强大的Th17反应和小鼠Th17细胞缺陷表现出延迟的细菌清除,证明衣原体特异性Th17细胞代表了被低估的保护性群体。此外,Th2缺陷小鼠能有效清除宫颈阴道感染。此外,我们表明,非造血细胞对IFN-γ的感知对于衣原体免疫是必不可少的,然而,FRT中的细菌清除不需要CD4T细胞分泌IFN-γ。尽管Th1细胞不是衣原体清除所必需的,对衣原体的保护性免疫仍然依赖于MHCII类限制性CD4T细胞和IL-12p40。一起,这些数据表明IL-12p40依赖性CD4效应成熟对衣原体免疫至关重要,和Th17细胞在较小程度上,然而Th1和Th2细胞的发育都不重要。未来的衣原体疫苗接种工作将更有效,如果他们专注于诱导这种保护性CD4T细胞群体。
    Chlamydia vaccine approaches aspire to induce Th1 cells for optimal protection, despite the fact that there is no direct evidence demonstrating Th1-mediated Chlamydia clearance from the female reproductive tract (FRT). We recently reported that T-bet-deficient mice can resolve primary Chlamydia infection normally, undermining the potentially protective role of Th1 cells in Chlamydia immunity. Here, we show that T-bet-deficient mice develop robust Th17 responses and that mice deficient in Th17 cells exhibit delayed bacterial clearance, demonstrating that Chlamydia-specific Th17 cells represent an underappreciated protective population. Additionally, Th2-deficient mice competently clear cervicovaginal infection. Furthermore, we show that sensing of IFN-γ by non-hematopoietic cells is essential for Chlamydia immunity, yet bacterial clearance in the FRT does not require IFN-γ secretion by CD4 T cells. Despite the fact that Th1 cells are not necessary for Chlamydia clearance, protective immunity to Chlamydia is still dependent on MHC class-II-restricted CD4 T cells and IL-12p40. Together, these data point to IL-12p40-dependent CD4 effector maturation as essential for Chlamydia immunity, and Th17 cells to a lesser extent, yet neither Th1 nor Th2 cell development is critical. Future Chlamydia vaccination efforts will be more effective if they focus on induction of this protective CD4 T cell population.
    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
    口服接种后,鼠衣原体下降到小鼠大肠进行持久定植。然而,由于工程化的提前终止密码子(称为CMpGP3S)而缺乏质粒编码的蛋白pGP3的突变型muridarum即使在空肠内接种后也未能这样做。这是因为CD4+T细胞依赖性免疫阻止了CMpGP3S从小肠向大肠的扩散。在目前的研究中,我们发现缺乏IL-22(IL-22-/-)的小鼠在空肠接种后第3天允许CMpGP3S从小肠扩散到大肠,表明IL-22在调节衣原体传播中的关键作用。由于IL-22-/-小鼠被拯救以阻断来自C57BL/6J小鼠的供体CD4+T细胞的CMpGP3S扩散,因此有责任的IL-22由CD4+T细胞产生。始终如一,缺乏IL-22的CD4+T细胞未能阻断CMpGP3S在Rag2-/-小鼠中的传播,而IL-22感受态CD4+T细胞确实阻断。此外,缺乏cathelicidin相关抗菌肽(CRAMP)的小鼠允许CMpGP3S传播,但是来自CRAMP-/-小鼠的供体CD4+T细胞仍然足以防止CMpGP3S在Rag2-/-小鼠中传播,表明CRAMP在调节衣原体扩散中的关键作用,并且负责的CRAMP不是由CD4+T细胞产生的。因此,产生IL-22的CD4+T细胞对衣原体铺展的依赖性调节与非CD4+T细胞产生的CRAMP相关。这些发现为进一步表征负责调节肠道中细菌扩散的CD4+T细胞亚群提供了平台。
    Following an oral inoculation, Chlamydia muridarum descends to the mouse large intestine for long-lasting colonization. However, a mutant C. muridarum that lacks the plasmid-encoded protein pGP3 due to an engineered premature stop codon (designated as CMpGP3S) failed to do so even following an intrajejunal inoculation. This was because a CD4+ T cell-dependent immunity prevented the spread of CMpGP3S from the small intestine to the large intestine. In the current study, we found that mice deficient in IL-22 (IL-22-/-) allowed CMpGP3S to spread from the small intestine to the large intestine on day 3 after intrajejunal inoculation, indicating a critical role of IL-22 in regulating the chlamydial spread. The responsible IL-22 is produced by CD4+ T cells since IL-22-/- mice were rescued to block the CMpGP3S spread by donor CD4+ T cells from C57BL/6J mice. Consistently, CD4+ T cells lacking IL-22 failed to block the spread of CMpGP3S in Rag2-/- mice, while IL-22-competent CD4+ T cells did block. Furthermore, mice deficient in cathelicidin-related antimicrobial peptide (CRAMP) permitted the CMpGP3S spread, but donor CD4+ T cells from CRAMP-/- mice were still sufficient for preventing the CMpGP3S spread in Rag2-/- mice, indicating a critical role of CRAMP in regulating chlamydial spreading, and the responsible CRAMP is not produced by CD4+ T cells. Thus, the IL-22-producing CD4+ T cell-dependent regulation of chlamydial spreading correlated with CRAMP produced by non-CD4+ T cells. These findings provide a platform for further characterizing the subset(s) of CD4+ T cells responsible for regulating bacterial spreading in the intestine.
    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
    为了寻找亚单位疫苗候选物,在三种不同的免疫方案下,在小鼠生殖道感染模型中评估了在人类中鉴定的免疫原性衣原体抗原对感染和病理的保护作用。肌内免疫方案首次用于评估106个衣原体抗原,这表明,两种抗原显着减少,而11增加生殖器衣原体负担。两种减少感染的抗原未能预防病理学,另外23种抗原甚至加剧了病理学。因此,接下来测试鼻内粘膜免疫,因为鼻内接种活的鼠衣原体可预防生殖器感染和病理。在评估的29种衣原体抗原中,有2种被发现可以预防生殖器感染,但不能预防病理学,还有3种加剧病理学。进一步提高保护功效,测试了组合方案(鼻内引发+肌内增强+第三次腹膜内/皮下增强)。该方案确定了四种减少感染的抗原,但其中只有一个能阻止病理学.不幸的是,这种保护性抗原由于其与几种人类分子的氨基酸序列同源性而没有进一步发展。还发现了两种病理加重抗原。然而,在对照组中,用活的C.muridarum进行鼻内粘膜引发可始终预防生殖器感染和病理,而与随后的助推器无关。因此,筛选140种不同的衣原体抗原,在17个实验中重复21次,未能鉴定亚单位疫苗候选物,但证明了活衣原体在诱导针对生殖器感染和病理学的免疫力方面的优越性。为开发减毒衣原体活疫苗奠定了基础。
    To search for subunit vaccine candidates, immunogenic chlamydial antigens identified in humans were evaluated for protection against both infection and pathology in a mouse genital tract infection model under three different immunization regimens. The intramuscular immunization regimen was first used to evaluate 106 chlamydial antigens, which revealed that two antigens significantly reduced while 11 increased genital chlamydial burden. The two infection-reducing antigens failed to prevent pathology and 23 additional antigens even exacerbated pathology. Thus, intranasal mucosal immunization was tested next since intranasal inoculation with live Chlamydia muridarum prevented both genital infection and pathology. Two of the 29 chlamydial antigens evaluated were found to prevent genital infection but not pathology and three exacerbate pathology. To further improve protection efficacy, a combinational regimen (intranasal priming + intramuscular boosting + a third intraperitoneal/subcutaneous boost) was tested. This regimen identified four infection-reducing antigens, but only one of them prevented pathology. Unfortunately, this protective antigen was not advanced further due to its amino acid sequence homology with several human molecules. Two pathology-exacerbating antigens were also found. Nevertheless, intranasal mucosal priming with viable C. muridarum in control groups consistently prevented both genital infection and pathology regardless of the subsequent boosters. Thus, screening 140 different chlamydial antigens with 21 repeated multiple times in 17 experiments failed to identify a subunit vaccine candidate but demonstrated the superiority of viable chlamydial organisms in inducing immunity against both genital infection and pathology, laying the foundation for developing a live-attenuated Chlamydia vaccine.
    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
    衣原体的IFNγ敏感突变体在生殖道中的致病性减弱,最近被许可作为细胞内口服疫苗载体或intrOv。IntrOv的口服递送诱导生殖道的经粘膜保护,但是intrOv本身通过来自第3组样先天淋巴细胞(ILC3s)的IFNγ从肠道中清除(不从外部脱落任何感染性颗粒)。我们在当前研究中进一步表征了与ILC3的IntrOv相互作用,因为相互作用可能会影响intrOv作为口服衣原体疫苗的安全性和有效性。结肠内接种intrOv诱导的IFNγ反过来抑制intrOv。Ov-IFNγ内相互作用依赖于RORγt,ILC3s的签名转录因子。始终如一,口服intrOv诱导的ILC3s从RORγt-GFP报告小鼠转移到IFNγ缺陷小鼠挽救了intrOv的抑制。因此,由intrOv诱导的ILC3产生的IFNγ可能负责抑制intrOv,口服intrOv确实诱导显着水平的产生IFNγ的LC3s(IFNγILC3s)的观察结果进一步支持了这一点。有趣的是,IL-23受体敲除(IL-23R-/-)小鼠不再抑制体内Ov,伴有结肠IFNγ降低。口服intrOv诱导的ILC3s的转移拯救了IL-23R-/-小鼠抑制intrOv,验证ILC3s对IL-23R信号传导的依赖性以抑制IntrOv。显然,IntrOv诱导肠道IFNγ+ILC3s在肠道中自身的抑制作用,这是由IL-23R信号促进。这些发现为确保intrOv作为口服衣原体疫苗的安全性提供了机制,并为研究口服intrOv如何诱导生殖道的经粘膜保护提供了平台。
    An IFNγ-susceptible mutant of Chlamydia muridarum is attenuated in pathogenicity in the genital tract and was recently licensed as an intracellular Oral vaccine vector or intrOv. Oral delivery of intrOv induces transmucosal protection in the genital tract, but intrOv itself is cleared from the gut (without shedding any infectious particles externally) by IFNγ from group 3-like innate lymphoid cells (ILC3s). We further characterized the intrOv interactions with ILC3s in the current study, since the interactions may impact both the safety and efficacy of intrOv as an oral Chlamydia vaccine. Intracolonic inoculation with intrOv induced IFNγ that in return inhibited intrOv. The intrOv-IFNγ interactions were dependent on RORγt, a signature transcriptional factor of ILC3s. Consistently, the transfer of oral intrOv-induced ILC3s from RORγt-GFP reporter mice to IFNγ-deficient mice rescued the inhibition of intrOv. Thus, IFNγ produced by intrOv-induced ILC3s is likely responsible for inhibiting intrOv, which is further supported by the observation that oral intrOv did induce significant levels of IFNγ-producing LC3s (IFNγ+ILC3s). Interestingly, IL-23 receptor knockout (IL-23R-/-) mice no longer inhibited intrOv, which was accompanied by reduced colonic IFNγ. Transfer of oral intrOv-induced ILC3s rescued the IL-23R-/- mice to inhibit intrOv, validating the dependence of ILC3s on IL-23R signaling for inhibiting intrOv. Clearly, intrOv induces intestinal IFNγ+ILC3s for its own inhibition in the gut, which is facilitated by IL-23R signaling. These findings have provided a mechanism for ensuring the safety of intrOv as an oral Chlamydia vaccine and a platform for investigating how oral intrOv induces transmucosal protection in the genital tract.
    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
    白细胞介素-21及其受体(IL-21/IL-21R)加重衣原体肺部感染,而巨噬细胞(Mφ)是衣原体感染的主要细胞之一,也是炎性细胞因子的主要来源。因此,研究IL-21/IL-21R是否通过调节Mφ加重衣原体呼吸道感染尤为重要。结合生物信息学分析,我们建立了小鼠衣原体IL-21R缺陷(IL-21R-/-)模型(C.muridarum)体内呼吸道感染,通过在体外添加rmIL-21研究了鼠尾草刺激的RAW264.7,并进行过继转移实验,以阐明IL-21/IL-21R与Mφ之间的关联。IL-21R-/-小鼠肺总Mφ浸润降低,肺泡巨噬细胞,和间质巨噬细胞与感染后的WT小鼠相比。转录组分析表明,M1相关基因在IL-21R-/-小鼠中下调,并且IL-21R缺陷会影响Mφ介导的炎症反应。体内实验证实,在IL-21R-/-小鼠中,肺M1型CD80+,CD86+,MHCII+,TNFα+,iNOS+Mφ下降,虽然M2型CD206+没有差异,TGF-β+,IL-10+和ARG1+Mφ。体外,rmIL-21对小鼠C.muridarum刺激的RAW264.7细胞的给药促进iNOS-NO的水平以及IL-12p40和TNFα的表达,但对TGFβ或IL-10无影响。Further,来自IL-21R-/-小鼠的M1样骨髓源性巨噬细胞的过继转移,与WT小鼠不同,有效地保护受者免受小鼠C.muridarum感染,并引起肺部病理缓解。这些发现有助于理解IL-21/IL-21R通过促进Mφ的促炎作用而加剧衣原体呼吸道感染的机制。
    Interleukin-21 and its receptors (IL-21/IL-21R) aggravate chlamydial lung infection, while macrophages (Mφ) are one of the main cells infected by chlamydia and the main source of inflammatory cytokines. Therefore, it is particularly important to study whether IL-21/IL-21R aggravates chlamydia respiratory infection by regulating Mφ. Combined with bioinformatics analysis, we established an IL-21R-deficient (IL-21R-/-) mouse model of Chlamydia muridarum (C. muridarum) respiratory tract infection in vivo, studied C. muridarum-stimulated RAW264.7 by the addition of rmIL-21 in vitro, and conducted adoptive transfer experiments to clarify the association between IL-21/IL-21R and Mφ. IL-21R-/- mice showed lower infiltration of pulmonary total Mφ, alveolar macrophages, and interstitial macrophages compared with WT mice following infection. Transcriptomic analysis suggested that M1-related genes are downregulated in IL-21R-/- mice and that IL-21R deficiency affects the Mφ-mediated inflammatory response during C. muridarum infection. In vivo experiments verified that in IL-21R-/- mice, pulmonary M1-type CD80+, CD86+, MHC II+, TNFα+, and iNOS+ Mφ decreased, while there were no differences in M2-type CD206+, TGF-β+, IL-10+ and ARG1+ Mφ. In vitro, administration of rmIL-21 to C. muridarum-stimulated RAW264.7 cells promoted the levels of iNOS-NO and the expression of IL-12p40 and TNFα, but had no effect on TGFβ or IL-10. Further, adoptive transfer of M1-like bone marrow-derived macrophages derived from IL-21R-/- mice, unlike those from WT mice, effectively protected the recipients against C. muridarum infection and induced relieved pulmonary pathology. These findings help in understanding the mechanism by which IL-21/IL-21R exacerbates chlamydia respiratory infection by promoting the proinflammatory effect of Mφ.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号