Chlamydia muridarum

鼠衣原体
  • 文章类型: Journal Article
    Pgp3亚单位疫苗可引发针对沙眼衣原体感染的免疫保护,但仍需要额外的佐剂来增强其免疫保护功效。鞭毛蛋白可以选择性地刺激免疫并充当佐剂。在这项研究中,成功表达并纯化了FliC-Pgp3重组体。用FliC-Pgp3疫苗在Balb/C小鼠中进行三免疫诱导快速和持续的生发中心B细胞反应和Tfh分化,促进显著高于Pgp3组的IgG抗体滴度。FliC-Pgp3免疫主要诱导Th1型细胞免疫,导致更高水平的IFN-γ,TNF-α,和IL-2分泌的CD4+T细胞比接种Pgp3的小鼠。鼠衣原体攻击结果表明,FliC-Pgp3疫苗接种的小鼠在下生殖道中表现出更快的清除鼠衣原体定植,确保较低的输卵管积水率和累积评分。组织学分析显示,与PBS和Pgp3对照相比,FliC-Pgp3接种的小鼠的输卵管和子宫角中的扩张和炎性浸润减少。重要的是,FliC-Pgp3三免疫有效激活CD4+T细胞和树突状细胞,正如收养转移所证实的那样,导致受体小鼠更好的免疫保护。总之,新型FliC-Pgp3嵌合蛋白有望成为一种新型疫苗,具有改善的抗鼠衣原体免疫保护作用.
    The Pgp3 subunit vaccine elicits immune protection against Chlamydia trachomatis infection, but additional adjuvants are still required to enhance its immunoprotective efficacy. Flagellin can selectively stimulate immunity and act as an adjuvant. In this research, the FliC-Pgp3 recombinant was successfully expressed and purified. Tri-immunization with the FliC-Pgp3 vaccine in Balb/C mice induced rapid and persistent germinal center B-cell response and Tfh differentiation, promoting a significantly higher IgG antibody titer compared to the Pgp3 group. FliC-Pgp3 immunization primarily induced Th1-type cellular immunity, leading to higher levels of IFN-γ, TNF-α, and IL-2 secreted by CD4+ T cells than in Pgp3-vaccinated mice. Chlamydia muridarum challenge results showed that FliC-Pgp3-vaccinated mice exhibited more rapid clearance of Chlamydia muridarum colonization in the lower genital tract, ensuring a lower hydrosalpinx rate and cumulative score. Histological analysis showed reduced dilation and inflammatory infiltration in the oviduct and uterine horn of FliC-Pgp3-vaccinated mice compared to the PBS and Pgp3 control. Importantly, tri-immunization with FliC-Pgp3 effectively activated CD4+ T cells and dendritic cells, as confirmed by the adoptive transfer, resulting in better immune protection in recipient mice. In summary, the novel FliC-Pgp3 chimeric is hoped to be a novel vaccine with improved immunoprotection against Chlamydia muridarum.
    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
    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)

  • 文章类型: Journal Article
    质粒编码蛋白Pgp3在衣原体发病机制中的关键作用已在各种动物模型中得到证实。先前的研究表明,Pgp3缺陷型C.muridarum突变体在小鼠阴道接种后无法诱导输卵管积水。沙眼衣原体Pgp3三聚体的结构分析表明,Trp234可能在三聚体晶体堆积相互作用中起关键作用,并且Tyr197参与主要的阳离子结合位点。在这项研究中,我们构建了具有Pgp3,Trp234或Tyr197点突变(Pgp3W234A和Pgp3Y197A)的C.muridarum转化体。感染Pgp3W234A突变体的C3H/HeJ小鼠未能在输卵管组织中诱导严重的输卵管积水,这在很大程度上表现了全长Pgp3缺陷的C.muridarum。Pgp3Y197A变体诱导中等严重程度的病理学。Pgp3W234A突变体引起的致病性减弱可能是由于其在小鼠下生殖道的存活率降低,减少了对输卵管的提升,和轻度诱导输卵管组织中炎性细胞浸润。因此,我们的结果表明,一个重要的氨基酸残基参与Pgp3的毒力,为衣原体感染提供潜在的治疗靶点。
    The crucial role of plasmid-encoded protein Pgp3 in Chlamydia pathogenesis has been demonstrated in various animal models. Previous studies have revealed that the Pgp3-deficient C. muridarum mutant fails to induce hydrosalpinx after vaginal inoculation in mice. Structural analysis of C. trachomatis Pgp3 trimer has indicated that Trp234 may play a critical role in trimeric crystal packing interactions and that Tyr197 is involved at predominant cation-binding sites. In this study, we constructed C. muridarum transformants harboring Pgp3, Trp234, or Tyr197 point mutations (Pgp3W234A and Pgp3Y197A). C3H/HeJ mice infected with Pgp3W234A mutant failed to induce severe hydrosalpinx in the oviduct tissue, which largely phenocopied the full-length Pgp3-deficient C. muridarum. The Pgp3Y197A variant induced an intermediate severity of pathology. The attenuated pathogenicity caused by the Pgp3W234A mutant may be due to its decreased survival in the lower genital tracts of mice, reduced ascension to the oviduct, and milder induction of inflammatory cell infiltration in the oviduct tissue. Thus, our results point to an important amino acid residue involved in Pgp3 virulence, providing a potential therapeutic target for chlamydial 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
    暂无摘要。
    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
    沙眼衣原体是导致不孕症的最常见的性感染之一,其生殖器感染引起输卵管粘连和输卵管积水。小鼠阴道内衣原体感染可引起上生殖道输卵管积水,已用于研究沙眼衣原体的致病性。已知DBA2/J品系小鼠对输卵管积水的衣原体诱导具有抗性。在这项研究中,我们利用DBA2/J小鼠的这一特征来评估抗生素诱导的菌群失调在衣原体致病性中的作用。口服抗生素(万古霉素和庆大霉素)以诱导DBA2/J小鼠肠道中的生态失调。评估具有或不具有抗生素治疗的小鼠的肠和生殖器菌群失调,然后由鼠梭菌阴道内攻击。测试衣原体负荷并评估生殖器病理。我们发现口服抗生素可显着增强生殖器输卵管积水的衣原体诱导。抗生素治疗引起了胃肠道严重的菌群失调,包括显着减少的粪便DNA和增加的硬体比细菌的比率。口服抗生素不会改变小鼠生殖道的衣原体感染或微生物群。我们的研究表明,口服抗生素增强的输卵管积水与肠道生态失调相关,为肠道微生物组与衣原体生殖器致病性相关提供证据。
    Chlamydia trachomatis is one of the most common sexually infections that cause infertility, and its genital infection induces tubal adhesion and hydrosalpinx. Intravaginal Chlamydia muridarum infection in mice can induce hydrosalpinx in the upper genital tract and it has been used for studying C. trachomatis pathogenicity. DBA2/J strain mice were known to be resistant to the chlamydial induction of hydrosalpinx. In this study, we took advantage of this feature of DBA2/J mice to evaluate the role of antibiotic induced dysbiosis in chlamydial pathogenicity. Antibiotics (vancomycin and gentamicin) were orally administrated to induce dysbiosis in the gut of DBA2/J mice. The mice with or without antibiotic treatment were evaluated for gut and genital dysbiosis and then intravaginally challenged by C. muridarum. Chlamydial burden was tested and genital pathologies were evaluated. We found that oral antibiotics significantly enhanced chlamydial induction of genital hydrosalpinx. And the antibiotic treatment induced severe dysbiosis in the GI tract, including significantly reduced fecal DNA and increased ratios of firmicutes over bacteroidetes. The oral antibiotic did not alter chlamydial infection or microbiota in the mouse genital tracts. Our study showed that the oral antibiotics-enhanced hydrosalpinx correlated with dysbiosis in gut, providing the evidence for associating gut microbiome with chlamydial genital pathogenicity.
    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
    衣原体已被用于研究衣原体的发病机理,因为它诱导小鼠发生输卵管积水,在沙眼衣原体感染的妇女中观察到的病理学。我们鉴定了不再能够诱导输卵管积水的C.muridarum突变体。在目前的研究中,我们评估了该突变体作为减毒疫苗的效果.用突变体阴道内免疫后,保护小鼠免受野生型C.muridarum诱导的输卵管积水。然而,该突变体本身有效地定植于小鼠生殖道,并在阴道拭子中产生感染性生物。然而,口服接种后,该突变体未能在直肠拭子中产生感染性脱落。重要的是,口服接种了突变的经粘膜免疫的小鼠,以抵抗生殖道中野生型C.muridarum的攻击感染。早在生殖器攻击感染后第3天就检测到了保护作用,并保护口服免疫的小鼠免受上生殖道的任何重大病理影响。然而,相同的口服免疫小鼠未能防止野生型鼠尾草在胃肠道中的定植。在气道中进一步验证了口服突变体诱导的跨粘膜免疫。口服接种的小鼠被保护免受肺部感染和由鼻内接种的野生型小鼠C.muridarum引起的全身毒性,尽管相同的小鼠仍然允许野生型C.muridarum的胃肠道定植。这些观察表明,突变型鼠类C.muridarum可以被开发成胞内口服疫苗载体(或IntrOv),用于在肠外组织中选择性诱导透粘膜免疫。
    Chlamydia muridarum has been used to study chlamydial pathogenesis because it induces mice to develop hydrosalpinx, a pathology observed in C. trachomatis-infected women. We identified a C. muridarum mutant that is no longer able to induce hydrosalpinx. In the current study, we evaluated the mutant as an attenuated vaccine. Following an intravaginal immunization with the mutant, mice were protected from hydrosalpinx induced by wild-type C. muridarum. However, the mutant itself productively colonized the mouse genital tract and produced infectious organisms in vaginal swabs. Nevertheless, the mutant failed to produce infectious shedding in the rectal swabs following an oral inoculation. Importantly, mice orally inoculated with the mutant mounted transmucosal immunity against challenge infection of wild-type C. muridarum in the genital tract. The protection was detected as early as day 3 following the genital challenge infection and the orally immunized mice were protected from any significant pathology in the upper genital tract. However, the same orally immunized mice failed to prevent the colonization of wild-type C. muridarum in the gastrointestinal tract. The transmucosal immunity induced by the oral mutant was further validated in the airway. The orally vaccinated mice were protected from both lung infection and systemic toxicity caused by intranasally inoculated wild-type C. muridarum although the same mice still permitted the gastrointestinal colonization by the wild-type C. muridarum. These observations suggest that the mutant C. muridarum may be developed into an intracellular oral vaccine vector (or IntrOv) for selectively inducing transmucosal immunity in extra-gut tissues.
    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
    Chlamydia trachomatis ( CT ) genital tract infection is insidious, and patients often have no conscious symptoms.Delayed treatment after infection can lead to serious complications. Chlamydia muridarum ( CM ) genital tract infection in female mice can simulate CT genital tract infection in women, which is an ideal model to investigate the pathogenesis of CT . CM plasmid protein pGP3, chromosomal protein TC0237/TC0668, CM -specific CD8 + T cells, TNF-α, and IL-13 can induce genital tract inflammation, CD4 + T cells are responsible for CM clearance. However, tubal inflammation persists after genital tract CM is removed. Genital tract CM can spread spontaneously in vivo and colonize the gastrointestinal (GI) tract, but the GI tract CM cannot reverse spread to the genital tract. The survival time and number of CM transmitted from genital tract to GI tract are positively correlated with the long-term lesion of oviduct, while the CM inoculated directly into the GI tract has no pathogenicity in both the genital and GI tract. The double attack pattern of Chlamydia -induced genital tract inflammatory lesions is as follows: CM infection of oviduct epithelial cells initiates the process of oviduct repair as the first attack. After genital CM spreads to the GI tract, activated chlamydia-specific CD8 + T cells are recruited to the genital tract and secreted pro-fibrotic cytokines such as TNF-α and IL-13. This process is called the second attack which transform tubal repair initiated by the first attack into long-term tubal fibrosis/hydrosalpinx. Elucidating the pathogenic mechanism of Chlamydia infection can provide new ideas for the development of Chlamydia vaccine, which is expected to solve the problems of infertility caused by repeated CT infection in women.
    沙眼衣原体( Chlamydia trachomatis , CT )生殖道感染较为隐匿,患者常无自觉症状,感染后延误治疗的女性可导致严重的并发症。鼠型衣原体( Chlamydia muridarum , CM )感染小鼠生殖道可模拟 CT 感染女性生殖道,是研究 CT 发病机制的理想模型。 CM 质粒蛋白pGP3、染色体蛋白TC0237/TC0668、 CM 特异性CD8 + T细胞和细胞因子TNF-α、IL-13能够诱导生殖道炎症,CD4 + T细胞则负责清除 CM 。然而,生殖道 CM 被清除后,输卵管炎症仍持续存在。生殖道 CM 可自行播散至胃肠道,胃肠道 CM 不能逆向返回生殖道。生殖道传播至胃肠道的 CM 在胃肠道存活的时间和数量与输卵管长期病变呈正相关,而直接接种至胃肠道的 CM 对生殖道与胃肠道均无致病性。衣原体致生殖道长期炎症反应的双重攻击模式被提出: CM 感染生殖道输卵管上皮细胞后启动输卵管修复过程为首次攻击;生殖道 CM 传播至胃肠道后,活化的衣原体特异性CD8 + T细胞被募集至生殖道,分泌促纤维化细胞因子如TNF-α和IL-13为二次攻击,将首次攻击启动的输卵管修复转化为长期输卵管纤维化/积水。研究衣原体感染的致病机制可为衣原体疫苗研发提供新思路,并为解决女性因 CT 反复感染导致不孕等问题提供参考依据。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号