humanized mice

人源化小鼠
  • 文章类型: Journal Article
    用于概括人类生物系统的人性化小鼠模型仍然存在局限性,例如致命的移植物抗宿主病(GvHD)的发作,可变的成功率,以及全身照射(TBI)的低可及性。最近,已经研究了用CD47-SIRPA轴修饰的小鼠以改善人源化小鼠模型。然而,这种试验很少应用于NOD小鼠。在这项研究中,我们创造了一种新的老鼠品系,NOD-CD47nullRag2nullIL-2Rγnull(RTKO)小鼠,并将其应用于产生人源化小鼠。
    用TBI或白消安(BSF)注射预处理的四周龄雌性NOD-Rag2nullIL-2Rγnull(RID)和RTKO小鼠用于产生人CD34造血干细胞(HSC)移植的人源化小鼠。每周观察两次临床体征,每周测量一次体重。以4周或2周的间隔进行人白细胞抗原的流式细胞术。在HSC注射后48周处死小鼠。
    移植后16至40周,hCD45的百分比在所有组中大多保持在25%以上,在RTKOBSF组中持续时间最长,最高。人白细胞的重建,包括hCD3在内,在RTKOBSF组中也最为突出。在所有组中,只有两只小鼠在移植后40周前死亡,除了死亡的小鼠,没有危及生命的GvHD病变。GvHD的发生已被鉴定为主要归因于人T细胞浸润组织及其相关细胞因子。
    在本研究中应用的所有条件下的人源化小鼠模型被认为是基于人类白细胞重建的改善和稳定的动物健康的长期实验的合适模型。尤其是,用BSF预处理的RTKO小鼠有望成为不仅用于产生人源化小鼠而且用于各种免疫研究领域的有价值的平台。
    UNASSIGNED: Humanized mouse models to recapitulate human biological systems still have limitations, such as the onset of lethal graft-versus-host disease (GvHD), a variable success rate, and the low accessibility of total body irradiation (TBI). Recently, mice modified with the CD47-SIRPA axis have been studied to improve humanized mouse models. However, such trials have been rarely applied in NOD mice. In this study, we created a novel mouse strain, NOD-CD47nullRag2nullIL-2rγnull (RTKO) mice, and applied it to generate humanized mice.
    UNASSIGNED: Four-week-old female NOD-Rag2nullIL-2rγnull (RID) and RTKO mice pre-conditioned with TBI or busulfan (BSF) injection were used for generating human CD34+ hematopoietic stem cell (HSC) engrafted humanized mice. Clinical signs were observed twice a week, and body weight was measured once a week. Flow cytometry for human leukocyte antigens was performed at intervals of four weeks or two weeks, and mice were sacrificed at 48 weeks after HSC injection.
    UNASSIGNED: For a long period from 16 to 40 weeks post transplantation, the percentage of hCD45 was mostly maintained above 25% in all groups, and it was sustained the longest and highest in the RTKO BSF group. Reconstruction of human leukocytes, including hCD3, was also most prominent in the RTKO BSF group. Only two mice died before 40 weeks post transplantation in all groups, and there were no life-threatening GvHD lesions except in the dead mice. The occurrence of GvHD has been identified as mainly due to human T cells infiltrating tissues and their related cytokines.
    UNASSIGNED: Humanized mouse models under all conditions applied in this study are considered suitable models for long-term experiments based on the improvement of human leukocytes reconstruction and the stable animal health. Especially, RTKO mice pretreated with BSF are expected to be a valuable platform not only for generating humanized mice but also for various immune research fields.
    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
    细菌感染继续在全球范围内构成重大的医疗保健负担。每年造成相当大的死亡率和发病率。多重耐药菌株的出现持续上升,对控制全球疾病爆发构成严重风险。开发新的和更有效的治疗和疫苗接种计划,需要临床相关的小动物模型。由于多种细菌物种对多种毒力因子和毒素具有人类特异性的向性,传统的小鼠模型不能完全代表人类疾病。几种人类疾病特征表型,如结核分枝杆菌感染的肺肉芽肿,在标准小鼠模型中不存在。或者,某些病原体,如肠沙门氏菌伤寒和金黄色葡萄球菌,在小鼠中可以很好地耐受并迅速清除。为了解决这个问题,多个小组开发了人源化小鼠模型,并观察到对感染的易感性增强,以及对人类疾病的更忠实的概述。在过去的二十年里,已经开发了多种人源化小鼠模型,以尝试在小动物模型中概括人类免疫系统。在这次审查中,我们首先讨论了能够植入人体组织的免疫缺陷小鼠的历史以及该领域目前使用的植入方法。然后,我们重点介绍了人源化小鼠模型如何成功地揭示了对各种细菌感染的关键人类免疫反应。包括伤寒沙门氏菌,结核分枝杆菌,和金黄色葡萄球菌。
    Bacterial infections continue to represent a significant healthcare burden worldwide, causing considerable mortality and morbidity every year. The emergence of multidrug-resistant bacterial strains continues to rise, posing serious risks to controlling global disease outbreaks. To develop novel and more effective treatment and vaccination programs, there is a need for clinically relevant small animal models. Since multiple bacterial species have human-specific tropism for numerous virulence factors and toxins, conventional mouse models do not fully represent human disease. Several human disease characteristic phenotypes, such as lung granulomas in the case of Mycobacterium tuberculosis infections, are absent in standard mouse models. Alternatively, certain pathogens, such as Salmonella enterica serovar typhi and Staphylococcus aureus, can be well tolerated in mice and cleared quickly. To address this, multiple groups have developed humanized mouse models and observed enhanced susceptibility to infection and a more faithful recapitulation of human disease. In the last two decades, multiple humanized mouse models have been developed to attempt to recapitulate the human immune system in a small animal model. In this review, we first discuss the history of immunodeficient mice that has enabled the engraftment of human tissue and the engraftment methods currently used in the field. We then highlight how humanized mouse models successfully uncovered critical human immune responses to various bacterial infections, including Salmonella enterica serovar Typhi, Mycobacterium tuberculosis, and Staphylococcus aureus.
    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
    近年来,病毒学,病态,和免疫学研究需要进行新兴的抗人类免疫缺陷病毒(HIV)疗法,如基因治疗,广泛中和抗体,和衍生的嵌合抗原受体(CAR)-T免疫疗法,这需要合适的,简单,以及用于准确定量HIV-1感染者中病毒基因组的廉价小动物模型和方法。在我们的研究中,通过双重标记的HIV假病毒感染对HIV-ΔENV-Jurkat-EGFP-mCherry细胞系进行了工程改造。建立了以细胞基因组为整合标准的巢式定量PCR(巢式qPCR)方法,用于定量HIV前病毒拷贝。我们对NOD/Prkdcscid/IL2rgnull(NPG)小鼠进行了健康人外周血单核细胞(PBMC)的静脉注射。要验证植入动力学,我们分析了hCD45+的百分比,hCD3+,hCD4+,hu-PBMC-NPG小鼠外周血中的hCD8+细胞。为了评估hu-PBMC-NPG小鼠中的HIV-1感染,我们通过腹膜内(IP)注射给这些小鼠接种HIVNL4-3-NanoLuc。然后,我们通过小动物成像系统监测荧光素酶表达,并通过qPCR测量脾脏中的病毒载量。静脉注射后3-5周检测小鼠体内人PBMC的浸润,在IP接种后5周,人源化小鼠PBMC中hCD45的百分比超过25%。感染27天后通过荧光素酶成像检测病毒相关荧光素酶蛋白的表达。此外,病毒的总DNA,RNA,前病毒DNA拷贝达到18000拷贝/106个细胞,15000拷贝/μgRNA,和15000个拷贝/106个细胞,分别,在老鼠的脾脏里.一起来看,我们报道了一种简便的方法,通过静脉注射hu-PBMC,无需先进的手术技巧和辐射,即可建立简单的人源化HuPBMC-NPG/严重联合免疫缺陷(SCID)小鼠模型。此外,我们建立了一种简便的方法来有效测定前病毒DNA,以评估体内HIV的复制,病毒库大小,包括CAR-T免疫疗法和基因治疗在内的新型抗HIV疗法的疗效。
    In recent years, virological, pathological, and immunological studies need to be carried out for the emerging anti-human immunodeficiency virus (HIV) therapies such as gene therapy, broadly neutralizing antibodies, and the derived chimeric antigen receptor (CAR)-T immunotherapy, which necessitates suitable, simple, and inexpensive small-animal models and methods for accurate quantification of the viral genome in the HIV-1 infected. In our research, the HIV-∆ENV-Jurkat-EGFP-mCherry cell line was engineered through the infection with a dual-labelled HIV pseudovirus. A nested quantitative PCR (nested-qPCR) method with the cellular genome as the integrated standard was established for the quantification of HIV proviral copies. We administered intravenous injections of healthy human peripheral blood mononuclear cell (PBMC) into NOD/Prkdcscid/IL2rgnull (NPG) mice. To verify engraftment kinetics, we analyzed the percentages of hCD45+, hCD3+, hCD4+, and hCD8+ cells in the peripheral blood of hu-PBMC-NPG mice. To evaluate HIV-1 infection in hu-PBMC-NPG mice, we inoculated these mice with HIV NL4-3-NanoLuc by intraperitoneal (IP) injection. We then monitored the luciferase expression by the small animal imaging system and measured the viral load in the spleen by qPCR. The infiltration of human PBMCs in mice was detected 3-5 weeks after intravenous injection, and the percentage of hCD45 in humanized mouse PBMCs were more than 25% five weeks after IP inoculation. The expression of the virus-associated luciferase protein was detected by luciferase imaging 27 days post infection. Moreover, the viral total DNA, RNA, and proviral DNA copies reached 18 000 copies/106 cells, 15 000 copies/μg RNA, and 15 000 copies/106 cells, respectively, in the mouse spleen. Taken together, we reported a convenient method for building a simple humanized mouse model of HuPBMC-NPG/severe combined immunodeficiency (SCID) by intravenous injection with hu-PBMCs without advanced surgical skills and irradiation. Furthermore, we established a convenient method for the efficient determination of proviral DNA to assess HIV replication in vivo, viral reservoir sizes, and efficacy of novel anti-HIV therapies including CAR-T immunotherapy and gene therapy.
    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
    HBV共价闭合环状DNA(cccDNA)通过作为病毒RNA转录的模板,在乙型肝炎病毒(HBV)感染的持久性中起着重要作用。为了治愈HBV感染,预计cccDNA需要消除或沉默。因此,精确的cccDNA定量是必不可少的。样品制备对于特异性检测cccDNA至关重要。Southern印迹被认为是特异性cccDNA检测的“金标准”,但缺乏敏感性。这里,我们描述了一种快速可靠的基于套件的修改,HBV无蛋白DNA提取方法以及使用分支DNA技术检测细胞培养和肝组织样品中HBVDNA的新型增强灵敏度Southern印迹。它对HBV分子生物学和抗病毒研究都很有用。
    HBV covalently closed circular DNA (cccDNA) plays an important role in the persistence of hepatitis B virus (HBV) infection by serving as the template for transcription of viral RNAs. To cure HBV infection, it is expected that cccDNA needs either to be eliminated or silenced. Hence, precise cccDNA quantification is essential. Sample preparation is crucial to specifically detect cccDNA. Southern blot is regarded as the \"gold standard\" for specific cccDNA detection but lacks sensitivity. Here, we describe a rapid and reliable modified kit-based, HBV protein-free DNA extraction method as well as a novel enhanced sensitivity Southern blot that uses branched DNA technology to detect HBV DNA in cell culture and liver tissue samples. It is useful for both HBV molecular biology and antiviral research.
    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
    广泛中和抗体(bNAb)已显示出预防和治疗HIV感染的巨大前景。bNAb中和的广度,在不同病毒分离株的面板上进行体外测量,通常用作临床潜力的预测因子。然而,最近的预防研究表明,广泛而有效的bNAb(VRC01)的临床疗效被循环菌株的中和抗性所破坏。使用HIV感染的人源化小鼠,我们发现,作为载体免疫疗法(VIT)递送的bNAb的治疗功效是在病毒逃逸过程中出现的突变的适应性成本和抗性益处的函数,我们称之为“escability”。应用这个机械框架,我们发现包膜V5环的序列改变了逃逸过程中出现的突变体的抗性益处,从而影响VIT介导的病毒抑制的治疗功效。我们还发现,基于恩曲他滨的抗逆转录病毒药物方案显着增强了VIT的疗效,通过降低逃生路径上突变体的适应性。我们的发现表明,bNAb的性能是在具有最大功效的抗体方案的合理设计中需要考虑的关键决定因素,并说明了将病毒从现有bNAb中逃逸的方法。
    Broadly neutralizing antibodies (bNAbs) have shown great promise for prevention and treatment of HIV infection. Breadth of bNAb neutralization, measured in vitro across panels of diverse viral isolates, is often used as a predictor of clinical potential. However, recent prevention studies demonstrate that the clinical efficacy of a broad and potent bNAb (VRC01) is undermined by neutralization resistance of circulating strains. Using HIV-infected humanized mice, we find that therapeutic efficacy of bNAbs delivered as Vectored ImmunoTherapy (VIT) is a function of both the fitness cost and resistance benefit of mutations that emerge during viral escape, which we term \'escapability\'. Applying this mechanistic framework, we find that the sequence of the envelope V5-loop alters the resistance benefits of mutants that arise during escape, thereby impacting the therapeutic efficacy of VIT-mediated viral suppression. We also find that an emtricitabine-based antiretroviral drug regimen dramatically enhances the efficacy of VIT, by reducing the fitness of mutants along the escape path. Our findings demonstrate that bNAb escapability is a key determinant to consider in the rational design of antibody regimens with maximal efficacy and illustrates a tractable means of minimizing viral escape from existing bNAbs.
    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
    恶性疟原虫会引起严重的疟疾,并在受感染的红细胞的寄生液泡膜(PVM)上组装蛋白质转位(PTEX)复合物,数百种蛋白质被出口以促进生长。感染的前一个肝脏阶段涉及肝细胞衍生的PVM的生长;然而,在恶性疟原虫肝脏感染期间,蛋白质输出的重要性仍未被探索。这里,我们使用FlpL/FRT系统有条件切除恶性疟原虫子孢子中的基因,用于功能性肝脏阶段研究。PTEX成员ptex150和exp2的破坏不会影响蚊子的子孢子发育或肝细胞的感染性,但会减弱人源化小鼠的肝脏阶段生长。虽然PTEX150缺乏使感染后第6天的适应性降低了40%,EXP2缺乏导致100%的肝脏寄生虫损失,证明PTEX成分在肝细胞中生长到不同程度是必需的。为了表征PTEX功能丧失突变,我们定位了四种肝期疟原虫输出元件(PEXEL)蛋白。恶性疟原虫肝脏特异性蛋白2(LISP2),肝期抗原3(LSA3),环子孢子蛋白(CSP),和一个伯氏疟原虫LISP2报告者都位于恶性疟原虫肝脏阶段的外围,但没有出口到PVM之外。在ptex150-FRT和exp2-FRT肝脏阶段,LISP2和CSP而不是LSA3的表达降低,提示某些PEXEL蛋白的表达受到PTEX破坏的直接或间接影响。这些结果表明,PTEX150和EMP2对于肝细胞中的恶性疟原虫发育是重要的,并且强调了PEXEL蛋白运输的新兴复杂性。
    Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.
    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:自从联合抗逆转录病毒疗法(cART)引入以来,由于在cART中使用的许多药物进入中枢神经系统(CNS)的渗透性相对较低,因此脑已经成为重要的人类免疫缺陷病毒(HIV)储库。鉴于直接评估HIV感染者(PLWH)大脑中急性HIV感染的固有局限性,动物模型,比如人性化的老鼠模型,提供了最有效的方法来研究不同病毒株的作用及其对CNS中HIV感染的影响。在人源化骨髓/肝脏/胸腺(BLT)小鼠模型中评估HIV-1感染期间的CNS病理学,对五个中枢神经系统区域进行了组织学分析,包括额叶皮层,海马体,纹状体,小脑,和脊髓,描绘神经元(MAP2ab,神经)和神经炎症(GFAP,Iba-1)在感染后2周和8周后由两种病毒株诱导的变化。
    结果:研究结果表明,感染HIV的BLT小鼠的大脑中有感染HIV的人类细胞,证明了HIV的神经入侵.Further,两种病毒株,HIV-1JR-CSF和HIV-1CH040在两个时间点感染HIV后,在所有CNS区域诱导神经元损伤和星形胶质增生,如MAP2ab的减少和GFAP荧光信号的增加所证明的,分别。重要的是,与HIV-1CH040感染相比,HIV-1JR-CSF感染对特定CNS区域的神经元健康有更突出的影响,随着NeuN+神经元数量的减少,特别是在额叶皮层。另一方面,感染HIV-1CH040对神经炎症表现出更突出的作用,通过GFAP信号的增加和/或Iba-1+小胶质细胞数量的增加来评估,在CNS地区。
    结论:这些发现表明,在急性HIV感染期间,中枢神经系统的病理分布很普遍。然而,中枢神经系统中的神经元损失和神经炎症的程度取决于菌株,表明HIV菌株会引起不同的中枢神经系统病理。
    BACKGROUND: Since the introduction of combination antiretroviral therapy (cART) the brain has become an important human immunodeficiency virus (HIV) reservoir due to the relatively low penetration of many drugs utilized in cART into the central nervous system (CNS). Given the inherent limitations of directly assessing acute HIV infection in the brains of people living with HIV (PLWH), animal models, such as humanized mouse models, offer the most effective means of studying the effects of different viral strains and their impact on HIV infection in the CNS. To evaluate CNS pathology during HIV-1 infection in the humanized bone marrow/liver/thymus (BLT) mouse model, a histological analysis was conducted on five CNS regions, including the frontal cortex, hippocampus, striatum, cerebellum, and spinal cord, to delineate the neuronal (MAP2ab, NeuN) and neuroinflammatory (GFAP, Iba-1) changes induced by two viral strains after 2 weeks and 8 weeks post-infection.
    RESULTS: Findings reveal HIV-infected human cells in the brain of HIV-infected BLT mice, demonstrating HIV neuroinvasion. Further, both viral strains, HIV-1JR-CSF and HIV-1CH040, induced neuronal injury and astrogliosis across all CNS regions following HIV infection at both time points, as demonstrated by decreases in MAP2ab and increases in GFAP fluorescence signal, respectively. Importantly, infection with HIV-1JR-CSF had more prominent effects on neuronal health in specific CNS regions compared to HIV-1CH040 infection, with decreasing number of NeuN+ neurons, specifically in the frontal cortex. On the other hand, infection with HIV-1CH040 demonstrated more prominent effects on neuroinflammation, assessed by an increase in GFAP signal and/or an increase in number of Iba-1+ microglia, across CNS regions.
    CONCLUSIONS: These findings demonstrate that CNS pathology is widespread during acute HIV infection. However, neuronal loss and the magnitude of neuroinflammation in the CNS is strain dependent indicating that strains of HIV cause differential CNS 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
    人类造血干细胞(HSC)转移的人源化小鼠是探索人类血液学和免疫学的有价值的模型。然而,在小鼠中充分重述人造血需要大量富集的人CD34+HSC和全身照射才能充分植入。最近,我们产生了在c-kit酪氨酸激酶结构域具有点突变的NOG小鼠品系(W41突变体;NOGW小鼠).在这项研究中,我们检查了NOGW小鼠重建人类造血细胞的能力。受辐照的NOGW小鼠在外周血中显示出较高的人CD45细胞植入水平,即使仅转移了5,000-10,000个CD34HSC。在用20,000-40,000个HSC转移的未照射的NOGW小鼠中也观察到人CD45+细胞的有效移植。与NOG小鼠的BM相比,NOGW小鼠的骨髓(BM)显示出明显更多的移植人HSC或祖细胞(CD34CD38-或CD34CD38-细胞)。此外,我们产生了人细胞因子(白细胞介素-3和粒细胞-巨噬细胞集落刺激因子)转基因NOG-W41(NOGW-EXL)小鼠,以实现人造血细胞充分植入的多谱系重建.未照射的NOGW-EXL小鼠显示出较高的人CD45+和髓系细胞移植水平,特别是粒细胞和血小板/巨核细胞,人CD34+细胞移植后,未照射的NOGW或照射的NOG-EXL小鼠。系列BM移植实验表明,与其他品系相比,NOGW小鼠表现出最高的长期HSC潜力。因此,c-kit突变体NOGW-EXL人源化小鼠代表了HSC转移的人源化小鼠的先进模型,并且由于其高通用性而有望广泛应用。
    Human hematopoietic stem cell (HSC)-transferred humanized mice are valuable models for exploring human hematology and immunology. However, sufficient recapitulation of human hematopoiesis in mice requires large quantities of enriched human CD34+ HSCs and total-body irradiation for adequate engraftment. Recently, we generated a NOG mouse strain with a point mutation in the c-kit tyrosine kinase domain (W41 mutant; NOGW mice). In this study, we examined the ability of NOGW mice to reconstitute human hematopoietic cells. Irradiated NOGW mice exhibited high engraftment levels of human CD45+ cells in the peripheral blood, even when only 5,000-10,000 CD34+ HSCs were transferred. Efficient engraftment of human CD45+ cells was also observed in non-irradiated NOGW mice transferred with 20,000-40,000 HSCs. The bone marrow (BM) of NOGW mice exhibited significantly more engrafted human HSCs or progenitor cells (CD34+CD38- or CD34+CD38+ cells) than the BM of NOG mice. Furthermore, we generated a human cytokine (interleukin-3 and granulocyte-macrophage colony-stimulating factor) transgenic NOG-W41 (NOGW-EXL) mouse to achieve multilineage reconstitution with sufficient engraftment of human hematopoietic cells. Non-irradiated NOGW-EXL mice showed significantly higher engraftment levels of human CD45+ and myeloid lineage cells, particularly granulocytes and platelets/megakaryocytes, than non-irradiated NOGW or irradiated NOG-EXL mice after human CD34+ cell transplantation. Serial BM transplantation experiments revealed that NOGW mice exhibited the highest potential for long-term HSC compared with other strains. Consequently, c-kit mutant NOGW-EXL humanized mice represent an advanced model for HSC-transferred humanized mice and hold promise for widespread applications owing to their high versatility.
    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
    中枢神经系统(CNS)的自身免疫疾病,例如多发性硬化症(MS)仅在当前的实验模型中部分表现出来,并且人源化免疫小鼠的开发对于更好地理解免疫发病机理和治疗方法的测试至关重要。我们描述了一种具有MS几个关键特征的人性化小鼠模型。将来自HLA-DRB1型MS和健康(HI)供体的外周血单核细胞(PBMC)移植到严重免疫缺陷的B2m-NOG小鼠中,并显示出人T和B淋巴细胞的快速植入。接受来自最近/正在进行的Epstein-Barr病毒再激活的MS患者的细胞的小鼠显示出高B细胞移植能力。HLA-DRB1*15(DR15)MS和DR15HI小鼠,不是HLA-DRB1*13MS小鼠,发展了中枢神经系统边界和实质的人类T细胞浸润。DR15MS小鼠在脑和脊髓灰质中独特地发展了炎性病变,自发的,hCD8T细胞病变,EAE免疫小鼠的混合hCD8/hCD4T细胞损伤,不同患者供体之间的定位和严重程度存在差异。该模型进一步发展的主要局限性是单核细胞植入不良和缺乏脱髓鞘,淋巴结组织,和IgG反应。这些结果表明PBMC人源化小鼠代表了用于以患者特异性方法研究MS免疫病理学的有希望的研究工具。
    Autoimmune diseases of the central nervous system (CNS) such as multiple sclerosis (MS) are only partially represented in current experimental models and the development of humanized immune mice is crucial for better understanding of immunopathogenesis and testing of therapeutics. We describe a humanized mouse model with several key features of MS. Severely immunodeficient B2m-NOG mice were transplanted with peripheral blood mononuclear cells (PBMCs) from HLA-DRB1-typed MS and healthy (HI) donors and showed rapid engraftment by human T and B lymphocytes. Mice receiving cells from MS patients with recent/ongoing Epstein-Barr virus reactivation showed high B cell engraftment capacity. Both HLA-DRB1*15 (DR15) MS and DR15 HI mice, not HLA-DRB1*13 MS mice, developed human T cell infiltration of CNS borders and parenchyma. DR15 MS mice uniquely developed inflammatory lesions in brain and spinal cord gray matter, with spontaneous, hCD8 T cell lesions, and mixed hCD8/hCD4 T cell lesions in EAE immunized mice, with variation in localization and severity between different patient donors. Main limitations of this model for further development are poor monocyte engraftment and lack of demyelination, lymph node organization, and IgG responses. These results show that PBMC humanized mice represent promising research tools for investigating MS immunopathology in a patient-specific approach.
    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
    数十年来,研究人员一直未能解开多种症状的海湾战争疾病(GWI)病理并找到有效的治疗方法。在小鼠模型中研究病因的慢性症状持续性和局限性与人类的病因显着不同,这对药物发现和寻找有效的治疗方案构成了挑战。GWI暴露在研究队列中存在显着差异,以上情况使得很难重建与GWI症状病理学非常相似的模型。我们已经使用双重植入策略来重建人类免疫系统以及人类微生物组转移,以创建GWI的人源化小鼠模型。采用全基因组鸟枪测序和血液免疫细胞因子酶联免疫吸附试验(ELISA),我们表明,我们用海湾战争(GW)化学物质处理的双人化小鼠显示出显着改变的肠道微生物组,与GWI的退伍军人队列中报道的相似。结果还显示了相似的细胞因子谱,如IL-1β水平升高,双人源化模型中的IL-6和TNF-R-1,正如以前在人类队列中发现的那样。Further,我们利用一种新型GWI退伍军人粪便微生物群转移方法创建了第二种替代模型,该模型与GWI退伍军人的微生物组和免疫系统相关病理非常相似.在人源化小鼠中的GWI退伍军人微生物群移植显示了人类微生物组重建和全身性炎症病理,正如白细胞介素1β的增加所反映的那样,6,8(IL-1β,IL-6,IL-8),肿瘤坏死因子受体1(TNF-R-1),和内毒素血症。总之,虽然是初步的,我们报道了一种新的体内模型,该模型具有人类微生物组重建和移植的人类免疫表型,可能有助于更好地了解GWI中的肠道-免疫相互作用.
    Unraveling the multisymptomatic Gulf War Illness (GWI) pathology and finding an effective cure have eluded researchers for decades. The chronic symptom persistence and limitations for studying the etiologies in mouse models that differ significantly from those in humans pose challenges for drug discovery and finding effective therapeutic regimens. The GWI exposome differs significantly in the study cohorts, and the above makes it difficult to recreate a model closely resembling the GWI symptom pathology. We have used a double engraftment strategy for reconstituting a human immune system coupled with human microbiome transfer to create a humanized-mouse model for GWI. Using whole-genome shotgun sequencing and blood immune cytokine enzyme linked immunosorbent assay (ELISA), we show that our double humanized mice treated with Gulf War (GW) chemicals show significantly altered gut microbiomes, similar to those reported in a Veteran cohort of GWI. The results also showed similar cytokine profiles, such as increased levels of IL-1β, IL-6, and TNF R-1, in the double humanized model, as found previously in a human cohort. Further, a novel GWI Veteran fecal microbiota transfer was used to create a second alternative model that closely resembled the microbiome and immune-system-associated pathology of a GWI Veteran. A GWI Veteran microbiota transplant in humanized mice showed a human microbiome reconstitution and a systemic inflammatory pathology, as reflected by increases in interleukins 1β, 6, 8 (IL-1β, IL-6, IL-8), tumor necrosis factor receptor 1 (TNF R-1), and endotoxemia. In conclusion, though preliminary, we report a novel in vivo model with a human microbiome reconstitution and an engrafted human immune phenotype that may help to better understand gut-immune interactions in GWI.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号