Plasmodium berghei

伯氏疟原虫
  • 文章类型: Journal Article
    背景:研究和发现疟原虫性发育的分子机制对于开发传播阻断药物和根除疟疾至关重要。这项研究的目的是研究使用磷酸酶抑制剂作为筛选疟原虫性发育所必需的蛋白质的工具的可行性,并发现影响疟疾寄生虫性发育的蛋白质。
    方法:使用磷酸化蛋白质组学方法评估了在体外卵形因子培养条件下与BVT-948孵育的疟原虫配子细胞之间蛋白质磷酸化的差异。进行基因本体论(GO)分析以预测BVT-948影响配子体-卵细胞转换的机制。评估了8种与伯氏疟原虫性发育有关的推定蛋白质的功能。生物信息学分析用于评估PBANKA_0100800在配子发生和随后的性发育中的可能机制。
    结果:用BVT-948治疗后,265个蛋白的磷酸化水平降低,67个蛋白的磷酸化水平升高。选择用于表型筛选的8个基因中的7个在伯氏疟原虫性发育中发挥作用,其中4个与配子细胞的发生有关。PBANKA_0100800在配子体-卵母细胞转化和向蚊子传播中起着至关重要的作用。
    结论:通过筛选鉴定出的7种蛋白质影响伯氏疟原虫的性发育,表明磷酸酶抑制剂可用于功能性蛋白质筛选。
    BACKGROUND: Studying and discovering the molecular mechanism of Plasmodium sexual development is crucial for the development of transmission blocking drugs and malaria eradication. The aim of this study was to investigate the feasibility of using phosphatase inhibitors as a tool for screening proteins essential for Plasmodium sexual development and to discover proteins affecting the sexual development of malaria parasites.
    METHODS: Differences in protein phosphorylation among Plasmodium gametocytes incubated with BVT-948 under in vitro ookinete culture conditions were evaluated using phosphoproteomic methods. Gene Ontology (GO) analysis was performed to predict the mechanism by which BVT-948 affected gametocyte-ookinete conversion. The functions of 8 putative proteins involved in Plasmodium berghei sexual development were evaluated. Bioinformatic analysis was used to evaluate the possible mechanism of PBANKA_0100800 in gametogenesis and subsequent sexual development.
    RESULTS: The phosphorylation levels of 265 proteins decreased while those of 67 increased after treatment with BVT-948. Seven of the 8 genes selected for phenotype screening play roles in P. berghei sexual development, and 4 of these were associated with gametocytogenesis. PBANKA_0100800 plays essential roles in gametocyte-ookinete conversion and transmission to mosquitoes.
    CONCLUSIONS: Seven proteins identified by screening affect P. berghei sexual development, suggesting that phosphatase inhibitors can be used for functional protein screening.
    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
    中性粒细胞和巨噬细胞通过激活TLR9功能将病原体截留在细胞外陷阱(ET)中来限制病原体。然而,疟原虫寄生虫分泌TatD样DNA酶(TatD)以抵消ET介导的免疫清除。我们发现TLR9突变小鼠对啮齿动物疟疾的易感性增加,表明TLR9是宿主防御的关键蛋白。我们发现,与WT小鼠相比,TLR9突变小鼠对疟原虫感染的中性粒细胞和巨噬细胞的比例显着降低。重要的是,PbTatD可以直接与巨噬细胞表面TLR9(sTLR9)结合,阻断丝裂原活化蛋白激酶和核因子-κB的磷酸化,负调节巨噬细胞和中性粒细胞形成ET的信号传导。这样,P.berheiTatD是一种寄生虫毒力因子,可以通过直接结合细胞表面的TLR9受体来抑制巨噬细胞和中性粒细胞的增殖,从而阻断下游MyD88-NF-kB途径的激活。
    Neutrophils and macrophages confine pathogens by entrapping them in extracellular traps (ETs) through activating TLR9 function. However, plasmodial parasites secreted TatD-like DNases (TatD) to counteract ETs-mediated immune clearance. We found that TLR9 mutant mice increased susceptibility to rodent malaria, suggesting TLR9 is a key protein for host defense. We found that the proportion of neutrophils and macrophages in response to plasmodial parasite infection in the TLR9 mutant mice was significantly reduced compared to that of the WT mice. Importantly, PbTatD can directly bind to the surface TLR9 (sTLR9) on macrophages, which blocking the phosphorylation of mitogen-activated protein kinase and nuclear factor-κB, negatively regulated the signaling of ETs formation by both macrophages and neutrophils. Such, P. berghei TatD is a parasite virulence factor that can inhibit the proliferation of macrophages and neutrophils through directly binding to TLR9 receptors on the cell surface, thereby blocking the activation of the downstream MyD88-NF-kB pathways.
    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
    临床上耐药的疟疾寄生虫的出现迫切需要开发新药物。蚊子是多种病原体的传播媒介,已经形成了对它们的抗性机制,通常涉及抗菌肽(AMP)。An-cecB是疟疾传播蚊子按蚊的AMP,我们在此报告了其体外对恶性疟原虫3D7,青蒿素抗性菌株803和氯喹抗性菌株Dd2的抗疟活性。我们还证明了它在体内的抗寄生虫活性,使用啮齿动物疟疾寄生虫伯氏疟原虫(ANKA)。我们表明,An-cecB具有有效的抗疟活性,其作用机制可能是通过直接杀死寄生虫或通过与感染的红细胞膜相互作用而发生的。不幸的是,发现An-cecB对哺乳动物细胞具有细胞毒性,并且在体内具有较差的抗疟活性。然而,其截短的肽An-cecB-1保留了其大部分抗疟疾活性,并避免了其在体外的细胞毒性。An-cecB-1在体内也显示出更好的抗疟活性。蚊源AMPs可能为开发抗耐药寄生虫的抗疟药物提供新思路,和An-cecB具有作为抗疟疾肽的模板的潜在用途。
    The emergence of clinically drug-resistant malaria parasites requires the urgent development of new drugs. Mosquitoes are vectors of multiple pathogens and have developed resistance mechanisms against them, which often involve antimicrobial peptides (AMPs). An-cecB is an AMP of the malaria-transmitting mosquito genus Anopheles, and we herein report its antimalarial activity against Plasmodium falciparum 3D7, the artemisinin-resistant strain 803, and the chloroquine-resistant strain Dd2 in vitro. We also demonstrate its anti-parasite activity in vivo, using the rodent malaria parasite Plasmodium berghei (ANKA). We show that An-cecB displays potent antimalarial activity and that its mechanism of action may occur through direct killing of the parasite or through interaction with infected red blood cell membranes. Unfortunately, An-cecB was found to be cytotoxic to mammalian cells and had poor antimalarial activity in vivo. However, its truncated peptide An-cecB-1 retained most of its antimalarial activity and avoided its cytotoxicity in vitro. An-cecB-1 also showed better antimalarial activity in vivo. Mosquito-derived AMPs may provide new ideas for the development of antimalarial drugs against drug-resistant parasites, and An-cecB has potential use as a template for antimalarial peptides.
    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
    背景:疟疾传播阻断疫苗(TBV)旨在抑制蚊子中疟疾寄生虫的发育,并防止进一步传播给人类宿主。推定分泌的卵形因子蛋白25(PSOP25),在疟原虫属中高度保守。,是一个有前途的TBV目标。这里,我们使用转基因鼠寄生虫伯氏疟原虫和临床间日疟原虫分离株,研究了间日疟原虫作为TBV候选物的PvPSOP25。
    结果:产生表达PvPSOP25(TrPvPSOP25Pb)的转基因伯氏疟原虫品系。全长PvPSOP25在巴斯德毕赤酵母中表达并用于免疫小鼠以获得抗rPvPSOP25血清。通过体外测定和蚊食实验评估了抗rPvPSOP25血清的传播阻断活性。通过用rPvPSOP25免疫产生的抗血清特异性识别在TrPvPSOP25Pb启动中表达的天然PvPSOP25抗原。体外实验表明,免疫血清可显着抑制TrPvPSOP25Pb寄生虫的鞭毛和卵形形成。与对照组相比,以感染转基因寄生虫并被动转移抗rPvPSOP25血清的小鼠为食的蚊子显示卵囊密度降低了70.7%。在对五种临床间日疟原虫分离物进行的直接膜饲喂试验中,小鼠抗rPvPSOP25抗体显著降低了卵囊密度,同时对蚊子感染率的影响可忽略不计.
    结论:该研究支持表达间日疟原虫抗原的转基因鼠疟疾寄生虫作为评估间日疟原虫TBV候选物的有用工具的可行性。同时,所产生的抗rPvPSOP25血清的中等传输减少活性需要进一步研究以优化其疗效.
    BACKGROUND: Malaria transmission-blocking vaccines (TBVs) aim to inhibit malaria parasite development in mosquitoes and prevent further transmission to the human host. The putative-secreted ookinete protein 25 (PSOP25), highly conserved in Plasmodium spp., is a promising TBV target. Here, we investigated PvPSOP25 from P. vivax as a TBV candidate using transgenic murine parasite P. berghei and clinical P. vivax isolates.
    RESULTS: A transgenic P. berghei line expressing PvPSOP25 (TrPvPSOP25Pb) was generated. Full-length PvPSOP25 was expressed in the yeast Pichia pastoris and used to immunize mice to obtain anti-rPvPSOP25 sera. The transmission-blocking activity of the anti-rPvPSOP25 sera was evaluated through in vitro assays and mosquito-feeding experiments. The antisera generated by immunization with rPvPSOP25 specifically recognized the native PvPSOP25 antigen expressed in TrPvPSOP25Pb ookinetes. In vitro assays showed that the immune sera significantly inhibited exflagellation and ookinete formation of the TrPvPSOP25Pb parasite. Mosquitoes feeding on mice infected with the transgenic parasite and passively transferred with the anti-rPvPSOP25 sera showed a 70.7% reduction in oocyst density compared to the control group. In a direct membrane feeding assay conducted with five clinical P. vivax isolates, the mouse anti-rPvPSOP25 antibodies significantly reduced the oocyst density while showing a negligible influence on mosquito infection prevalence.
    CONCLUSIONS: This study supported the feasibility of transgenic murine malaria parasites expressing P. vivax antigens as a useful tool for evaluating P. vivax TBV candidates. Meanwhile, the moderate transmission-reducing activity of the generated anti-rPvPSOP25 sera necessitates further research to optimize its efficacy.
    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
    雄性配子细胞分化为鞭毛可育的雄性配子依赖于轴突的组装,疟疾寄生虫蚊子传播的男性发育的主要组成部分。RNA结合蛋白(RBP)介导的mRNA转录后调控在真核细胞性发育中发挥重要作用,包括雌性疟原虫的发育。然而,RBP在疟原虫雄性转录组的定义及其在雄性配子发生中的功能中的作用尚不完全清楚.这里,我们对性别特异性RBPs进行了全基因组筛查,并在疟原虫中发现了一个未描述的男性特异性RBP基因Rbpm1.RBPm1定位于雄性配子细胞核中。缺乏RBPm1的寄生虫无法组装轴突以进行雄性配子发生,从而无法传播蚊子。RBPm1与剪接体E复合物相互作用,并调节一组26个轴突基因中某些内含子的剪接起始。RBPm1缺乏导致这些轴突基因的内含子保留和蛋白质丢失。内含子缺失可恢复RBPm1空配子细胞中的轴突蛋白表达并部分矫正轴突缺陷。报告基因和内源基因中的进一步剪接测定均显示出RBPm1对轴突内含子的严格识别。剪接激活因子RBPm1及其靶内含子构成了疟原虫雄性发育所必需的转录后调节中的轴突内含子剪接程序。
    Differentiation of male gametocytes into flagellated fertile male gametes relies on the assembly of axoneme, a major component of male development for mosquito transmission of the malaria parasite. RNA-binding protein (RBP)-mediated post-transcriptional regulation of mRNA plays important roles in eukaryotic sexual development, including the development of female Plasmodium. However, the role of RBP in defining the Plasmodium male transcriptome and its function in male gametogenesis remains incompletely understood. Here, we performed genome-wide screening for gender-specific RBPs and identified an undescribed male-specific RBP gene Rbpm1 in the Plasmodium. RBPm1 is localized in the nucleus of male gametocytes. RBPm1-deficient parasites fail to assemble the axoneme for male gametogenesis and thus mosquito transmission. RBPm1 interacts with the spliceosome E complex and regulates the splicing initiation of certain introns in a group of 26 axonemal genes. RBPm1 deficiency results in intron retention and protein loss of these axonemal genes. Intron deletion restores axonemal protein expression and partially rectifies axonemal defects in RBPm1-null gametocytes. Further splicing assays in both reporter and endogenous genes exhibit stringent recognition of the axonemal introns by RBPm1. The splicing activator RBPm1 and its target introns constitute an axonemal intron splicing program in the post-transcriptional regulation essential for Plasmodium male development.
    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
    背景:脑型疟疾(CM)是疟疾最致命的并发症,幸存者通常会忍受神经系统后遗症。值得注意的是,浸润性疟原虫激活的CD8+T细胞对脑微血管内皮细胞的细胞毒性作用是具有血脑屏障破坏的实验性CM(ECM)模型的突出特征.然而,CD8+T细胞浸润脑实质对神经元的损伤作用尚不清楚.基于PD-1/PD-L1通路对T细胞的免疫抑制作用,我们之前的研究表明,全身上调PD-L1抑制CD8+T细胞功能可以有效缓解ECM小鼠的症状。然而,神经元是否可以通过PD-1/PD-L1阴性免疫调节途径抑制CD8+T细胞的致病作用尚未见报道。作为CM的重要炎症因子,干扰素可以根据神经免疫微环境通过不同的分子机制诱导PD-L1的表达。因此,本研究旨在探讨CD8+T细胞与神经元之间的直接相互作用,以及神经元通过上调IFN诱导的PD-L1减轻CD8+T细胞致病作用的机制。
    方法:使用感染伯氏疟原虫ANKA(PbA)的C57BL/6J小鼠的ECM模型,通过电子显微镜和IF染色进行体内形态学观察。通过IF染色和延时摄影观察了ECMCD8T细胞(来自ECM小鼠脾脏的免疫磁珠分选)与体外原代培养的皮质神经元之间的相互作用。RNA-seq分析IFNβ或IFNγ诱导的神经元PD-L1上调的信号通路,并通过q-PCR验证,WB,IF染色,和使用IFNAR或IFNGR基因敲除小鼠的体外和体内流式细胞术。腺病毒介导的PD-L1IgGFc融合蛋白表达的保护作用在体内脑立体定向注射的ECM小鼠和体外通过病毒感染的原代培养神经元中得到验证。
    结果:体内,ECM小鼠脑实质显示活化的CD8+T细胞浸润和神经元损伤。体外,ECMCD8+T细胞与神经元直接接触并诱导轴突损伤,作为一种积极的行为。在ECM小鼠神经元和IFNβ诱导的原代培养神经元中PD-L1蛋白水平升高,IFNγ,或体外ECMCD8+T细胞。此外,IFNβ或IFNγ诱导的PD-L1神经元表达是通过IFN受体增加STAT1/IRF1途径介导的。在删除IFNAR或IFNGR后,PbA感染期间神经元中PD-L1表达的增加减弱。腺病毒增加的PD-L1表达在体外和体内部分保护神经元免受CD8+T细胞介导的损伤。
    结论:我们的研究表明,I型和II型IFN均可通过IFN受体介导的STAT1/IRF1途径诱导神经元上调PD-L1,以防止活化的CD8+T细胞介导的损伤,提供了一个靶向途径来减轻ECM过程中的神经炎症。
    BACKGROUND: Cerebral malaria (CM) is the most lethal complication of malaria, and survivors usually endure neurological sequelae. Notably, the cytotoxic effect of infiltrating Plasmodium-activated CD8+ T cells on cerebral microvasculature endothelial cells is a prominent feature of the experimental CM (ECM) model with blood-brain barrier disruption. However, the damage effect of CD8+ T cells infiltrating the brain parenchyma on neurons remains unclear. Based on the immunosuppressive effect of the PD-1/PD-L1 pathway on T cells, our previous study demonstrated that the systemic upregulation of PD-L1 to inhibit CD8+ T cell function could effectively alleviate the symptoms of ECM mice. However, it has not been reported whether neurons can suppress the pathogenic effect of CD8+ T cells through the PD-1/PD-L1 negative immunomodulatory pathway. As the important inflammatory factor of CM, interferons can induce the expression of PD-L1 via different molecular mechanisms according to the neuro-immune microenvironment. Therefore, this study aimed to investigate the direct interaction between CD8+ T cells and neurons, as well as the mechanism of neurons to alleviate the pathogenic effect of CD8+ T cells through up-regulating PD-L1 induced by IFNs.
    METHODS: Using the ECM model of C57BL/6J mice infected with Plasmodium berghei ANKA (PbA), morphological observations were conducted in vivo by electron microscope and IF staining. The interaction between the ECM CD8+ T cells (immune magnetic bead sorting from spleen of ECM mice) and primary cultured cortical neurons in vitro was observed by IF staining and time-lapse photography. RNA-seq was performed to analyze the signaling pathway of PD-L1 upregulation in neurons induced by IFNβ or IFNγ, and verified through q-PCR, WB, IF staining, and flow cytometry both in vitro and in vivo using IFNAR or IFNGR gene knockout mice. The protective effect of adenovirus-mediated PD-L1 IgGFc fusion protein expression was verified in ECM mice with brain stereotaxic injection in vivo and in primary cultured neurons via viral infection in vitro.
    RESULTS: In vivo, ECM mice showed infiltration of activated CD8+ T cells and neuronal injury in the brain parenchyma. In vitro, ECM CD8+ T cells were in direct contact with neurons and induced axonal damage, as an active behavior. The PD-L1 protein level was elevated in neurons of ECM mice and in primary cultured neurons induced by IFNβ, IFNγ, or ECM CD8+ T cells in vitro. Furthermore, the IFNβ or IFNγ induced neuronal expression of PD-L1 was mediated by increasing STAT1/IRF1 pathway via IFN receptors. The increase of PD-L1 expression in neurons during PbA infection was weakened after deleting the IFNAR or IFNGR. Increased PD-L1 expression by adenovirus partially protected neurons from CD8+ T cell-mediated damage both in vitro and in vivo.
    CONCLUSIONS: Our study demonstrates that both type I and type II IFNs can induce neurons to upregulate PD-L1 via the STAT1/IRF1 pathway mediated by IFN receptors to protect against activated CD8+ T cell-mediated damage, providing a targeted pathway to alleviate neuroinflammation during ECM.
    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
    恶性疟原虫感染导致脑型疟疾时通常是致命的,这与神经病理学相关,被描述为压倒性的炎症反应和脑微血管的机械性阻塞。PI3Kγ是细胞内信号转导的重要组成部分,在调节细胞趋化性中起着核心作用,迁移,和激活。本研究的目的是研究抑制PI3Kγ途径与感染小鼠疟原虫的C57BL/6J小鼠实验性脑型疟疾(ECM)结局之间的关系,伯氏疟原虫安卡。我们观察到在感染后口服施用PI3Kγ抑制剂IPI549完全保护小鼠免受ECM。IPI549治疗可显着抑制炎症反应的程度,随着促炎因子的产生减少,减少T细胞活化,和改变抗原呈递细胞的分化。IPI549治疗保护受感染的小鼠免受神经病理学的影响,通过观察到的大脑中致病性T细胞的减少来评估。在寄生虫接种后三天用IPI549处理感染的小鼠改善了鼠血脑屏障(BBB)的完整性并帮助小鼠通过ECM的发作。一起,这些数据表明,口服PI3Kγ抑制剂IPI549对宿主炎症具有抑制作用,并减轻脑病理学,它支持IPI549作为一种新的疟疾治疗选择,对脑型疟疾具有潜在的治疗意义。
    Infection with Plasmodium falciparum is often deadly when it results in cerebral malaria, which is associated with neuropathology described as an overwhelming inflammatory response and mechanical obstruction of cerebral microvascular. PI3Kγ is a critical component of intracellular signal transduction and plays a central role in regulating cell chemotaxis, migration, and activation. The purpose of this study was to examine the relationship between inhibiting the PI3Kγ pathway and the outcome of experimental cerebral malaria (ECM) in C57BL/6J mice infected with the mouse malaria parasite, Plasmodium berghei ANKA. We observed that oral administration of the PI3Kγ inhibitor IPI549 after infection completely protected mice from ECM. IPI549 treatment significantly dampened the magnitude of inflammatory responses, with reduced production of pro-inflammatory factors, decreased T cell activation, and altered differentiation of antigen-presenting cells. IPI549 treatment protected the infected mice from neuropathology, as assessed by an observed reduction of pathogenic T cells in the brain. Treating the infected mice with IPI549 three days after parasite inoculation improved the murine blood brain barrier (BBB) integrity and helped the mice pass the onset of ECM. Together, these data indicate that oral administration of the PI3Kγ inhibitor IPI549 has a suppressive role in host inflammation and alleviates cerebral pathology, which supports IPI549 as a new malaria treatment option with potential therapeutic implications for cerebral malaria.
    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
    RTS,S是第一个建议在有风险的幼儿中实施的疟疾疫苗。然而,疫苗效力是适度和短暂的。为了减轻5岁以下儿童的脑型疟疾(CM)风险,必须开发新的疫苗。EV是潜在的疫苗候选物,因为它们在感染期间获得脑靶向递送和转移疟原虫抗原和免疫调节剂的能力。本研究从感染约氏疟原虫17XNL(P。y17XNL)。在第0天和第20天,对C57BL/6J小鼠进行EV静脉内免疫(EV-I.V.CM组)或皮下注射EV和CpGODN-1826的组合(EVCPGODN-S.C.CM组),然后感染伯氏疟原虫ANKA(P。bANKA)在第二次免疫后第20天。我们监测了寄生虫血症和生存率。使用伊文思蓝染色检查血脑屏障(BBB)的完整性。使用Luminex评估细胞因子和粘附分子的水平,RT-qPCR,和WB。通过苏木精和曙红和免疫组织化学染色评估脑病理学。血清IgG水平,通过酶联免疫吸附测定分析IgG1和IgG2a。与P.bANKA感染组相比,寄生虫血症缓慢增加,死亡延迟(感染后第10天),EV-I.V.ECM和EVCPGODN-S.C.ECM组的存活率达到75%-83.3%。同时,与EV+CPGODN-S.C.+ECM组相比,虽然寄生虫血症几乎是一样的,EV-I.V.+ECM组的存活率增加。此外,EV免疫显着下调脾脏和大脑中的炎症反应,并改善脑部病理变化,包括BBB破坏和感染的红细胞(iRBC)隔离。此外,与正常对照组相比,EV免疫组表现出增强的抗体应答(IgG1和IgG2a产生上调).EV免疫发挥了保护作用,提高BBB的完整性,下调脑组织的炎症反应,结果降低了CM的发生率。保护作用由EV引起的免疫途径和脑靶标确定。静脉免疫比皮下免疫表现更好。这可能与电动汽车相关,能自然穿越血脑屏障发挥较好的脑保护作用。
    RTS,S is the first malaria vaccine recommended for implementation among young children at risk. However, vaccine efficacy is modest and short-lived. To mitigate the risk of cerebral malaria (CM) among children under the age of 5, it is imperative to develop new vaccines. EVs are potential vaccine candidates as they obtain the ability of brain-targeted delivery and transfer plasmodium antigens and immunomodulators during infections. This study extracted EVs from BALB/c mice infected with Plasmodium yoelii 17XNL (P.y17XNL). C57BL/6J mice were intravenously immunized with EVs (EV-I.V. + CM group) or subcutaneously vaccinated with the combination of EVs and CpG ODN-1826 (EV + CPG ODN-S.C. + CM group) on days 0 and 20, followed by infection with Plasmodium berghei ANKA (P.bANKA) on day 20 post-second immunization. We monitored Parasitemia and survival rate. The integrity of the Blood-brain barrier (BBB) was examined using Evans blue staining.The levels of cytokines and adhesion molecules were evaluated using Luminex, RT-qPCR, and WB. Brain pathology was evaluated by hematoxylin and eosin and immunohistochemical staining. The serum levels of IgG, IgG1, and IgG2a were analyzed by enzyme-linked immunosorbent assay. Compared with those in the P.bANKA-infected group, parasitemia increased slowly, death was delayed (day 10 post-infection), and the survival rate reached 75 %-83.3 % in the EV-I.V. + ECM and EV + CPG ODN-S.C. + ECM groups. Meanwhile, compared with the EV + CPG ODN-S.C. + ECM group, although parasitemia was almost the same, the survival rate increased in the EV-I.V. + ECM group.Additionally, EVs immunization markedly downregulated inflammatory responses in the spleen and brain and ameliorated brain pathological changes, including BBB disruption and infected red blood cell (iRBC) sequestration. Furthermore, the EVs immunization group exhibited enhanced antibody responses (upregulation of IgG1 and IgG2a production) compared to the normal control group. EV immunization exerted protective effects, improving the integrity of the BBB, downregulating inflammation response of brain tissue, result in reduces the incidence of CM. The protective effects were determined by immunological pathways and brain targets elicited by EVs. Intravenous immunization exhibited better performance than subcutaneous immunization, which perhaps correlated with EVs, which can naturally cross BBB to play a better role in brain protection.
    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
    寄生虫特异性CD4+Th1细胞反应是控制疟疾感染的主要免疫效应;然而,潜在的监管机制在很大程度上仍然未知。这项研究表明,骨髓细胞中的ATG5缺乏可以通过选择性增强寄生虫特异性CD4Th1细胞反应来显着抑制啮齿动物血液阶段疟疾寄生虫的生长。这种作用与ATG5介导的规范和非规范自噬无关。机械上,ATG5缺乏症抑制了FAS介导的LY6G-ITGAM/CD11bADGRE1/F4/80-细胞的凋亡,随后增加了寄生虫感染小鼠的CCL2/MCP-1产量。LY6G-ITGAM+ADGRE1-细胞衍生的CCL2通过JAK2-STAT4途径与CD4+Th1细胞上的CCR2选择性相互作用以优化其应答。重组CCL2的施用显著促进寄生虫特异性CD4+Th1应答并抑制疟疾感染。最后,我们的研究强调了ATG5在调节骨髓细胞凋亡并依次影响CCL2产生中先前未被识别的作用,选择性地促进CD4+Th1细胞反应。我们的发现为免疫干预和有效抗疟疾疫苗的开发提供了新的见解。
    Parasite-specific CD4+ Th1 cell responses are the predominant immune effector for controlling malaria infection; however, the underlying regulatory mechanisms remain largely unknown. This study demonstrated that ATG5 deficiency in myeloid cells can significantly inhibit the growth of rodent blood-stage malarial parasites by selectively enhancing parasite-specific CD4+ Th1 cell responses. This effect was independent of ATG5-mediated canonical and non-canonical autophagy. Mechanistically, ATG5 deficiency suppressed FAS-mediated apoptosis of LY6G- ITGAM/CD11b+ ADGRE1/F4/80- cells and subsequently increased CCL2/MCP-1 production in parasite-infected mice. LY6G- ITGAM+ ADGRE1- cell-derived CCL2 selectively interacted with CCR2 on CD4+ Th1 cells for their optimized responses through the JAK2-STAT4 pathway. The administration of recombinant CCL2 significantly promoted parasite-specific CD4+ Th1 responses and suppressed malaria infection. Conclusively, our study highlights the previously unrecognized role of ATG5 in modulating myeloid cells apoptosis and sequentially affecting CCL2 production, which selectively promotes CD4+ Th1 cell responses. Our findings provide new insights into the development of immune interventions and effective anti-malarial vaccines.Abbreviations: ATG5: autophagy related 5; CBA: cytometric bead array; CCL2/MCP-1: C-C motif chemokine ligand 2; IgG: immunoglobulin G; IL6: interleukin 6; IL10: interleukin 10; IL12: interleukin 12; MFI: mean fluorescence intensity; JAK2: Janus kinase 2; LAP: LC3-associated phagocytosis; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; pRBCs: parasitized red blood cells; RUBCN: RUN domain and cysteine-rich domain containing, Beclin 1-interacting protein; STAT4: signal transducer and activator of transcription 4; Th1: T helper 1 cell; Tfh: follicular helper cell; ULK1: unc-51 like kinase 1.
    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
    背景:基于对妊娠相关疟疾(PAM)胎盘病理特征和安全用药的了解,迫切需要建立稳定的疟原虫感染妊娠小鼠模型。
    方法:将检测到阴道栓子的ICR小鼠随机分为妊娠后感染(疟疾)和未感染妊娠(疟疾)组。未交配的年龄匹配小鼠被感染为孕前感染组(维珍对照),随后与ICR雄性交配。所有小鼠腹腔注射1×106L伯氏疟原虫ANKA感染的红细胞,并给予疟疾组相同量的生理盐水。我们记录了各组不良妊娠结局的发生率和后代的数量。
    结果:维珍组小鼠无法正常受孕,阴道出血,流产,或死产出现在疟疾+组中。不良妊娠结局发生率极高,与对照组(疟疾组)比较差异有统计学意义(P<0.05),其中胎盘表现出与人类妊娠期疟疾相关的病理特征。
    结论:腹腔注射1×106伯氏疟原虫ANKA感染的红细胞可建立ICR小鼠妊娠相关性疟疾模型。
    BACKGROUND: Based on understanding of placental pathological features and safe medication in pregnancy-associated malaria (PAM), establishment of a stable pregnant mouse infection model with Plasmodium was urgently needed.
    METHODS: ICR mice with vaginal plugs detected were randomly divided into post-pregnancy infection (Malaria+) and uninfected pregnancy (Malaria-) cohorts. Age-matched mice that had not been mated were infected as pre-pregnancy infection group (Virgin control), which were subsequently mated with ICR males. All mice were inoculated with 1 × 106Plasmodium berghei ANKA-infected RBCs by intraperitoneal injection, and the same amount of saline was given to Malaria- group. We recorded the incidence of adverse pregnancy outcomes and the amounts of offspring in each group.
    RESULTS: The Virgin group mice were unable to conceive normally, and vaginal bleeding, abortion, or stillbirth appeared in the Malaria+ group. The incidence of adverse pregnancy outcomes was extremely high and statistically significant compared with the control (Malaria-) group (P < 0.05), of which placenta exhibited pathological features associated with human gestational malaria.
    CONCLUSIONS: The intraperitoneal injection of 1 × 106Plasmodium berghei ANKA-infected RBCs could establish a model of pregnancy-associated malaria in ICR mouse.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号