Malaria, Cerebral

疟疾,大脑
  • 文章类型: Journal Article
    背景:在脑型疟疾(CM)中观察到氨基酸代谢紊乱。这项研究旨在确定从昏迷中恢复的儿童中异常的氨基酸浓度是否与意识水平相关。纵向定量21个氨基酸和昏迷评分,并分析数据的相关性。
    方法:在一项前瞻性观察研究中,纳入了42名CM儿童。在进入时和之后的频繁间隔测量氨基酸水平,并通过Blantyre昏迷评分(BCS)评估意识。36名健康儿童作为国内正常氨基酸范围的对照。使用广义线性混合效应模型采用Logistic回归来评估超出范围的氨基酸水平与BCS之间的关联。
    结果:在条目16/21氨基酸水平超出范围。纵向分析显示10/21个超范围氨基酸与BCS显著相关。升高的苯丙氨酸水平显示与低BCS的相关性最高。当在每个采样时间分析超出正常范围的数据时,这一发现成立。
    结论:提供了氨基酸水平异常与CM恢复之间关联的纵向数据。在与BCS显著相关的10个氨基酸中,升高的苯丙氨酸可能是炎症的醚脂质介质清除受损的替代品,并且可能有助于CM的发病机理。
    BACKGROUND: Disordered amino acid metabolism is observed in cerebral malaria (CM). This study sought to determine whether abnormal amino acid concentrations were associated with level of consciousness in children recovering from coma. Twenty-one amino acids and coma scores were quantified longitudinally and the data were analysed for associations.
    METHODS: In a prospective observational study, 42 children with CM were enrolled. Amino acid levels were measured at entry and at frequent intervals thereafter and consciousness was assessed by Blantyre Coma Scores (BCS). Thirty-six healthy children served as controls for in-country normal amino acid ranges. Logistic regression was employed using a generalized linear mixed-effects model to assess associations between out-of-range amino acid levels and BCS.
    RESULTS: At entry 16/21 amino acid levels were out-of-range. Longitudinal analysis revealed 10/21 out-of-range amino acids were significantly associated with BCS. Elevated phenylalanine levels showed the highest association with low BCS. This finding held when out-of-normal-range data were analysed at each sampling time.
    CONCLUSIONS: Longitudinal data is provided for associations between abnormal amino acid levels and recovery from CM. Of 10 amino acids significantly associated with BCS, elevated phenylalanine may be a surrogate for impaired clearance of ether lipid mediators of inflammation and may contribute to CM pathogenesis.
    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
    使用非侵入性方法很难研究脑型疟疾对脑组织转录谱的影响。我们从脑型疟疾和社区对照患者中分离了血浆细胞外囊泡(EV),并对其mRNA含量进行了测序。去卷积分析显示,来自脑型疟疾的电动汽车富含脑源性转录本。我们根据从横截面收集的样本中的EV转录谱和推断的疾病轨迹,同时使用健康的社区对照作为起点,对患有脑型疟疾的患者进行了排序。我们发现血浆EV中的神经元转录本随着疾病轨迹而减少,而来自神经胶质的转录本,内皮,免疫细胞增加。疾病轨迹与死亡等严重程度指标呈正相关,并与VEGFA-VEGFR和谷氨酸能信号的增加相关。以及血小板和中性粒细胞活化。这些数据表明,脑型疟疾的脑组织反应可以使用外周血中循环的电动汽车进行非侵入性研究。
    The impact of cerebral malaria on the transcriptional profiles of cerebral tissues is difficult to study using noninvasive approaches. We isolated plasma extracellular vesicles (EVs) from patients with cerebral malaria and community controls and sequenced their mRNA content. Deconvolution analysis revealed that EVs from cerebral malaria are enriched in transcripts of brain origin. We ordered the patients with cerebral malaria based on their EV-transcriptional profiles from cross-sectionally collected samples and inferred disease trajectory while using healthy community controls as a starting point. We found that neuronal transcripts in plasma EVs decreased with disease trajectory, whereas transcripts from glial, endothelial, and immune cells increased. Disease trajectory correlated positively with severity indicators like death and was associated with increased VEGFA-VEGFR and glutamatergic signaling, as well as platelet and neutrophil activation. These data suggest that brain tissue responses in cerebral malaria can be studied noninvasively using EVs circulating in peripheral blood.
    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
    脑型疟疾(CM)的标志是在脑微脉管系统中隔离恶性疟原虫感染的红细胞(IE)。IE与内皮的结合减少了微血管流量,结合炎症反应,扰乱内皮屏障功能,导致血脑屏障(BBB)的破坏。细胞粘附导致内皮的激活并改变一系列影响信号通路的细胞过程。受体表达,凝血,和BBB完整性的破坏。这里,我们调查了CM来源的寄生虫是否对人脑微血管内皮细胞(HBMECs)产生不同的影响,与简单的疟疾(UM)衍生的寄生虫相比。来自UM和CM临床病例的患者源性IE,以及非结合骨架结合蛋白1敲除寄生虫,覆盖在肿瘤坏死因子(TNF)激活的HBMECs上。使用一组参与炎症的基因的基于探针的测定进行内皮反应的基因表达分析。凋亡,内皮屏障功能,和前列环素合成途径。我们观察到在IE存在下对内皮转录反应的显著影响,然而,HBMEC反应与临床综合征(UM或CM)类型之间没有显着相关性。此外,HBMEC基因表达与本身的结合和IE与HBMECs的结合水平之间没有相关性,因为我们在使用结合和非结合寄生虫时检测到相同的内皮反应变化。我们的结果表明,在这种共培养模型中,IE与内皮细胞的相互作用诱导了一些内皮反应,这些反应与临床起源无关,也与IE表面上主要变异抗原恶性疟原虫红细胞膜蛋白1的表达无关。
    目的:脑型疟疾(CM)是严重恶性疟原虫感染最普遍和致命的并发症。这种疾病的标志是恶性疟原虫感染的红细胞(IE)在脑微脉管系统中的隔离,其最终导致血脑屏障的破坏。这里,我们比较了一组基因中来自无并发症疟疾(UM)和CM病例的恶性疟原虫寄生虫对人脑微血管内皮细胞(HBMECs)相对基因表达的影响。我们观察到在IE存在下对内皮转录反应的显著影响,然而,HBMEC反应与临床综合征(UM或CM)类型之间没有显着相关性。此外,HBMEC基因表达与本身的结合以及IE与HBMECs的结合水平之间没有相关性。我们的结果表明,IE与内皮细胞的相互作用诱导了与临床起源无关的内皮反应,而不是完全由表面恶性疟原虫红细胞膜蛋白1表达驱动。
    A hallmark of cerebral malaria (CM) is sequestration of Plasmodium falciparum-infected erythrocytes (IE) within the brain microvasculature. Binding of IE to endothelium reduces microvascular flow and, combined with an inflammatory response, perturbs endothelial barrier function, resulting in breakdown of the blood-brain barrier (BBB). Cytoadherence leads to activation of the endothelium and alters a range of cell processes affecting signaling pathways, receptor expression, coagulation, and disruption of BBB integrity. Here, we investigated whether CM-derived parasites elicit differential effects on human brain microvascular endothelial cells (HBMECs), as compared to uncomplicated malaria (UM)-derived parasites. Patient-derived IE from UM and CM clinical cases, as well as non-binding skeleton-binding protein 1 knockout parasites, were overlaid onto tumour necrosis factor (TNF)-activated HBMECs. Gene expression analysis of endothelial responses was performed using probe-based assays of a panel of genes involved in inflammation, apoptosis, endothelial barrier function, and prostacyclin synthesis pathway. We observed a significant effect on endothelial transcriptional responses in the presence of IE, yet there was no significant correlation between HBMEC responses and type of clinical syndrome (UM or CM). Furthermore, there was no correlation between HBMEC gene expression and both binding itself and level of IE binding to HBMECs, as we detected the same change in endothelial responses when employing both binding and non-binding parasites. Our results suggest that interaction of IE with endothelial cells in this co-culture model induces some endothelial responses that are independent of clinical origin and independent of the expression of the major variant antigen Plasmodium falciparum erythrocyte membrane protein 1 on the IE surface.
    OBJECTIVE: Cerebral malaria (CM) is the most prevalent and deadly complication of severe Plasmodium falciparum infection. A hallmark of this disease is sequestration of P. falciparum-infected erythrocytes (IE) in brain microvasculature that ultimately results in breakdown of the blood-brain barrier. Here, we compared the effect of P. falciparum parasites derived from uncomplicated malaria (UM) and CM cases on the relative gene expression of human brain microvascular endothelial cells (HBMECs) for a panel of genes. We observed a significant effect on the endothelial transcriptional response in the presence of IE, yet there is no significant correlation between HBMEC responses and the type of clinical syndrome (UM or CM). Furthermore, there was no correlation between HBMEC gene expression and both binding itself and the level of IE binding to HBMECs. Our results suggest that interaction of IE with endothelial cells induces endothelial responses that are independent of clinical origin and not entirely driven by surface Plasmodium falciparum erythrocyte membrane protein 1 expression.
    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
    在神经系统受累(中枢神经系统[CNS]疟疾)的疟疾中幸存下来的儿童中有三分之一会出现后遗症。较高的最高温度(Tmax)和癫痫发作是后遗症的危险因素。
    比较使用对乙酰氨基酚和布洛芬的积极退热治疗与仅在38.5°C或更高温度下单独给予对乙酰氨基酚的常规治疗。
    这项随机临床试验是在赞比亚的1个三级护理和1个地区医院以及马拉维的一个三级护理中心的住院儿科服务中进行的。包括2至11岁的中枢神经系统疟疾儿童(不包括肌酐>1.2mg/dL的儿童),他们从2019年到2022年注册。数据分析于2022年12月至2023年4月进行。
    积极退热组接受对乙酰氨基酚(30mg/kg负荷,然后15毫克/千克)加布洛芬,10mg/kg,每6小时,无论临床温度为72小时。常规护理组在38.5°C或更高的温度下每6小时根据需要接受15mg/kg对乙酰氨基酚。
    主要结果变量是72小时内的Tmax,随访的总持续时间。次要结果包括癫痫发作和寄生虫清除。
    对553名患者进行了筛查,226(40.9%)不合格,和57(10.3%)下跌。总共256名参与者(n=128/组)的平均(SD)年龄为4.3(2.1)岁;115(45%)为女性,141(55%)为男性。积极退热组的Tmax较低,38.6与39.2°C(差异,-0.62°C;95%CI,-0.82至-0.42;P<.001)并且经历多次或长时间癫痫发作的几率较低,常规护理组10名(8%)vs34名儿童(27%)(比值比[OR],0.26;95%CI,0.12至0.56)。未检测到寄生虫清除时间的组间差异。40名儿童(15%)发生严重不良事件,常规护理组中25例(20%),积极退热组中15例(12%),包括13例死亡(10[8%]和3[2%],分别)。肌酐升高导致常规治疗组的8名儿童(6%)和积极退热组的13名儿童(10%)的研究药物停药(OR,1.74;95%CI,0.63至5.07)。
    这项研究发现,在先前的观察性研究中,积极的解热治疗可将无神经损伤儿童的平均Tmax降低至与Tmax相当的温度水平,并改善急性癫痫发作的结局,而没有寄生虫血症的延长。
    ClinicalTrials.gov标识符:NCT03399318。
    UNASSIGNED: A third of children who survive malaria with neurological involvement (central nervous system [CNS] malaria) develop sequelae. A higher maximum temperature (Tmax) and seizures are risk factors for sequelae.
    UNASSIGNED: To compare aggressive antipyretic therapy using scheduled acetaminophen and ibuprofen vs usual care with acetaminophen alone given only for a temperature of 38.5 °C or higher.
    UNASSIGNED: This randomized clinical trial was conducted at inpatient pediatric services of 1 tertiary care and 1 district hospital in Zambia and a tertiary care center in Malawi. Included were children aged 2 to 11 years with CNS malaria (excluding those with creatinine >1.2 mg/dL), who were enrolled from 2019 to 2022. Data analysis took place from December 2022 to April 2023.
    UNASSIGNED: The aggressive antipyretic group received acetaminophen (30 mg/kg load, then 15 mg/kg) plus ibuprofen, 10 mg/kg, every 6 hours, regardless of clinical temperature for 72 hours. The usual care group received 15 mg/kg of acetaminophen as needed every 6 hours for a temperature of 38.5 °C or higher.
    UNASSIGNED: The primary outcome variable was Tmax over 72 hours, the total duration of follow-up. Secondary outcomes included seizures and parasite clearance.
    UNASSIGNED: Five hundred fifty-three patients were screened, 226 (40.9%) were ineligible, and 57 (10.3%) declined. A total 256 participants (n = 128/group) had a mean (SD) age of 4.3 (2.1) years; 115 (45%) were female, and 141 (55%) were male. The aggressive antipyretic group had a lower Tmax, 38.6 vs 39.2 °C (difference, -0.62 °C; 95% CI, -0.82 to -0.42; P < .001) and lower odds of experiencing multiple or prolonged seizures, 10 (8%) vs 34 children (27%) in the usual care group (odds ratio [OR], 0.26; 95% CI, 0.12 to 0.56). No group difference in parasite clearance time was detected. Severe adverse events occurred in 40 children (15%), 25 (20%) in the usual care group and 15 (12%) in the aggressive antipyretic group, including 13 deaths (10 [8%] and 3 [2%], respectively). Increased creatinine resulted in study drug discontinuation in 8 children (6%) in the usual care group and 13 children (10%) in the aggressive antipyretic group (OR, 1.74; 95% CI, 0.63 to 5.07).
    UNASSIGNED: This study found that aggressive antipyretic therapy reduced mean Tmax to temperature levels comparable with the Tmax among children without neurological impairments in prior observational studies and improved acute seizure outcomes with no prolongation of parasitemia.
    UNASSIGNED: ClinicalTrials.gov Identifier: NCT03399318.
    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
    先天免疫对于减轻寄生虫负担和促进严重疟疾的生存至关重要。单核细胞是先天反应的关键参与者,像巨噬细胞一样,是塑料细胞,其功能和表型受到来自微环境的信号调节。在脑型疟疾(CM)的背景下,单核细胞反应构成一个重要的问题来理解。我们先前证明,非经典单核细胞百分比的降低与CM儿童的死亡结局有关。在目前的研究中,我们推测单核细胞吞噬功能受疟疾感染严重程度的影响.
    为了研究这一假设,我们比较了贝宁患有非复杂性疟疾(UM)和CM的儿童循环单核细胞的调理和非调理吞噬能力。对于CM组,在包含时(D0)和处理后3天和30天(D3,D30)获得样品。单核细胞及其亚群的吞噬能力通过流式细胞术和转录谱分析来表征,方法是研究已知其在感染的红细胞(iRBC)消除或免疫逃逸中的功能意义的基因。
    我们的结果证实了我们的假设,并强调了非经典单核细胞吞噬iRBC的更高能力。我们还证实,与UM相比,低数量的非经典单核细胞与CM结果相关,提示这个亚群动员到大脑炎症部位。最后,我们的结果提示抑制性受体LILRB1,LILRB2和Tim3在吞噬作用控制中的作用.
    放在一起,这些数据提供了更好地了解单核细胞和疟疾感染在CM致病性中的相互作用。
    UNASSIGNED: Innate immunity is crucial to reducing parasite burden and contributing to survival in severe malaria. Monocytes are key actors in the innate response and, like macrophages, are plastic cells whose function and phenotype are regulated by the signals from the microenvironment. In the context of cerebral malaria (CM), monocyte response constitutes an important issue to understand. We previously demonstrated that decreased percentages of nonclassical monocytes were associated with death outcomes in CM children. In the current study, we postulated that monocyte phagocytosis function is impacted by the severity of malaria infection.
    UNASSIGNED: To study this hypothesis, we compared the opsonic and nonopsonic phagocytosis capacity of circulant monocytes from Beninese children with uncomplicated malaria (UM) and CM. For the CM group, samples were obtained at inclusion (D0) and 3 and 30 days after treatment (D3, D30). The phagocytosis capacity of monocytes and their subsets was characterized by flow cytometry and transcriptional profiling by studying genes known for their functional implication in infected-red blood cell (iRBC) elimination or immune escape.
    UNASSIGNED: Our results confirm our hypothesis and highlight the higher capacity of nonclassical monocytes to phagocyte iRBC. We also confirm that a low number of nonclassical monocytes is associated with CM outcome when compared to UM, suggesting a mobilization of this subpopulation to the cerebral inflammatory site. Finally, our results suggest the implication of the inhibitory receptors LILRB1, LILRB2, and Tim3 in phagocytosis control.
    UNASSIGNED: Taken together, these data provide a better understanding of the interplay between monocytes and malaria infection in the pathogenicity of CM.
    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
    Oelschlegel等人的一系列复杂的研究。在脑型疟疾的鼠模型中,建立了将静脉外排减少与灌注受损联系起来的事件的时间序列,水肿,和神经炎症。讨论了与人脑型疟疾的相关性,包括在最近的脑血管血流动力学研究中认识到的异质性。
    A complex series of studies by Oelschlegel et al. in a murine model of cerebral malaria establishes a temporal sequence of events linking decreased venous efflux to impaired perfusion, edema, and neuroinflammation. The relevance to human cerebral malaria is discussed, including the heterogeneity recognized in recent investigations of cerebrovascular hemodynamics.
    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
    脑肿胀的同时,与液体积聚有关,是小儿脑型疟疾(CM)的已知特征,在从CM恢复过程中,液体和大分子是如何从大脑中排出的,目前尚不清楚。使用实验CM(ECM)模型,我们表明,CM期间大脑中的液体积聚是由血管源性水肿驱动的,而不是由血管周围脑脊液(CSF)流入驱动的。我们发现,在具有ECM的小鼠中,液体和分子极快地从大脑中去除到深颈部淋巴结(dcLN),主要通过基础途径,穿过筛板和鼻淋巴管。在协议中,我们证明,引流到dcLN的传入淋巴管结扎会显著损害脑液引流,并降低抗疟疾药物从ECM综合征的恢复.总的来说,我们的结果提供了对协调从CM恢复的途径的洞察。
    While brain swelling, associated with fluid accumulation, is a known feature of pediatric cerebral malaria (CM), how fluid and macromolecules are drained from the brain during recovery from CM is unknown. Using the experimental CM (ECM) model, we show that fluid accumulation in the brain during CM is driven by vasogenic edema and not by perivascular cerebrospinal fluid (CSF) influx. We identify that fluid and molecules are removed from the brain extremely quickly in mice with ECM to the deep cervical lymph nodes (dcLNs), predominantly through basal routes and across the cribriform plate and the nasal lymphatics. In agreement, we demonstrate that ligation of the afferent lymphatic vessels draining to the dcLNs significantly impairs fluid drainage from the brain and lowers anti-malarial drug recovery from the ECM syndrome. Collectively, our results provide insight into the pathways that coordinate recovery from CM.
    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
    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
    背景:血脑屏障(BBB)破坏是脑型疟疾(CM)的主要特征,恶性疟原虫(Pf)感染的严重并发症。在CM中,Pf感染的红细胞(Pf-iRBC)与脑内皮细胞的隔离并伴有炎症,溶血,微血管阻塞和内皮功能障碍介导BBB破坏,导致严重的神经系统症状,包括昏迷和癫痫发作,可能导致死亡或长期后遗症。体外模型提高了我们对CM介导的BBB破坏的认识,但是它们的生理相关性仍然不确定。使用人类诱导的多能干细胞来源的脑微血管内皮细胞(hiPSC-BMECs),我们旨在开发一种新的CMBBB体外模型,表现出增强的阻隔性能。
    方法:将hiPSC-BMEC与HB3var03菌株Pf-iRBC共培养长达9小时。使用跨内皮电阻(TEER)和荧光素钠通透性测定来测量屏障完整性。紧密连接(TJ)蛋白的定位和表达(occludin,小带闭合-1,claudin-5),细胞粘附分子(ICAM-1,VCAM-1),使用免疫荧光成像(IF)和蛋白质印迹(WB)确定内皮表面标志物(EPCR)。使用多重蛋白质组分析器阵列测量血管生成和细胞应激标志物的表达。
    结果:与Pf-iRBC共培养6小时后,与未感染的红细胞共培养相比,hiPSC-BMEC显示TEER降低和荧光素钠通透性增加,表明有一个泄漏的屏障。我们观察到occludin的定位中断,IF的小带闭塞-1和claudin-5,但在Pf-iRBC共培养物中,WB蛋白表达没有变化。与未感染的RBC共培养相比,在具有Pf-iRBC共培养的hiPSC-BMEC中ICAM-1和VCAM-1而非EPCR的表达升高。此外,血管生成素的表达增加,与未感染的RBC共培养物相比,Pf-iRBC共培养物中的血小板因子4和磷酸热休克蛋白27。
    结论:这些发现证明了我们基于hiPSC-BMEC的BBB模型的有效性,显示出增强的屏障完整性和适当的TJ蛋白定位。在hiPSC-BMEC与Pf-iRBC共培养中,减少TEER,细胞旁通透性增加,TJ蛋白定位的变化,增加粘附分子的表达,血管生成和细胞应激的标记都指向一种具有增强屏障特性的新型模型,适用于研究CM中BBB破坏的致病机制。
    BACKGROUND: Blood-brain barrier (BBB) disruption is a central feature of cerebral malaria (CM), a severe complication of Plasmodium falciparum (Pf) infections. In CM, sequestration of Pf-infected red blood cells (Pf-iRBCs) to brain endothelial cells combined with inflammation, hemolysis, microvasculature obstruction and endothelial dysfunction mediates BBB disruption, resulting in severe neurologic symptoms including coma and seizures, potentially leading to death or long-term sequelae. In vitro models have advanced our knowledge of CM-mediated BBB disruption, but their physiological relevance remains uncertain. Using human induced pluripotent stem cell-derived brain microvascular endothelial cells (hiPSC-BMECs), we aimed to develop a novel in vitro model of the BBB in CM, exhibiting enhanced barrier properties.
    METHODS: hiPSC-BMECs were co-cultured with HB3var03 strain Pf-iRBCs up to 9 h. Barrier integrity was measured using transendothelial electrical resistance (TEER) and sodium fluorescein permeability assays. Localization and expression of tight junction (TJ) proteins (occludin, zonula occludens-1, claudin-5), cellular adhesion molecules (ICAM-1, VCAM-1), and endothelial surface markers (EPCR) were determined using immunofluorescence imaging (IF) and western blotting (WB). Expression of angiogenic and cell stress markers were measured using multiplex proteome profiler arrays.
    RESULTS: After 6-h of co-culture with Pf-iRBCs, hiPSC-BMECs showed reduced TEER and increased sodium fluorescein permeability compared to co-culture with uninfected RBCs, indicative of a leaky barrier. We observed disruptions in localization of occludin, zonula occludens-1, and claudin-5 by IF, but no change in protein expression by WB in Pf-iRBC co-cultures. Expression of ICAM-1 and VCAM-1 but not EPCR was elevated in hiPSC-BMECs with Pf-iRBC co-culture compared to uninfected RBC co-culture. In addition, there was an increase in expression of angiogenin, platelet factor-4, and phospho-heat shock protein-27 in the Pf-iRBCs co-culture compared to uninfected RBC co-culture.
    CONCLUSIONS: These findings demonstrate the validity of our hiPSC-BMECs based model of the BBB, that displays enhanced barrier integrity and appropriate TJ protein localization. In the hiPSC-BMEC co-culture with Pf-iRBCs, reduced TEER, increased paracellular permeability, changes in TJ protein localization, increase in expression of adhesion molecules, and markers of angiogenesis and cellular stress all point towards a novel model with enhanced barrier properties, suitable for investigating pathogenic mechanisms underlying BBB disruption in CM.
    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:在脑型疟疾(CM)中,消耗性凝血病和血栓炎症的特征是突出的。我们假设血栓形成自身抗体有助于CM的促凝血状态。
    方法:来自无并发症疟疾儿童的血浆(UM,n=124)和CM(n=136)通过ELISA分析了一组8种自身抗体,包括抗血小板因子4/聚阴离子(抗PF4/P),抗磷脂,抗磷脂酰丝氨酸,抗髓过氧化物酶,抗蛋白酶3,抗dsDNA,抗β-2-糖蛋白I(β2GPI),和抗心磷脂.非疟疾昏迷(NMC,n=49)和健康对照(HC,n=56)进行比较。使用单变量和逻辑回归分析确定与临床和免疫生物标志物的关联。
    结果:疟疾感染后,相对于HC(P<0.001)和NMC(PF4/P:P<0.001),抗PF4/P和抗PSIgG中位数水平升高。与UM(中位数=0.19,IQR:0.14-0.22,P≤0.0001)相比,CM中的抗PF4/PIgG水平升高(中位数=0.27,IQR:0.19-0.41)。UM和CM之间的抗PSIgG水平没有差异(P=0.39)。当CM病例按疟疾视网膜病变(Ret)状态分层时,抗PF4/PIgG水平与Ret+CM患者外周血血小板计数呈负相关(Rs=0.201,P=0.04),与死亡率呈正相关(OR=15.2,95%CI:1.02~275,P=0.048)。在离体测定中,来自CM患者的血浆相对于来自UM患者的血浆诱导更大的血小板活化能力(P=0.02)。血小板活化与抗PF4/PIgG水平相关(Rs=0.293,P=0.035)。
    结论:抗PF4/P自身抗体升高介导的血栓形成可能是导致CM临床并发症的机制之一。
    BACKGROUNDFeatures of consumptive coagulopathy and thromboinflammation are prominent in cerebral malaria (CM). We hypothesized that thrombogenic autoantibodies contribute to a procoagulant state in CM.METHODSPlasma from children with uncomplicated malaria (UM) (n = 124) and CM (n = 136) was analyzed by ELISA for a panel of 8 autoantibodies including anti-platelet factor 4/polyanion (anti-PF4/P), anti-phospholipid, anti-phosphatidylserine, anti-myeloperoxidase, anti-proteinase 3, anti-dsDNA, anti-β-2-glycoprotein I, and anti-cardiolipin. Plasma samples from individuals with nonmalarial coma (NMC) (n = 49) and healthy controls (HCs) (n = 56) were assayed for comparison. Associations with clinical and immune biomarkers were determined using univariate and logistic regression analyses.RESULTSMedian anti-PF4/P and anti-PS IgG levels were elevated in individuals with malaria infection relative to levels in HCs (P < 0.001) and patients with NMC (PF4/P: P < 0.001). Anti-PF4/P IgG levels were elevated in children with CM (median = 0.27, IQR: 0.19-0.41) compared with those with UM (median = 0.19, IQR: 0.14-0.22, P < 0.0001). Anti-PS IgG levels did not differ between patients with UM and those with CM (P = 0.39). When patients with CM were stratified by malaria retinopathy (Ret) status, the levels of anti-PF4/P IgG correlated negatively with the peripheral platelet count in patients with Ret+ CM (Spearman\'s rho [Rs] = 0.201, P = 0.04) and associated positively with mortality (OR = 15.2, 95% CI: 1.02-275, P = 0.048). Plasma from patients with CM induced greater platelet activation in an ex vivo assay relative to plasma from patients with UM (P = 0.02), and the observed platelet activation was associated with anti-PF4/P IgG levels (Rs= 0.293, P = 0.035).CONCLUSIONSThrombosis mediated by elevated anti-PF4/P autoantibodies may be one mechanism contributing to the clinical complications of CM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号