Microphysiological system

微生理系统
  • 文章类型: Journal Article
    这项研究的重点是在微流控细胞研究中使用脉冲电场(PEF)。常用的软光刻材料,聚二甲基硅氧烷(PDMS),不能完全确保这些系统中必要的耐化学性和耐机械性。将特定的分析测量设置集成到微生理系统(MPS)中也具有挑战性。我们提出了一种基于非化学计量的硫醇-烯(OSTE)的微芯片,包含用于PEF和跨上皮电阻(TEER)测量的集成电极以及用于原位监测pH和氧气浓度的设备。MPS的有效性通过C6细胞的PEF处理经验证明。在两种模式下测试PEF处理对细胞活力和对荧光染料DapI的渗透性的影响:停止流动和连续流动。最大磁导率在1.8kV/cm下实现,停流模式下16个脉冲,连续流模式下每个电池64个脉冲。不损害细胞活力。两个集成传感器检测到PEF治疗前后氧气浓度的变化,PEF处理后,pH向碱度移动。因此,我们的概念验证技术作为哺乳动物细胞PEF治疗的MPS,实现原位生理监测。
    This study focuses on the use of pulsed electric fields (PEF) in microfluidics for controlled cell studies. The commonly used material for soft lithography, polydimethylsiloxane (PDMS), does not fully ensure the necessary chemical and mechanical resistance in these systems. Integration of specific analytical measurement setups into microphysiological systems (MPS) are also challenging. We present an off-stoichiometry thiol-ene (OSTE)-based microchip, containing integrated electrodes for PEF and transepithelial electrical resistance (TEER) measurement and the equipment to monitor pH and oxygen concentration in situ. The effectiveness of the MPS was empirically demonstrated through PEF treatment of the C6 cells. The effects of PEF treatment on cell viability and permeability to the fluorescent dye DapI were tested in two modes: stop flow and continuous flow. The maximum permeability was achieved at 1.8 kV/cm with 16 pulses in stop flow mode and 64 pulses per cell in continuous flow mode, without compromising cell viability. Two integrated sensors detected changes in oxygen concentration before and after the PEF treatment, and the pH shifted towards alkalinity following PEF treatment. Therefore, our proof-of-concept technology serves as an MPS for PEF treatment of mammalian cells, enabling in situ physiological monitoring.
    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
    人类心脏组织生长为三维球体,并由源自多能干细胞(hiPSC)的不同心脏细胞类型组成,在体外比标准二维模型更好地概括了人类生理学的各个方面。它们通常由少于5000个细胞组成,用于测量收缩动力学,尽管不是收缩力。相比之下,围绕两个柔性支柱形成的工程心脏组织(EHTs),可以测量收缩力,但传统的EHT通常需要0.5到2百万个细胞。这使得许多EHTs的大规模筛选成本很高。我们在这里的目标是(i)创建一个生理相关的模型,需要比标准EHTs更少的细胞,使它们更便宜,和(ii)确保该小型化模型保持正确的功能。我们证明,完全功能的EHTs可以从生理相关的组合产生hiPSC衍生的心肌细胞(70%),心脏成纤维细胞(15%)和心脏内皮细胞(15%),使用少至1.6×104个细胞。我们的结果表明,这些EHTs在形成后14天内都是可行的和起作用的。EHT可以在0.6和3Hz之间的频率范围内进行电起搏,最佳在0.6和2Hz之间。这在测试的三个缩小的EHT尺寸之间是一致的。这些发现表明,小型化的EHT可以代表一种经济有效的微生理系统,用于疾病建模和检查药物反应,特别是在用于发现药物的二次筛选中。
    Human heart tissues grown as three-dimensional spheroids and consisting of different cardiac cell types derived from pluripotent stem cells (hiPSCs) recapitulate aspects of human physiology better than standard two-dimensional models in vitro. They typically consist of less than 5000 cells and are used to measure contraction kinetics although not contraction force. By contrast, engineered heart tissues (EHTs) formed around two flexible pillars, can measure contraction force but conventional EHTs often require between 0.5 and 2 million cells. This makes large-scale screening of many EHTs costly. Our goals here were (i) to create a physiologically relevant model that required fewer cells than standard EHTs making them less expensive, and (ii) to ensure that this miniaturized model retained correct functionality. We demonstrated that fully functional EHTs could be generated from physiologically relevant combinations of hiPSC-derived cardiomyocytes (70%), cardiac fibroblasts (15%) and cardiac endothelial cells (15%), using as few as 1.6 × 104 cells. Our results showed that these EHTs were viable and functional up to 14 days after formation. The EHTs could be electrically paced in the frequency range between 0.6 and 3 Hz, with the optimum between 0.6 and 2 Hz. This was consistent across three downscaled EHT sizes tested. These findings suggest that miniaturized EHTs could represent a cost-effective microphysiological system for disease modelling and examining drug responses particularly in secondary screens for drug discovery.
    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
    为了道德,经济,和科学原因,动物实验,用于评估化学品在市场上发布之前的潜在神经毒性,需要用新的方法来取代。为了说明新方法方法的使用,人类诱导的多能干细胞衍生的3D模型BrainSpheres急性(48小时)或反复(7天)暴露于胺碘酮(0.625-15µM),一种据报道对神经系统有有害影响的亲脂性抗心律失常药物。使用转录组学评估神经毒性,细胞类型特异性标记的免疫组织化学,以及与脂质代谢有关的各种基因的实时逆转录聚合酶链反应。通过将分布动力学模型与神经毒性读数相结合,我们表明,胺碘酮的神经毒性作用的时间和浓度依赖性增加是由重复给药后胺碘酮的细胞积累驱动的.室性体外分布动力学模型的发展使我们能够预测脑球中细胞相关浓度随时间和不同暴露情况的变化。结果表明,人类细胞本质上对胺碘酮比啮齿动物细胞更敏感。首次在脑细胞中观察到胺碘酮诱导的脂质代谢基因调节。星形胶质细胞似乎是体外最敏感的人脑细胞类型。总之,在重复给药人类诱导多能干细胞衍生的BrainSppheres后,结合体外动力学的隔室模型评估不同分子水平的读数,提供了一种评估神经毒性途径和完善人类化学安全性评估的机制手段。
    For ethical, economical, and scientific reasons, animal experimentation, used to evaluate the potential neurotoxicity of chemicals before their release in the market, needs to be replaced by new approach methodologies. To illustrate the use of new approach methodologies, the human induced pluripotent stem cell-derived 3D model BrainSpheres was acutely (48 h) or repeatedly (7 days) exposed to amiodarone (0.625-15 µM), a lipophilic antiarrhythmic drug reported to have deleterious effects on the nervous system. Neurotoxicity was assessed using transcriptomics, the immunohistochemistry of cell type-specific markers, and real-time reverse transcription-polymerase chain reaction for various genes involved in the lipid metabolism. By integrating distribution kinetics modeling with neurotoxicity readouts, we show that the observed time- and concentration-dependent increase in the neurotoxic effects of amiodarone is driven by the cellular accumulation of amiodarone after repeated dosing. The development of a compartmental in vitro distribution kinetics model allowed us to predict the change in cell-associated concentrations in BrainSpheres with time and for different exposure scenarios. The results suggest that human cells are intrinsically more sensitive to amiodarone than rodent cells. Amiodarone-induced regulation of lipid metabolism genes was observed in brain cells for the first time. Astrocytes appeared to be the most sensitive human brain cell type in vitro. In conclusion, assessing readouts at different molecular levels after the repeat dosing of human induced pluripotent stem cell-derived BrainSpheres in combination with the compartmental modeling of in vitro kinetics provides a mechanistic means to assess neurotoxicity pathways and refine chemical safety assessment for humans.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Review
    微生理系统(MPS)是能够模拟组织微环境的2D或3D多细胞构建体。最新的模型包括一系列的技术,包括各种细胞类型的共培养,支架和细胞外基质材料的利用,灌注系统,3D培养方法,3D生物打印,器官芯片技术,和组织结构的检查。在过去的十年中,已经出现了几种人脑3D培养物或脑MPS(BMPS)。这些类器官或球体是源自诱导多能细胞或胚胎干细胞的3D培养系统,其含有神经元和神经胶质群体,并概括人脑的结构和生理方面。BMPS最近已被引入神经感染性疾病的研究和建模中,并已被证明可用于建立病毒感染的神经嗜性。感染所需的细胞-病原体相互作用,评估细胞病理学影响,基因组和蛋白质组谱,和筛选治疗化合物。在这里,我们回顾了神经传染病中使用的类器官的不同方法,包括球体,引导和非引导方案以及含有小胶质细胞和血脑屏障的模型,它们的具体应用,和限制。该综述概述了包括寨卡在内的特定感染的现有模型,登革热,JC病毒,日本脑炎,麻疹,疱疹,SARS-CoV2和流感病毒等,并在疾病和抗病毒剂筛选的建模中提供有用的概念。
    Microphysiological systems (MPS) are 2D or 3D multicellular constructs able to mimic tissue microenvironments. The latest models encompass a range of techniques, including co-culturing of various cell types, utilization of scaffolds and extracellular matrix materials, perfusion systems, 3D culture methods, 3D bioprinting, organ-on-a-chip technology, and examination of tissue structures. Several human brain 3D cultures or brain MPS (BMPS) have emerged in the last decade. These organoids or spheroids are 3D culture systems derived from induced pluripotent cells or embryonic stem cells that contain neuronal and glial populations and recapitulate structural and physiological aspects of the human brain. BMPS have been introduced recently in the study and modeling of neuroinfectious diseases and have proven to be useful in establishing neurotropism of viral infections, cell-pathogen interactions needed for infection, assessing cytopathological effects, genomic and proteomic profiles, and screening therapeutic compounds. Here we review the different methodologies of organoids used in neuroinfectious diseases including spheroids, guided and unguided protocols as well as microglia and blood-brain barrier containing models, their specific applications, and limitations. The review provides an overview of the models existing for specific infections including Zika, Dengue, JC virus, Japanese encephalitis, measles, herpes, SARS-CoV2, and influenza viruses among others, and provide useful concepts in the modeling of disease and antiviral agent screening.
    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
    骨关节炎(OA)是一种痛苦和致残的关节疾病,影响着全世界数百万人。缺乏临床相关模型限制了我们在临床试验之前预测治疗结果的能力,大多数药物失败的地方。因此,需要一种能够准确概括人类OA的全关节疾病性质的模型。新兴的微生理系统提供了新的机会。我们最近建立了一个微型膝关节系统,被称为迷你关节,其中使用人骨髓间充质干细胞(hBMSCs)来创建骨软骨复合体,滑膜样纤维组织,和脂肪组织类似物。在这项研究中,我们通过检验与抗炎和软骨诱导剂共同治疗可以抑制关节炎症和相关软骨降解的假设,探索了小关节在开发新型OA治疗中的潜力.具体来说,我们通过用白细胞介素-1β(IL-1β)治疗滑膜样组织,在小关节中创建了一个“滑膜炎”相关的OA模型,然后介绍了抑制核因子κβ(NF-κB)遗传途径和骨形态发生蛋白7(BMP-7)的寡脱氧核苷酸(ODN)的联合治疗。BMP-7和ODN的联合治疗减少了滑膜样纤维组织中的炎症,并显示了骨软骨复合体软骨部分中糖胺聚糖形成的增加。第一次,这项研究证明了小关节在开发改善疾病的OA药物方面的潜力.与NF-κBODN和BMP-7共同治疗的疗效可以在未来的临床研究中进一步验证。
    Osteoarthritis (OA) is a painful and disabling joint disease affecting millions worldwide. The lack of clinically relevant models limits our ability to predict therapeutic outcomes prior to clinical trials, where most drugs fail. Therefore, there is a need for a model that accurately recapitulates the whole-joint disease nature of OA in humans. Emerging microphysiological systems provide a new opportunity. We recently established a miniature knee joint system, known as the miniJoint, in which human bone-marrow-derived mesenchymal stem cells (hBMSCs) were used to create an osteochondral complex, synovial-like fibrous tissue, and adipose tissue analogs. In this study, we explored the potential of the miniJoint in developing novel treatments for OA by testing the hypothesis that co-treatment with anti-inflammation and chondroinducing agents can suppress joint inflammation and associated cartilage degradation. Specifically, we created a \"synovitis\"-relevant OA model in the miniJoint by treating synovial-like tissues with interleukin-1β (IL-1β), and then a combined treatment of oligodeoxynucleotides (ODNs) suppressing the nuclear factor kappa beta (NF-κB) genetic pathway and bone morphogenic protein-7 (BMP-7) was introduced. The combined treatment with BMP-7 and ODNs reduced inflammation in the synovial-like fibrous tissue and showed an increase in glycosaminoglycan formation in the cartilage portion of the osteochondral complex. For the first time, this study demonstrated the potential of the miniJoint in developing disease-modifying OA drugs. The therapeutic efficacy of co-treatment with NF-κB ODNs and BMP-7 can be further validated in future clinical studies.
    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
    疼痛是骨关节炎(OA)的主要症状,其驱使患者寻求医疗护理。目前,没有药物治疗可以逆转或阻止OA的进展.用于长期治疗OA疼痛的安全和有效的药物也不可用。了解OA疼痛在发病时和随时间产生的机制对于开发有效的治疗方法至关重要。在这篇叙述性评论中,我们首先总结一下我们目前对膝关节神经支配的认识,然后讨论目前认为是OA疼痛的分子机制。特别是,我们专注于每个关节成分对疼痛产生的贡献。接下来,总结了目前研究OA疼痛的实验模型,并介绍了评估啮齿动物疼痛的方法。尤其突出了新兴的微生理系统在OA疼痛研究中的潜在应用。最后,我们讨论了当前在标准化模型和选择适当的系统来解决特定问题方面的挑战。
    Pain is the predominant symptom of osteoarthritis (OA) that drives patients to seek medical care. Currently, there are no pharmacological treatments that can reverse or halt the progression of OA. Safe and efficacious medications for long-term management of OA pain are also unavailable. Understanding the mechanisms behind OA pain generation at onset and over time is critical for developing effective treatments. In this narrative review, we first summarize our current knowledge on the innervation of the knee joint, and then discuss the molecular mechanism(s) currently thought to underlie OA pain. In particular, we focus on the contribution of each joint component to the generation of pain. Next, the current experimental models for studying OA pain are summarized, and the methods to assess pain in rodents are presented. The potential application of emerging microphysiological systems in OA pain research is especially highlighted. Lastly, we discuss the current challenge in standardizing models and the selection of appropriate systems to address specific questions.
    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
    氧化应激是由活性氧和氮(ROS/RNS)的产生和解毒之间的不平衡引起的。这种失衡在脑老化和年龄相关的神经退行性疾病中起着重要作用。在帕金森病(PD)的背景下,黑质致密部多巴胺能神经元对氧化应激的敏感性被认为是PD发病的关键因素。在这里,我们研究了不同的氧化应激诱导化合物(6-OHDA,MPTP或MPP)对iPSC衍生的人脑3D模型(又名BrainSpheres)中的多巴胺能神经元群体。用6-OHDA治疗,在分化4周时,MPTP或MPP破坏了脑球中多巴胺能神经元的表型(分别为50、5000、1000μM)。6-OHDA最有效地增加ROS产生并降低线粒体功能。它进一步诱导了与氧化应激和线粒体功能障碍相关的基因表达和代谢产物的最大变化。使用transwell系统将脑球与内皮屏障共培养,可以评估测试化合物的不同渗透能力及其在脑球内多巴胺能神经元中引起的损害。在BrainSphere模型中,用已知在体内诱导PD样表型的化合物处理引起分子缺陷和多巴胺能神经元的损失。因此,这种方法概括了类似高剂量的PD中神经变性过程的常见动物模型。讨论了作为药物发现工具的相关性。
    Oxidative stress is caused by an imbalance between the generation and detoxification of reactive oxygen and nitrogen species (ROS/RNS). This imbalance plays an important role in brain aging and age-related neurodegenerative diseases. In the context of Parkinson\'s disease (PD), the sensitivity of dopaminergic neurons in the substantia nigra pars compacta to oxidative stress is considered a key factor of PD pathogenesis. Here we study the effect of different oxidative stress-inducing compounds (6-OHDA, MPTP or MPP+) on the population of dopaminergic neurons in an iPSC-derived human brain 3D model (aka BrainSpheres). Treatment with 6-OHDA, MPTP or MPP+ at 4 weeks of differentiation disrupted the dopaminergic neuronal phenotype in BrainSpheres at (50, 5000, 1000 μM respectively). 6-OHDA increased ROS production and decreased mitochondrial function most efficiently. It further induced the greatest changes in gene expression and metabolites related to oxidative stress and mitochondrial dysfunction. Co-culturing BrainSpheres with an endothelial barrier using a transwell system allowed the assessment of differential penetration capacities of the tested compounds and the damage they caused in the dopaminergic neurons within the BrainSpheres In conclusion, treatment with compounds known to induce PD-like phenotypes in vivo caused molecular deficits and loss of dopaminergic neurons in the BrainSphere model. This approach therefore recapitulates common animal models of neurodegenerative processes in PD at similarly high doses. The relevance as tool for drug discovery is discussed.
    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
    肺的气体交换单元,肺泡,是机械活动的,并在呼吸过程中经历循环变形。排列肺泡的上皮细胞通过表面活性剂分泌降低表面张力来促进肺功能,受到与呼吸相关的机械提示的高度影响。这些空间异质的机械线索与几种生理和病理生理状态有关。这里,我们描述了微流体辅助肺细胞培养模型的发展,该模型结合了异质循环拉伸来模拟肺泡呼吸运动。使用这个设备,我们研究了呼吸生物力学(与呼吸样运动相关)和应变异质性对肺泡上皮细胞功能的影响.此外,我们评估了该平台在模拟与肺发病机制和呼吸机诱导的肺损伤相关的基质顺应性改变方面的潜在应用.结合人体细胞和动态生物力学的肺微生理平台可以作为描绘肺泡微力学在肺生理和病理结果中的作用的重要工具。
    The gas exchange units of the lung, the alveoli, are mechanically active and undergo cyclic deformation during breathing. The epithelial cells that line the alveoli contribute to lung function by reducing surface tension via surfactant secretion, which is highly influenced by the breathing-associated mechanical cues. These spatially heterogeneous mechanical cues have been linked to several physiological and pathophysiological states. Here, we describe the development of a microfluidically assisted lung cell culture model that incorporates heterogeneous cyclic stretching to mimic alveolar respiratory motions. Employing this device, we have examined the effects of respiratory biomechanics (associated with breathing-like movements) and strain heterogeneity on alveolar epithelial cell functions. Furthermore, we have assessed the potential application of this platform to model altered matrix compliance associated with lung pathogenesis and ventilator-induced lung injury. Lung microphysiological platforms incorporating human cells and dynamic biomechanics could serve as an important tool to delineate the role of alveolar micromechanics in physiological and pathological outcomes in the lung.
    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
    侵袭性肺曲霉病与高死亡率相关,并对免疫功能低下的患者构成直接威胁。这里,我们提出了侵袭性曲霉菌病芯片(IAC)模型来研究烟曲霉的体外感染。该模型可以研究肺泡芯片模型中真菌菌丝从上皮到内皮细胞层的侧向生长和侵袭行为。我们为三维共聚焦显微镜图像建立了基于算法的分析管道,以可视化和量化真菌形态,包括菌丝生长和分枝。IAC模型中的人巨噬细胞部分抑制真菌的生长,促炎细胞因子(IL-1,IL-6,TNF)和趋化因子(IL-8和MCP-1)的释放与侵袭性菌丝数量的增加有关。类似于体内,抑制真菌的药物卡泊芬净的应用限制了真菌的生长,并导致了先前在其他研究中描述的菌丝树的形态变化。IAC感染模型允许细胞感染靶标的鉴定和表征以及临床相关浓度的抗真菌药物的体外测试。因此,它代表了一种有希望的工具,可以扩大对侵袭性曲霉病的致病性和病理生理学的理解。
    Invasive pulmonary aspergillosis is associated with a high mortality rate and poses a direct threat to immunocompromised patients. Here, we present the invasive aspergillosis-on-chip (IAC) model to investigate Aspergillus fumigatus infection in vitro. The model allows the study of the lateral growth and the invasive behaviour of fungal hyphae from the epithelium into the endothelial cell layer in an alveolus-on-chip model. We established an algorithm-based analysis pipeline for three-dimensional confocal microscopy images to visualize and quantify fungal morphology, including hyphal growth and branching. Human macrophages in the IAC model partially inhibited the growth of the fungus, contributed to the release of proinflammatory cytokines (IL-1, IL-6, TNF) and chemokines (IL-8 and MCP-1) associated with an increased number of invasive hyphae. Similar to in vivo, the application of the fungistatic drug caspofungin limited the fungal growth and resulted in morphological changes of the hyphal tree previously described in other studies. The IAC infection model allows the identification and characterization of cellular infection targets and in vitro testing of antifungal drugs in clinically relevant concentrations. It thus represents a promising tool to broaden the understanding of pathogenicity and pathophysiology of invasive aspergillosis.
    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
    进行性多灶性白质脑病(PML)是免疫抑制患者常见的神经系统并发症。PML是由JC病毒(JCV)引起的,一种感染少突胶质细胞和星形胶质细胞的嗜神经DNA多瘤病毒,引起炎症和脱髓鞘,导致神经功能障碍。由于缺乏体外或动物模型来研究疾病机制,因此对PML的发病机理知之甚少,因为病毒最有效地仅感染人类细胞。我们开发了人类来源的脑器官型系统(也称为脑类器官)来模拟JCV感染。该模型是通过使用人类诱导的多能干细胞(iPSC)并在3D中培养它们以生成包含神经元的器官型模型来开发的,星形胶质细胞,以及概括人脑环境的少突胶质细胞。我们用PML患者的JCVMAD4菌株或脑脊液感染了脑类器官。通过qPCR评估类器官的感染证据,免疫荧光,暴露后1、2和3周的电子显微镜检查。通过免疫细胞化学研究证实了JCVMAD4菌株和PMLCSF暴露的脑类器官中的JCV感染,这些研究表明病毒抗原和电子显微镜显示少突胶质细胞和星形胶质细胞的核室中的病毒体颗粒。没有可见神经元感染的证据。在暴露于病毒的类器官及其培养基中,通过JCVqPCR也证实了感染。总之,JCV感染的脑类器官模型建立了一个适用于研究JCV感染机制和PML发病机制的人类模型,可能有助于探索治疗方法。
    Progressive multifocal leukoencephalopathy (PML) is a frequent neurological complication in immunosuppressed patients. PML is caused by the JC virus (JCV), a neurotropic DNA polyomavirus that infects oligodendrocytes and astrocytes, causing inflammation and demyelination which lead to neurological dysfunction. The pathogenesis of PML is poorly understood due to the lack of in vitro or animal models to study mechanisms of disease as the virus most efficiently infects only human cells. We developed a human-derived brain organotypic system (also called brain organoid) to model JCV infection. The model was developed by using human-induced pluripotent stem cells (iPSC) and culturing them in 3D to generate an organotypic model containing neurons, astrocytes, and oligodendrocytes which recapitulates aspects of the environment of the human brain. We infected the brain organoids with the JCV MAD4 strain or cerebrospinal fluid of a patient with PML. The organoids were assessed for evidence of infection by qPCR, immunofluorescence, and electron microscopy at 1, 2, and 3 weeks post-exposure. JCV infection in both JCV MAD4 strain and PML CSF-exposed brain organoids was confirmed by immunocytochemical studies demonstrating viral antigens and electron microscopy showing virion particles in the nuclear compartment of oligodendrocytes and astrocytes. No evidence of neuronal infection was visualized. Infection was also demonstrated by JCV qPCR in the virus-exposed organoids and their media. In conclusion, the brain organoid model of JCV infection establishes a human model suitable for studying the mechanisms of JCV infection and pathogenesis of PML and may facilitate the exploration of therapeutic approaches.
    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