gut-on-chip

芯片上消化道
  • 文章类型: Journal Article
    人类诱导多能干细胞(hiPSC)来源的肠道类器官是研究发育生物学和个性化治疗的有价值的工具,但是它们的封闭拓扑和相对不成熟的状态限制了应用。这里,我们使用芯片上器官技术在更生理的体外微环境中开发出具有顶端和基底外侧通路的hiPSC衍生肠屏障。为了沿着隐窝-绒毛轴复制生长因子梯度,我们将细胞局部暴露于扩增和分化培养基中。在这些条件下,肠上皮细胞自组织成具有生理屏障完整性的绒毛样褶皱,肌成纤维细胞和神经元在底部通道中出现并形成上皮下组织。生长因子梯度有效地平衡分裂和成熟细胞类型,并诱导肠上皮成分,包括吸收和分泌谱系,类似于人类小肠的成分。这种特征良好的hiPSC衍生的芯片肠系统可以促进对人类小肠中的生理过程和治疗开发的个性化研究。
    Human induced pluripotent stem cell (hiPSC)-derived intestinal organoids are valuable tools for researching developmental biology and personalized therapies, but their closed topology and relative immature state limit applications. Here, we use organ-on-chip technology to develop a hiPSC-derived intestinal barrier with apical and basolateral access in a more physiological in vitro microenvironment. To replicate growth factor gradients along the crypt-villus axis, we locally expose the cells to expansion and differentiation media. In these conditions, intestinal epithelial cells self-organize into villus-like folds with physiological barrier integrity, and myofibroblasts and neurons emerge and form a subepithelial tissue in the bottom channel. The growth factor gradients efficiently balance dividing and mature cell types and induce an intestinal epithelial composition, including absorptive and secretory lineages, resembling the composition of the human small intestine. This well-characterized hiPSC-derived intestine-on-chip system can facilitate personalized studies on physiological processes and therapy development in the human small intestine.
    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
    微生理系统(MPS)是有前途的体外技术,用于生理相关的人体吸收预测,分布,新陈代谢,和候选药物的排泄(ADME)特性。然而,聚二甲基硅氧烷(PDMS),MPS中使用的常用材料,可以吸附和吸收小分子,从而损害实验结果。本研究旨在评估使用基于PDMS的Emulate芯片肠来确定首过肠道药物清除的可行性。在无细胞的PDMS器官芯片中,我们评估了17种药物的损失,其中睾酮基于其对器官芯片的大量吸附和吸收以及其广泛的首过肠代谢以及特征明确的代谢物而被选择作为进一步研究的模型化合物。建立了由上皮Caco-2细胞和原代人脐静脉内皮细胞(HUVECs)组成的肠芯片模型。用参考化合物和药物流出的抑制测试模型的屏障完整性。在无细胞器官芯片和芯片上肠模型中测量睾酮的浓度-时间曲线。提供了推断代谢清除的方法。我们的结果表明,尽管芯片上有大量化合物损失,但可以使用基于PDMS的MPS确定代谢清除。总的来说,这项研究提供了一个实用的协议,通过实验评估基于PDMS的MPS中的ADME特性。
    Microphysiological systems (MPSs) are promising in vitro technologies for physiologically relevant predictions of the human absorption, distribution, metabolism, and excretion (ADME) properties of drug candidates. However, polydimethylsiloxane (PDMS), a common material used in MPSs, can both adsorb and absorb small molecules, thereby compromising experimental results. This study aimed to evaluate the feasibility of using the PDMS-based Emulate gut-on-chip to determine the first-pass intestinal drug clearance. In cell-free PDMS organ-chips, we assessed the loss of 17 drugs, among which testosterone was selected as a model compound for further study based on its substantial ad- and absorptions to organ chips and its extensive first-pass intestinal metabolism with well-characterized metabolites. A gut-on-chip model consisting of epithelial Caco-2 cells and primary human umbilical vein endothelial cells (HUVECs) was established. The barrier integrity of the model was tested with reference compounds and inhibition of drug efflux. Concentration-time profiles of testosterone were measured in cell-free organ chips and in gut-on-chip models. A method to deduce the metabolic clearance was provided. Our results demonstrate that metabolic clearance can be determined with PDMS-based MPSs despite substantial compound loss to the chip. Overall, this study offers a practical protocol to experimentally assess ADME properties in PDMS-based MPSs.
    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
    慢性消化系统疾病在世界范围内的发病率越来越高,治疗费用昂贵,尚无治愈方法。现有的治疗方案主要依靠症状缓解,患者对这种治疗的反应具有很大程度的变异性,强调需要新的治疗策略。有强烈的迹象表明,肠道微生物群的贡献似乎是疾病活动和患者治疗反应的关键调节剂。因此,人们一直致力于理解宿主-微生物相互作用以及支撑这种变异性的机制。动物模型,作为黄金标准,提供对宿主-微生物相互作用的有价值的机械见解。然而,它们不能免除促使替代方法发展的限制。新兴的微流控技术和芯片上肠道模型被证明反映了肠道生理和疾病状态的主要特征,反映微生物群的改变,并包括用于研究宿主反应的功能读数。在这篇评论中,我们讨论了动物模型在理解宿主-微生物相互作用方面的相关性,以及芯片上肠道技术如何有望解决患者对慢性消化系统疾病治疗反应的变异性.
    Chronic digestive disorders are of increasing incidence worldwide with expensive treatments and no available cure. Available therapeutic schemes mainly rely on symptom relief, with large degrees of variability in patients\' response to such treatments, underlining the need for new therapeutic strategies. There are strong indications that the gut microbiota\'s contribution seems to be a key modulator of disease activity and patients\' treatment responses. Hence, efforts have been devoted to understanding host-microbe interactions and the mechanisms underpinning such variability. Animal models, being the gold standard, provide valuable mechanistic insights into host-microbe interactions. However, they are not exempt from limitations prompting the development of alternative methods. Emerging microfluidic technologies and gut-on-chip models were shown to mirror the main features of gut physiology and disease state, reflect microbiota modification, and include functional readouts for studying host responses. In this commentary, we discuss the relevance of animal models in understanding host-microbe interactions and how gut-on-chip technology holds promises for addressing patient variability in responses to chronic digestive disease treatment.
    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
    探索硫化氢(H2S)的胃肠道作用是困难的,因为它的挥发性和缺乏用于操纵肠道环境的精确可控的模型系统。Hayesetal.通过工程大肠杆菌在不透气的片上肠设备中滴定H2S水平来解决这个问题。
    Exploring the gastrointestinal role of hydrogen sulfide (H2S) is difficult because of its volatility and the absence of a precisely controllable model system for manipulating the gut environment. Hayes et al. address this issue by engineering Escherichia coli to titrate H2S levels in a gas-impermeable gut-on-chip device.
    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
    炎症性肠病对患者和医疗保健系统造成重大负担,提高了开发有效疗法的必要性。细胞培养技术的进步,与道德问题结盟,推动体外模型作为研究疾病病因的重要工具,其进展,和可能的疗法。已经使用了几种基于细胞的肠道炎症体外模型,它们的复杂性和诱发炎症的方法各不相同。永生化细胞系由于其长期存活而被广泛使用。与短暂但患者特异性的原代培养相反。最近,类器官和器官芯片通过在生理上更相关而显示出巨大的潜力。这篇综述旨在阐明肠道炎症的复杂性,并涵盖最近报道人类肠道炎症的基于细胞的体外模型的工作。包括不同的方法和结果。
    Inflammatory bowel disease causes a major burden to patients and healthcare systems, raising the need to develop effective therapies. Technological advances in cell culture, allied with ethical issues, have propelled in vitro models as essential tools to study disease aetiology, its progression, and possible therapies. Several cell-based in vitro models of intestinal inflammation have been used, varying in their complexity and methodology to induce inflammation. Immortalized cell lines are extensively used due to their long-term survival, in contrast to primary cultures that are short-lived but patient-specific. Recently, organoids and organ-chips have demonstrated great potential by being physiologically more relevant. This review aims to shed light on the intricate nature of intestinal inflammation and cover recent works that report cell-based in vitro models of human intestinal inflammation, encompassing diverse approaches and outcomes.
    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
    炎症性肠病(IBD)是一种慢性,复发性胃肠道疾病。溃疡性结肠炎和克罗恩病是炎症性肠病的类型。几十年来,这种疾病一直是研究的主题,专家们仍在试图找出其原因和病理。研究人员已经建立了许多体内动物模型,体外细胞系,和离体系统,以最终了解其原因并充分确定治疗方法。然而,体内动物模型不能被认为是研究IBD的良好模型,因为它们不能完全模拟疾病。此外,因为物种差异是一个至关重要的问题,可以采用体外细胞系和离体系统来适当地重建该条件。体外模型是生物学和医学研究的起点。已经开发了用于复制肠道生理学的离体和体外模型。这篇综述旨在清楚地了解IBD的几种体外和离体模型,并提供对它们的益处和局限性及其在理解肠道生理学方面的价值的见解。
    Inflammatory bowel disease (IBD) is a chronic, relapsing gastrointestinal condition. Ulcerative colitis and Crohn\'s disease are types of inflammatory bowel disease. Over many decades, the disease has been a topic of study, with experts still trying to figure out its cause and pathology. Researchers have established many in vivo animal models, in vitro cell lines, and ex vivo systems to understand its cause ultimately and adequately identify a therapy. However, in vivo animal models cannot be regarded as good models for studying IBD since they cannot completely simulate the disease. Furthermore, because species differences are a crucial subject of concern, in vitro cell lines and ex vivo systems can be employed to recreate the condition properly. In vitro models serve as the starting point for biological and medical research. Ex vivo and in vitro models for replicating gut physiology have been developed. This review aims to present a clear understanding of several in vitro and ex vivo models of IBD and provide insights into their benefits and limits and their value in understanding intestinal physiology.
    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
    肠粘液是支持肠道微生物群生长和过滤分子的生物屏障。为了执行这些功能,粘液具有优化的微观结构和粘弹性,并且可以稳定地补充,从而沿着肠道流动。可用的体外肠粘液模型是研究微生物群-人细胞相互作用的有用工具,并用作细菌培养的基质或用作微流体设备的静态组件,例如芯片上的肠道。这项工作的目的是设计一种体外粘液模型(I-Bac3Gel),以解决单个系统的生理粘弹性特性(即,2-200Pa),动态细菌培养的3D结构和适用性。优化均匀交联的藻酸盐水凝胶的组成,以获得目标粘弹性和微观结构性能。然后,利用流变测试来先验地评估水凝胶承受流动动态条件的能力。我们通过将动态流应用于负载细菌的粘液模型并通过监测大肠杆菌的生长和存活来实验评估I-Bac3Gels在微流体发展领域中的适用性。工程模型代表了粘液建模的进步,因为他们可以满足不同的紧急需求,比如3D结构,生物启发特性和与动态系统的兼容性。
    The intestinal mucus is a biological barrier that supports the intestinal microbiota growth and filters molecules. To perform these functions, mucus possesses optimized microstructure and viscoelastic properties and it is steadily replenished thus flowing along the gut. The available in vitro intestinal mucus models are useful tools in investigating the microbiota-human cells interaction, and are used as matrices for bacterial culture or as static component of microfluidic devices like gut-on-chips. The aim of this work is to engineer an in vitro mucus models (I-Bac3Gel) addressing in a single system physiological viscoelastic properties (i.e., 2-200 Pa), 3D structure and suitability for dynamic bacterial culture. Homogeneously crosslinked alginate hydrogels are optimized in composition to obtain target viscoelastic and microstructural properties. Then, rheological tests are exploited to assess a priori the hydrogels capability to withstand the flow dynamic condition. We experimentally assess the suitability of I-Bac3Gels in the evolving field of microfluidics by applying a dynamic flow to a bacterial-loaded mucus model and by monitoring E. coli growth and survival. The engineered models represent a step forward in the modelling of the mucus, since they can answer to different urgent needs such as a 3D structure, bioinspired properties and compatibility with dynamic system.
    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
    结直肠癌(CRC)是影响人类的最普遍的癌症之一,具有复杂的遗传和环境病因。与已知环境的癌症不同,可遗传,或者与性别相关的原因,散发性CRC很难预测,并且在临床应用中没有具有风险的分子生物标志物.二十个CRC病例中有一个具有既定的可遗传成分。其余病例是散发性的,与部分模糊的遗传有关,表观遗传,再生,微生物,饮食,和生活方式因素。为了解决这种复杂性,我们应该改进结肠镜检查的做法,建议统一超过一定年龄,包括对指示个体CRC风险的生物标志物的评估。理想情况下,这类生物标志物将导致该疾病,并且在饮食或治疗干预下可能会改变.多组学分析,包括转录,表观遗传以及宏基因组,和代谢组学概况,迫切需要为风险分析提供数据。本文的目的是提供一个视角,对导致CRC启动的体内平衡的多因素脱轨,这可以通过多组学和芯片肠道分析来探索,以确定急需的预测性生物标志物。
    Colorectal cancer (CRC) is one of the most prevalent cancers affecting humans, with a complex genetic and environmental aetiology. Unlike cancers with known environmental, heritable, or sex-linked causes, sporadic CRC is hard to foresee and has no molecular biomarkers of risk in clinical use. One in twenty CRC cases presents with an established heritable component. The remaining cases are sporadic and associated with partially obscure genetic, epigenetic, regenerative, microbiological, dietary, and lifestyle factors. To tackle this complexity, we should improve the practice of colonoscopy, which is recommended uniformly beyond a certain age, to include an assessment of biomarkers indicative of individual CRC risk. Ideally, such biomarkers will be causal to the disease and potentially modifiable upon dietary or therapeutic interventions. Multi-omics analysis, including transcriptional, epigenetic as well as metagenomic, and metabolomic profiles, are urgently required to provide data for risk analyses. The aim of this article is to provide a perspective on the multifactorial derailment of homeostasis leading to the initiation of CRC, which may be explored via multi-omics and Gut-on-Chip analysis to identify much-needed predictive biomarkers.
    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
    人们对了解肠道微生物组是否以及如何与疾病的发病机理和/或进展有因果关系越来越感兴趣。虽然体外细胞系模型通常用于研究宿主-微生物相互作用的特定方面,侏儒小鼠模型被认为是研究微生物组研究因果关系的首选平台。然而,动物研究的发现为描述人类肠道微生物组研究的各个感兴趣领域提供了有限的机会。芯片上的肠道是模拟肠道生理学的仿生物质,可以研究宿主和微生物组的双向作用。我们认为,它们可以在三个主要领域推进因果和生态肠道微生物组研究:(i)饮食-微生物组和药物-微生物组相互作用;(ii)微生物组靶向治疗药物生态学;(iii)肠道微生物组和微生物组靶向干预肠外病变的机理研究。
    There is a growing interest to understand if and how the gut microbiome is causally linked to the pathogenesis and/or progression of diseases. While in vitro cell line models are commonly used for studying specific aspects of the host-microbe interaction, gnotobiotic murine models are considered the preferred platform for studying causality in microbiome research. Nevertheless, findings from animal studies provide limited opportunity for delineating various areas of interest to the human gut microbiome research. Gut-on-chips are biomimetics recapitulating intestinal physiology which enable investigation of bidirectional effects of the host and microbiome. We posit that they could advance causal and ecological gut microbiome research in three major areas: (i) diet-microbiome and drug-microbiome interaction; (ii) microbiome-targeted therapeutics pharmacoecology; and (iii) mechanistic studies of gut microbiome and microbiome-targeted intervention in extraintestinal pathologies.
    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
    芯片器官(OoC)技术充满了工程和生物挑战,但它有可能彻底改变消费产品和化学品新成分的下一代风险评估。将OoC技术成功纳入下一代风险评估工具箱取决于所使用的微流体设备和器官组织模型的鲁棒性。标准化设备制造的最新进展,器官组织培养和生长方案提供了弥合实施器官芯片技术的差距的能力。下一代风险评估是一种暴露主导和假设驱动的分层方法,使用详细的人体暴露信息和适当的新(非动物)毒理学测试方法的应用进行风险评估。通过将人细胞培养与动态微流体相结合以改善生理模拟,芯片上器官提出了一种有前途的体外方法。这里,我们严格审查商业器官芯片设备,以及最近的皮肤组织培养模型研究,肠屏障和肝脏作为主要代谢器官,用于下一代风险评估。最后,对芯片上器官设备中的微流体连接的组织组合,例如皮肤-肝脏和肠-肝脏进行了审查,因为它们构成了推进毒物代谢动力学和毒物动力学研究的相关方面。我们指出最近的成就和需要克服的挑战,推进非动物,与人类相关的安全性研究。
    Organ-on-chip (OoC) technology is full of engineering and biological challenges, but it has the potential to revolutionize the Next-Generation Risk Assessment of novel ingredients for consumer products and chemicals. A successful incorporation of OoC technology into the Next-Generation Risk Assessment toolbox depends on the robustness of the microfluidic devices and the organ tissue models used. Recent advances in standardized device manufacturing, organ tissue cultivation and growth protocols offer the ability to bridge the gaps towards the implementation of organ-on-chip technology. Next-Generation Risk Assessment is an exposure-led and hypothesis-driven tiered approach to risk assessment using detailed human exposure information and the application of appropriate new (non-animal) toxicological testing approaches. Organ-on-chip presents a promising in vitro approach by combining human cell culturing with dynamic microfluidics to improve physiological emulation. Here, we critically review commercial organ-on-chip devices, as well as recent tissue culture model studies of the skin, intestinal barrier and liver as the main metabolic organ to be used on-chip for Next-Generation Risk Assessment. Finally, microfluidically linked tissue combinations such as skin-liver and intestine-liver in organ-on-chip devices are reviewed as they form a relevant aspect for advancing toxicokinetic and toxicodynamic studies. We point to recent achievements and challenges to overcome, to advance non-animal, human-relevant safety studies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号