organ‐on‐chip

  • 文章类型: Journal Article
    微生理系统(MPS)重建组织界面和器官功能,在药物开发中提出了一种有希望的动物模型替代品。然而,传统材料如聚二甲基硅氧烷(PDMS)通常通过吸收疏水分子来干扰,影响药物测试的准确性。增材制造,包括3D生物打印,提供可行的解决方案。GlioFlow3D,介绍了一种结合挤出生物打印和立体光刻(SLA)的新型微流体平台。GlioFlow3D在基于水凝胶的模拟脉管系统的微通道中整合了原代人细胞和胶质母细胞瘤(GBM)系,在SLA树脂框架内使用具有成本效益的材料。该研究引入了一种稳健的方案来减轻SLA树脂的细胞毒性。与PDMS相比,GlioFlow3D展示了较低的小分子吸收,这与诸如替莫唑胺(TMZ)之类的小分子的准确测试有关。计算建模用于优化无泵设置,以模拟组织中的间质流体流动动力学。在GlioFlow3D中共培养GBM与脑内皮细胞显示增强的CD133表达和血管界面附近的TMZ阻力,突出空间耐药机制。这个无PDMS平台承诺进行先进的药物测试,通过阐明受组织微环境影响的复杂GBM耐药机制,改善临床前研究和个性化治疗。
    Microphysiological systems (MPSs) reconstitute tissue interfaces and organ functions, presenting a promising alternative to animal models in drug development. However, traditional materials like polydimethylsiloxane (PDMS) often interfere by absorbing hydrophobic molecules, affecting drug testing accuracy. Additive manufacturing, including 3D bioprinting, offers viable solutions. GlioFlow3D, a novel microfluidic platform combining extrusion bioprinting and stereolithography (SLA) is introduced. GlioFlow3D integrates primary human cells and glioblastoma (GBM) lines in hydrogel-based microchannels mimicking vasculature, within an SLA resin framework using cost-effective materials. The study introduces a robust protocol to mitigate SLA resin cytotoxicity. Compared to PDMS, GlioFlow3D demonstrated lower small molecule absorption, which is relevant for accurate testing of small molecules like Temozolomide (TMZ). Computational modeling is used to optimize a pumpless setup simulating interstitial fluid flow dynamics in tissues. Co-culturing GBM with brain endothelial cells in GlioFlow3D showed enhanced CD133 expression and TMZ resistance near vascular interfaces, highlighting spatial drug resistance mechanisms. This PDMS-free platform promises advanced drug testing, improving preclinical research and personalized therapy by elucidating complex GBM drug resistance mechanisms influenced by the tissue microenvironment.
    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
    银河宇宙辐射(GCR)是宇航员在执行月球和火星任务期间面临的最严重风险之一。能够概括人体生理的实验模型对于理解辐射对人体器官的影响以及制定针对太空旅行暴露的辐射防护措施至关重要。使用包含人类骨髓(造血和急性辐射损伤部位)的工程组织模型的多器官芯片(multi-OoC)平台研究了全身辐射的影响,心肌(慢性辐射损伤部位)和肝脏(代谢部位),通过血管循环与内皮屏障相连,将各个组织腔与血管灌注液分开。在长期的中子辐射之后,深空中最具破坏性的辐射成分,与急性递送的相同累积剂量相比,观察到更大的组织功能偏差。Further,通过表征循环中的工程骨髓(eBM)衍生的免疫细胞,鉴定了58个特定于延长中子剂量效应的独特基因,与急性照射和健康组织相比。它提出,这个生物工程平台允许在“宇航员芯片”模型中研究人类对延长辐射暴露的反应,该模型可以为减轻宇宙辐射伤害的措施提供信息。
    Galactic cosmic radiation (GCR) is one of the most serious risks posed to astronauts during missions to the Moon and Mars. Experimental models capable of recapitulating human physiology are critical to understanding the effects of radiation on human organs and developing radioprotective measures against space travel exposures. The effects of systemic radiation are studied using a multi-organ-on-a-chip (multi-OoC) platform containing engineered tissue models of human bone marrow (site of hematopoiesis and acute radiation damage), cardiac muscle (site of chronic radiation damage) and liver (site of metabolism), linked by vascular circulation with an endothelial barrier separating individual tissue chambers from the vascular perfusate. Following protracted neutron radiation, the most damaging radiation component in deep space, a greater deviation of tissue function is observed as compared to the same cumulative dose delivered acutely. Further, by characterizing engineered bone marrow (eBM)-derived immune cells in circulation, 58 unique genes specific to the effects of protracted neutron dosing are identified, as compared to acutely irradiated and healthy tissues. It propose that this bioengineered platform allows studies of human responses to extended radiation exposure in an \"astronaut-on-a-chip\" model that can inform measures for mitigating cosmic radiation injury.
    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
    纤维化,其主要标志是过度的细胞外基质(ECM)沉积,是与许多疾病相关的病理生理过程,最终导致器官功能障碍和患者预后不良。尽管纤维化的患病率很高,目前很少有治疗选择,而且重要的是,缺乏准确研究纤维化的体外模型。这篇综述从发展器官芯片(OoC)疾病模型的角度讨论了纤维化的多面性,专注于五个关键特征:ECM组件,炎症,机械提示,缺氧,和血管化。在研究纤维化疾病的背景下,探索了OoC技术的潜力,以更好地对这些特征进行建模,并强调了各种关键特征之间的相互作用。本文回顾了如何在OoC平台上对器官特异性纤维化疾病进行建模,这些现有模型中包含哪些元素,并强调了新颖研究方向的途径。最后,这篇综述最后提出了如何解决目前在纳入多个特征以OoC格式产生更复杂和相关的纤维化疾病模型方面的差距的观点.
    Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
    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
    组织稳态和疾病状态依赖于通过血管生成发芽形成新血管,受周围细胞外基质的特性严格调节。虽然物理线索,如基体刚度或降解性,已经发展成为组织微环境中细胞功能的主要调节因子,目前尚不清楚物理线索是否以及如何在血管生成过程中调节内皮细胞迁移。为了调查这一点,在可调的合成水凝胶内形成血管生成发芽的仿生模型。结果表明,内皮细胞感知周围基质对蛋白水解裂解的抵抗力,并通过调节其迁移表型做出反应。电阻细胞遭遇受到共价基质交联的数量的影响,交联降解性,和细胞的蛋白水解活性。当基体电阻较高时,细胞从集体转变为依赖于肌动球蛋白收缩性的单细胞迁移模式。这种集体转换伴随着肌动蛋白细胞骨架的重大重组,应力纤维不再可见,和F-肌动蛋白聚集在大的点状簇中。基质抗性被确定为先前未知的血管生成发芽调节剂,因此,提供了基质的物理性质通过细胞骨架重塑影响细胞迁移模式的机制。
    Tissue homeostasis and disease states rely on the formation of new blood vessels through angiogenic sprouting, which is tightly regulated by the properties of the surrounding extracellular matrix. While physical cues, such as matrix stiffness or degradability, have evolved as major regulators of cell function in tissue microenvironments, it remains unknown whether and how physical cues regulate endothelial cell migration during angiogenesis. To investigate this, a biomimetic model of angiogenic sprouting inside a tunable synthetic hydrogel is created. It is shown that endothelial cells sense the resistance of the surrounding matrix toward proteolytic cleavage and respond by adjusting their migration phenotype. The resistance cells encounter is impacted by the number of covalent matrix crosslinks, crosslink degradability, and the proteolytic activity of cells. When matrix resistance is high, cells switch from a collective to an actomyosin contractility-dependent single cellular migration mode. This switch in collectivity is accompanied by a major reorganization of the actin cytoskeleton, where stress fibers are no longer visible, and F-actin aggregates in large punctate clusters. Matrix resistance is identified as a previously unknown regulator of angiogenic sprouting and, thus, provides a mechanism by which the physical properties of the matrix impact cell migration modes through cytoskeletal remodeling.
    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
    人体微生物组显着影响通过肠-肝轴的药物代谢,导致改变的药物反应和潜在的毒性。由于人类肠道环境的复杂性,我们对微生物群对这些过程的影响的理解是有限的。为了解决这个问题,我们引入了一个多器官芯片(MOoC)平台,该平台结合了人类微生物串扰(HuMiX)芯片上肠道(GoC)和Dynamic42芯片上肝脏(LoC),模拟肠道和肝脏之间的双向互连,称为肠-肝轴。该平台支持肠和肝细胞的活力和功能。在概念验证研究中,我们复制了伊立替康的代谢,一种广泛使用的结直肠癌药物,在我们的MOOC内。利用液相色谱与串联质谱联用(LC-MS/MS),我们追踪了伊立替康代谢物,确认平台代表沿着肠-肝轴的药物代谢的能力。此外,使用我们的肠道-肝脏平台,我们发现结直肠癌相关的肠道细菌,大肠杆菌,通过将其非活性代谢物SN-38G转化为毒性代谢物SN-38来修饰伊立替康的代谢。这个平台是一个强大的工具,用于研究肠道微生物和药物之间的复杂相互作用。提供动物模型的代表性替代方案,并提供新的药物开发策略。本文受版权保护。保留所有权利。
    The human microbiome significantly influences drug metabolism through the gut-liver axis, leading to modified drug responses and potential toxicity. Due to the complex nature of the human gut environment, the understanding of microbiome-driven impacts on these processes is limited. To address this, a multiorgan-on-a-chip (MOoC) platform that combines the human microbial-crosstalk (HuMiX) gut-on-chip (GoC) and the Dynamic42 liver-on-chip (LoC), mimicking the bidirectional interconnection between the gut and liver known as the gut-liver axis, is introduced. This platform supports the viability and functionality of intestinal and liver cells. In a proof-of-concept study, the metabolism of irinotecan, a widely used colorectal cancer drug, is imitated within the MOoC. Utilizing liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS), irinotecan metabolites are tracked, confirming the platform\'s ability to represent drug metabolism along the gut-liver axis. Further, using the authors\' gut-liver platform, it is shown that the colorectal cancer-associated gut bacterium, Escherichia coli, modifies irinotecan metabolism through the transformation of its inactive metabolite SN-38G into its toxic metabolite SN-38. This platform serves as a robust tool for investigating the intricate interplay between gut microbes and pharmaceuticals, offering a representative alternative to animal models and providing novel drug development strategies.
    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
    尽管淋巴管生成在发育过程中和对组织再生有影响的几种疾病中起着至关重要的作用,豁免权,和癌症,相对于血管生成,了解这一过程的工具明显较少。虽然在用微生理系统模拟血管生成方面有很大的进展,它们尚未经过严格的优化或标准化,以重新创建淋巴管生成的动力学。这里,设计了淋巴管生成芯片(L-Chip),在其中,新的芽形成和成熟取决于间质流的强加,生长因子梯度,和用生长因子预处理内皮细胞。L-Chip揭示了淋巴管生成的这些机械和生化决定因素的独立和组合作用,因此最终导致芽从亲本血管中出现,并在4天内成熟成长度达1毫米的管状结构,超过现有技术。Further,当预处理鸡尾酒和用于启动和促进淋巴管生成的生长因子鸡尾酒的组成被解剖时,已发现,相对于血管生成,内皮蛋白聚糖(ESM-1)导致更显性的淋巴管生成。因此,L-Chip为标准化针对淋巴管生成的微流体测定以及加速其与血管生成同等的基础和转化科学提供了基础。
    Despite the crucial role of lymphangiogenesis during development and in several diseases with implications for tissue regeneration, immunity, and cancer, there are significantly fewer tools to understand this process relative to angiogenesis. While there has been a major surge in modeling angiogenesis with microphysiological systems, they have not been rigorously optimized or standardized to enable the recreation of the dynamics of lymphangiogenesis. Here, a Lymphangiogenesis-Chip (L-Chip) is engineered, within which new sprouts form and mature depending upon the imposition of interstitial flow, growth factor gradients, and pre-conditioning of endothelial cells with growth factors. The L-Chip reveals the independent and combinatorial effects of these mechanical and biochemical determinants of lymphangiogenesis, thus ultimately resulting in sprouts emerging from a parent vessel and maturing into tubular structures up to 1 mm in length within 4 days, exceeding prior art. Further, when the constitution of the pre-conditioning cocktail and the growth factor cocktail used to initiate and promote lymphangiogenesis are dissected, it is found that endocan (ESM-1) results in more dominant lymphangiogenesis relative to angiogenesis. Therefore, The L-Chip provides a foundation for standardizing the microfluidics assays specific to lymphangiogenesis and for accelerating its basic and translational science at par with angiogenesis.
    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
    中枢神经系统和周围神经系统的神经回路的改变与神经退行性疾病(NDD)的发作密切相关。尽管有大量的研究工作,关于NDD病理过程的知识,由于无法获取和复制神经系统的成分及其复杂的微环境,有效药物的开发仍然受到限制。2D培养系统过于简单而不能准确地表示体内细胞的更复杂和动态的情况,因此已经被3D系统所超越。然而,这两种模型都受到各种限制,可以通过采用两种创新技术来克服:芯片上器官和3D打印。在这次审查中,将概述微流体平台和细胞外基质样生物材料的优点和缺点。然后,通过分析用于神经退行性研究的3D片上大脑的最新进展,将微流体和水凝胶的组合作为研究神经疾病的新的协同方法进行探索。
    The alteration in the neural circuits of both central and peripheral nervous systems is closely related to the onset of neurodegenerative disorders (NDDs). Despite significant research efforts, the knowledge regarding NDD pathological processes, and the development of efficacious drugs are still limited due to the inability to access and reproduce the components of the nervous system and its intricate microenvironment. 2D culture systems are too simplistic to accurately represent the more complex and dynamic situation of cells in vivo and have therefore been surpassed by 3D systems. However, both models suffer from various limitations that can be overcome by employing two innovative technologies: organ-on-chip and 3D printing. In this review, an overview of the advantages and shortcomings of both microfluidic platforms and extracellular matrix-like biomaterials will be given. Then, the combination of microfluidics and hydrogels as a new synergistic approach to study neural disorders by analyzing the latest advances in 3D brain-on-chip for neurodegenerative research will be explored.
    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
    2型糖尿病(T2DM),肥胖,和代谢功能障碍相关的脂肪变性肝病(MASLD)是流行病学相关的疾病,在全球范围内患病率不断上升。虽然导致这些疾病发作和发展的机制尚未完全了解,用于研究调节能量代谢的中央器官之间的协调相互作用的预测性组织表示,尤其是肝脏和胰岛,是需要的。这里,提出了一种双无泵再循环芯片上器官(dual-rOoC)平台,该平台结合了人类多能干细胞(sc)来源的sc-肝脏和sc-胰岛类器官。该平台再现了两个器官之间代谢串扰的关键方面,包括葡萄糖水平和选定的激素,并支持sc-胰岛和sc-肝脏类器官的活力和功能,同时保留促炎细胞因子的减少释放。在对高脂和果糖治疗的代谢破坏模型中,sc-肝类器官表现出脂肪变性和胰岛素抵抗的标志,而sc-胰岛在芯片上产生促炎细胞因子。最后,该平台在芯片上再现抗糖尿病药物的已知作用。一起来看,该平台为肥胖的功能研究提供了基础,T2DM,和芯片上的MASLD,以及测试潜在的治疗干预措施。本文受版权保护。保留所有权利。
    Type 2 diabetes mellitus (T2DM), obesity, and metabolic dysfunction-associated steatotic liver disease (MASLD) are epidemiologically correlated disorders with a worldwide growing prevalence. While the mechanisms leading to the onset and development of these conditions are not fully understood, predictive tissue representations for studying the coordinated interactions between central organs that regulate energy metabolism, particularly the liver and pancreatic islets, are needed. Here, a dual pump-less recirculating organ-on-chip platform that combines human pluripotent stem cell (sc)-derived sc-liver and sc-islet organoids is presented. The platform reproduces key aspects of the metabolic cross-talk between both organs, including glucose levels and selected hormones, and supports the viability and functionality of both sc-islet and sc-liver organoids while preserving a reduced release of pro-inflammatory cytokines. In a model of metabolic disruption in response to treatment with high lipids and fructose, sc-liver organoids exhibit hallmarks of steatosis and insulin resistance, while sc-islets produce pro-inflammatory cytokines on-chip. Finally, the platform reproduces known effects of anti-diabetic drugs on-chip. Taken together, the platform provides a basis for functional studies of obesity, T2DM, and MASLD on-chip, as well as for testing potential therapeutic interventions.
    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
    背景器官芯片技术加速了血管系统的体外临床前研究,该平台的一个关键优势在于,它有望通过提供原代人类细胞培养环境来影响个性化医疗,在该环境中,直接从单个组织中对内皮细胞进行活检或通过干细胞生物技术进行分化。然而,这些方法很难在实验室采用,并经常导致细胞的杂质和异质性。这限制了器官芯片做出准确生理预测的能力。在这项研究中,我们报道了使用血液来源的内皮细胞替代原代和诱导多能干细胞来源的内皮细胞.方法和结果,基因型,表型,和器官芯片功能特征的血液来源的生长内皮细胞与市售和最常用的原代内皮细胞和诱导多能干细胞来源的内皮细胞进行了比较。这些方法包括RNA测序,以及细胞标记表达的标准标准测定,生长动力学,迁移潜力,和血管生成。最后,使用经血液来源的内皮细胞工程化的血管芯片评估剪切下的血栓炎症反应.血液来源的内皮细胞表现出典型内皮细胞的标准标准标志。不同来源的内皮细胞基因表达谱存在差异,但是血液来源的细胞比诱导多能干细胞来源的细胞更接近原代细胞。此外,血液来源的内皮细胞更容易从个体中获得,与原代细胞或诱导多能干细胞来源的细胞相比,它们在功能研究和器官芯片方面同样有效。结论血液来源的内皮细胞可用于临床前研究,以使用芯片上器官开发更强大和个性化的下一代疾病模型。
    Background Organ-on-chip technology has accelerated in vitro preclinical research of the vascular system, and a key strength of this platform is its promise to impact personalized medicine by providing a primary human cell-culture environment where endothelial cells are directly biopsied from individual tissue or differentiated through stem cell biotechniques. However, these methods are difficult to adopt in laboratories, and often result in impurity and heterogeneity of cells. This limits the power of organ-chips in making accurate physiological predictions. In this study, we report the use of blood-derived endothelial cells as alternatives to primary and induced pluripotent stem cell-derived endothelial cells. Methods and Results Here, the genotype, phenotype, and organ-chip functional characteristics of blood-derived outgrowth endothelial cells were compared against commercially available and most used primary endothelial cells and induced pluripotent stem cell-derived endothelial cells. The methods include RNA-sequencing, as well as criterion standard assays of cell marker expression, growth kinetics, migration potential, and vasculogenesis. Finally, thromboinflammatory responses under shear using vessel-chips engineered with blood-derived endothelial cells were assessed. Blood-derived endothelial cells exhibit the criterion standard hallmarks of typical endothelial cells. There are differences in gene expression profiles between different sources of endothelial cells, but blood-derived cells are relatively closer to primary cells than induced pluripotent stem cell-derived. Furthermore, blood-derived endothelial cells are much easier to obtain from individuals and yet, they serve as an equally effective cell source for functional studies and organ-chips compared with primary cells or induced pluripotent stem cell-derived cells. Conclusions Blood-derived endothelial cells may be used in preclinical research for developing more robust and personalized next-generation disease models using organ-on-chips.
    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