1
|
Hsieh HC, Han Q, Brenes D, Bishop KW, Wang R, Wang Y, Poudel C, Glaser AK, Freedman BS, Vaughan JC, Allbritton NL, Liu JTC. Imaging 3D cell cultures with optical microscopy. Nat Methods 2025; 22:1167-1190. [PMID: 40247123 DOI: 10.1038/s41592-025-02647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2025] [Indexed: 04/19/2025]
Abstract
Three-dimensional (3D) cell cultures have gained popularity in recent years due to their ability to represent complex tissues or organs more faithfully than conventional two-dimensional (2D) cell culture. This article reviews the application of both 2D and 3D microscopy approaches for monitoring and studying 3D cell cultures. We first summarize the most popular optical microscopy methods that have been used with 3D cell cultures. We then discuss the general advantages and disadvantages of various microscopy techniques for several broad categories of investigation involving 3D cell cultures. Finally, we provide perspectives on key areas of technical need in which there are clear opportunities for innovation. Our goal is to guide microscope engineers and biomedical end users toward optimal imaging methods for specific investigational scenarios and to identify use cases in which additional innovations in high-resolution imaging could be helpful.
Collapse
Affiliation(s)
- Huai-Ching Hsieh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - David Brenes
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Kevin W Bishop
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Rui Wang
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Adam K Glaser
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Benjamin S Freedman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Nephrology, Kidney Research Institute and Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
- Plurexa LLC, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan T C Liu
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
2
|
Chi KY, Kim G, Son JS, Han J, Kim JH. Recent Advances in Three-Dimensional In Vitro Models for Studies of Liver Fibrosis. Tissue Eng Regen Med 2025:10.1007/s13770-025-00719-8. [PMID: 40358834 DOI: 10.1007/s13770-025-00719-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/02/2025] [Accepted: 03/11/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Liver fibrosis is a reversible but complex pathological condition associated with chronic liver diseases, affecting over 1.5 billion people worldwide. It is characterized by excessive extracellular matrix deposition resulting from sustained liver injury, often advancing to cirrhosis and cancer. As its progression involves various cell types and pathogenic factors, understanding the intricate mechanisms is essential for the development of effective therapies. In this context, extensive efforts have been made to establish three-dimensional (3D) in vitro platforms that mimic the progression of liver fibrosis. METHODS This review outlines the pathophysiology of liver fibrosis and highlights recent advancements in 3D in vitro liver models, including spheroids, organoids, assembloids, bioprinted constructs, and microfluidic systems. It further assesses their biological relevance, with particular focus on their capacity to reproduce fibrosis-related characteristics. RESULTS 3D in vitro liver models offer significant advantages over conventional two-dimensional cultures. Although each model exhibits unique strengths, they collectively recapitulate key fibrotic features, such as extracellular matrix remodeling, hepatic stellate cell activation, and collagen deposition, in a physiologically relevant 3D setting. In particular, multilineage liver organoids and assembloids integrate architectural complexity with scalability, enabling deeper mechanistic insights and supporting therapeutic evaluation with improved translational relevance. CONCLUSION 3D in vitro liver models represent a promising strategy to bridge the gap between in vitro studies and in vivo realities by faithfully replicating liver-specific architecture and microenvironments. With enhanced reproducibility through standardized protocols, these models hold great potential for advancing drug discovery and facilitating the development of personalized therapies for liver fibrosis.
Collapse
Affiliation(s)
- Kyun Yoo Chi
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Gyeongmin Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea
| | - Jeong Sang Son
- User Convenience Technology R&D Department, Korea Institute of Industrial Technology (KITECH), Ansan, 15588, South Korea
| | - Jiyou Han
- Department of Biomedical and Chemical Sciences, Hyupsung University, Hwasung-Si, 18330, South Korea
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
3
|
Zhou Y, Zhong Y, Lauschke VM. Evaluating the synergistic use of advanced liver models and AI for the prediction of drug-induced liver injury. Expert Opin Drug Metab Toxicol 2025; 21:563-577. [PMID: 39893552 DOI: 10.1080/17425255.2025.2461484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Drug-induced liver injury (DILI) is a leading cause of acute liver failure. Hepatotoxicity typically occurs only in a subset of individuals after prolonged exposure and constitutes a major risk factor for the termination of drug development projects. AREAS COVERED We provide an overview of available human liver models for DILI research and discuss how they have been used to aid in early risk assessments and to mitigate the risk of project closures due to DILI in clinical stages. We summarize the different data that can be provided by such models and illustrate how these diverse data types can be interfaced with machine learning strategies to improve predictions of liver safety liabilities. EXPERT OPINION Advanced human liver models closely mimic human liver phenotypes and functions for many weeks, allowing for the recapitulation of hepatotoxicity events in vitro. Integration of the biochemical, histological, and toxicogenomic output data from these models with physicochemical compound properties using different machine learning architectures holds promise to enhance preclinical DILI predictions. However, to realize this aim, it is important to benchmark the available liver models on test sets of DILI positive and negative compounds and to carefully annotate and share the resulting data.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Yi Zhong
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
4
|
Abdurakhmanova МM, Leonteva AA, Vasilieva NS, Kuligina EV, Nushtaeva AA. 3D cell culture models: how to obtain and characterize the main models. Vavilovskii Zhurnal Genet Selektsii 2025; 29:175-188. [PMID: 40264808 PMCID: PMC12011624 DOI: 10.18699/vjgb-25-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 04/24/2025] Open
Abstract
For many years, the gold standard in the study of malignant tumors has been the in vitro culture of tumor cells, in vivo xenografts or genetically modified animal models. Meanwhile, three-dimensional cell models (3D cultures) have been added to the arsenal of modern biomedical research. 3D cultures reproduce tissue-specific features of tissue topology. This makes them relevant tissue models in terms of cell differentiation, metabolism and the development of drug resistance. Such models are already being used by many research groups for both basic and translational research, and may substantially reduce the number of animal studies, for example in the field of oncological research. In the current literature, 3D cultures are classified according to the technique of their formation (with or without a scaffold), cultivation conditions (static or dynamic), as well as their cellular organization and function. In terms of cellular organization, 3D cultures are divided into "spheroid models", "organoids", "organs-on-a-chip" and "microtissues". Each of these models has its own unique features, which should be taken into account when using a particular model in an experiment. The simplest 3D cultures are spheroid models which are floating spherical cell aggregates. An organoid is a more complex 3D model, in which a self-organizing 3D structure is formed from stem cells (SCs) capable of self-renewal and differentiation within the model. Organ-on-a-chip models are chips of microfluidic systems that simulate dynamic physical and biological processes found in organs and tissues in vitro. By combining different cell types into a single structure, spheroids and organoids can act as a basis for the formation of a microtissue - a hybrid 3D model imitating a specific tissue phenotype and containing tissue-specific extracellular matrix (ECM) components. This review presents a brief history of 3D cell culture. It describes the main characteristics and perspectives of the use of "spheroid models", "organoids", "organ-on-a-chip" models and "microtissues" in immune oncology research of solid tumors.
Collapse
Affiliation(s)
- М M Abdurakhmanova
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Leonteva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N S Vasilieva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E V Kuligina
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Nushtaeva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
5
|
Liao Y, Chai D, Wang Q, Wang X, Yong Q, Cheng Z, Zhang C, Zhang D, Liu B, Liu R, Li Z. Sensor-combined organ-on-a-chip for pharmaceutical and medical sciences: from design and materials to typical biomedical applications. MATERIALS HORIZONS 2025; 12:2161-2178. [PMID: 39801302 DOI: 10.1039/d4mh01174k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Organ-on-a-chip (OoC) is a breakthrough technology in biomedicine. As microphysiological systems constructed in vitro, OoCs can simulate the main structures and functions of human organs, thereby providing a powerful tool for drug screening and disease model construction. Furthermore, the coupling of OoCs and sensors has been an innovative discovery in the field of biomedical and electronic engineering in recent years. The integration of sensors into OoCs allows the real-time monitoring of the changes in the microenvironmental parameters within the chip, reflecting the physiological responses of cells or tissues in the OoC and providing more accurate data support for drug development and disease treatment. In this work, we briefly outline the design ideas of OoCs, summarize the commonly used materials for OoCs and their advantages and disadvantages, and provide the most recent practical examples of the combination of OoCs and sensors in pharmaceutical and medical sciences. Furthermore, perspectives, challenges and their solutions in the future development of this technology are provided, with the aim to inspire the researchers to work toward the subsequent development of OoCs having improved reliability.
Collapse
Affiliation(s)
- Yumeng Liao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Danni Chai
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Quan Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xueqi Wang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qian Yong
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zhaoming Cheng
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Chuanjun Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Di Zhang
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Boshi Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Rui Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Zheng Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin 301617, PR China
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin 301617, PR China
| |
Collapse
|
6
|
Brandauer K, Schweinitzer S, Lorenz A, Krauß J, Schobesberger S, Frauenlob M, Ertl P. Advances of dual-organ and multi-organ systems for gut, lung, skin and liver models in absorption and metabolism studies. LAB ON A CHIP 2025; 25:1384-1403. [PMID: 39973270 DOI: 10.1039/d4lc01011f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Drug development is a costly and timely process with high risks of failure during clinical trials. Although in vitro tissue models have significantly advanced over the years, thus fostering a transition from animal-derived models towards human-derived models, failure rates still remain high. Current cell-based assays are still not able to provide an accurate prediction of the clinical success or failure of a drug candidate. To overcome the limitations of current methods, a variety of microfluidic systems have been developed as powerful tools that are capable of mimicking (micro)physiological conditions more closely by integrating physiological fluid flow conditions, mechanobiological cues and concentration gradients, to name only a few. One major advantage of these biochip-based tissue cultures, however, is their ability to seamlessly connect different organ models, thereby allowing the study of organ-crosstalk and metabolic byproduct effects. This is especially important when assessing absorption, distribution, metabolism, and excretion (ADME) processes of drug candidates, where an interplay between various organs is a prerequisite. In the current review, a number of in vitro models as well as microfluidic dual- and multi-organ systems are summarized with a focus on absorption (skin, lung, gut) and metabolism (liver). Additionally, the advantage of multi-organ chips in identifying a drug's on and off-target toxicity is discussed. Finally, the potential high-throughput implementation and modular chip design of multi-organ-on-a-chip systems within the pharmaceutical industry is highlighted, outlining the necessity of reducing handling complexity.
Collapse
Affiliation(s)
- Konstanze Brandauer
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Sophie Schweinitzer
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Alexandra Lorenz
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Judith Krauß
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | | | - Martin Frauenlob
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Peter Ertl
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| |
Collapse
|
7
|
Xue C, Yin Y, Xu X, Tian K, Su J, Hu G. Particle manipulation under X-force fields. LAB ON A CHIP 2025; 25:956-978. [PMID: 39774586 DOI: 10.1039/d4lc00794h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Particle manipulation is a central technique that enhances numerous scientific and medical applications by exploiting micro- and nanoscale control within fluidic environments. In this review, we systematically explore the multifaceted domain of particle manipulation under the influence of various X-force fields, integral to lab-on-a-chip technologies. We dissect the fundamental mechanisms of hydrodynamic, gravitational, optical, magnetic, electrical, and acoustic forces and detail their individual and synergistic applications. In particular, our discourse extends to advanced multi-modal manipulation strategies that harness the combined power of these forces, revealing their enhanced efficiency and precision in complex assays and diagnostic frameworks. The integration of cutting-edge technologies such as artificial intelligence and autonomous systems further enhances the capabilities of these microfluidic platforms, leading to transformative innovations in personalized medicine and point-of-care diagnostics. This review not only highlights current technological advances, but also forecasts the trajectory of future developments, emphasizing the escalating precision and scalability essential for advancing lab-on-a-chip applications.
Collapse
Affiliation(s)
- Chundong Xue
- Institute of Cardio-cerebrovascular Medicine, Central Hospital of Dalian University of Technology, Dalian 116033, China
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Yifan Yin
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Xiaoyu Xu
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian 116024, China
| | - Kai Tian
- School of Mechanical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jinghong Su
- Department of Energy and Power Engineering, Tsinghua University, Beijing 100084, China
| | - Guoqing Hu
- Department of Engineering Mechanics, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
8
|
Janssen R, Benito-Zarza L, Cleijpool P, Valverde MG, Mihăilă SM, Bastiaan-Net S, Garssen J, Willemsen LEM, Masereeuw R. Biofabrication Directions in Recapitulating the Immune System-on-a-Chip. Adv Healthc Mater 2025; 14:e2304569. [PMID: 38625078 DOI: 10.1002/adhm.202304569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/19/2024] [Indexed: 04/17/2024]
Abstract
Ever since the implementation of microfluidics in the biomedical field, in vitro models have experienced unprecedented progress that has led to a new generation of highly complex miniaturized cell culture platforms, known as Organs-on-a-Chip (OoC). These devices aim to emulate biologically relevant environments, encompassing perfusion and other mechanical and/or biochemical stimuli, to recapitulate key physiological events. While OoCs excel in simulating diverse organ functions, the integration of the immune organs and immune cells, though recent and challenging, is pivotal for a more comprehensive representation of human physiology. This comprehensive review covers the state of the art in the intricate landscape of immune OoC models, shedding light on the pivotal role of biofabrication technologies in bridging the gap between conceptual design and physiological relevance. The multifaceted aspects of immune cell behavior, crosstalk, and immune responses that are aimed to be replicated within microfluidic environments, emphasizing the need for precise biomimicry are explored. Furthermore, the latest breakthroughs and challenges of biofabrication technologies in immune OoC platforms are described, guiding researchers toward a deeper understanding of immune physiology and the development of more accurate and human predictive models for a.o., immune-related disorders, immune development, immune programming, and immune regulation.
Collapse
Affiliation(s)
- Robine Janssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Laura Benito-Zarza
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Pim Cleijpool
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Marta G Valverde
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Silvia M Mihăilă
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, 6708 WG, The Netherlands
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
- Danone Global Research & Innovation Center, Danone Nutricia Research B.V., Utrecht, 3584 CT, The Netherlands
| | - Linette E M Willemsen
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Rosalinde Masereeuw
- Department of Pharmaceutical Sciences, Pharmacology, Utrecht University, Utrecht, 3584 CG, The Netherlands
| |
Collapse
|
9
|
Fan X, Hou K, Liu G, Shi R, Wang W, Liang G. Strategies to overcome the limitations of current organoid technology - engineered organoids. J Tissue Eng 2025; 16:20417314251319475. [PMID: 40290859 PMCID: PMC12033597 DOI: 10.1177/20417314251319475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/26/2025] [Indexed: 04/30/2025] Open
Abstract
Organoids, as 3D in vitro models derived from stem cells, have unparalleled advantages over traditional cell and animal models for studying organogenesis, disease mechanisms, drug screening, and personalized diagnosis and treatment. Despite the tremendous progress made in organoid technology, the translational application of organoids still presents enormous challenges due to the complex structure and function of human organs. In this review, the limitations of the translational application of traditional organoid technologies are first described. Next, we explore ways to address many of the limitations of traditional organoid cultures by engineering various dimensions of organoid systems. Finally, we discuss future directions in the field, including potential roles in drug screening, simulated microphysiology system and personalized diagnosis and treatment. We hope that this review inspires future research into organoids and microphysiology system.
Collapse
Affiliation(s)
- Xulong Fan
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
| | - Kun Hou
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, China
| | - Gaojian Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
| | - Ruolin Shi
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, China
| | - Wenjie Wang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, China
| |
Collapse
|
10
|
Maulana TI, Wevers NR, Kristoforus T, Chandler M, Lanz HL, Joore J, Vulto P, Villenave R, Kustermann S, Loskill P, Bircsak KM. Opportunities for Microphysiological Systems in Toxicity Testing of New Drug Modalities. Annu Rev Pharmacol Toxicol 2025; 65:47-69. [PMID: 39227343 DOI: 10.1146/annurev-pharmtox-061724-080621] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
New drug modalities offer life-saving benefits for patients through access to previously undruggable targets. Yet these modalities pose a challenge for the pharmaceutical industry, as side effects are complex, unpredictable, and often uniquely human. With animal studies having limited predictive value due to translatability challenges, the pharmaceutical industry seeks out new approach methodologies. Microphysiological systems (MPS) offer important features that enable complex toxicological processes to be modeled in vitro such as (a) an adjustable complexity of cellular components, including immune components; (b) a modifiable tissue architecture; (c) integration and monitoring of dynamic mechanisms; and (d) a multiorgan connection. Here we review MPS studies in the context of four clinical adverse events triggered by new drug modalities: peripheral neuropathy, thrombocytopenia, immune-mediated hepatotoxicity, and cytokine release syndrome. We conclude that while the use of MPS for testing new drug modality-induced toxicities is still in its infancy, we see strong potential going forward.
Collapse
Affiliation(s)
- Tengku Ibrahim Maulana
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | | - Theodora Kristoforus
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | - Jos Joore
- MIMETAS BV, Oegstgeest, The Netherlands
| | | | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stefan Kustermann
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical Engineering, Faculty of Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | | |
Collapse
|
11
|
Lee Y, Min J, Kim S, Park W, Ko J, Jeon NL. Recapitulating the Cancer-Immunity Cycle on a Chip. Adv Healthc Mater 2025; 14:e2401927. [PMID: 39221688 DOI: 10.1002/adhm.202401927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/11/2024] [Indexed: 09/04/2024]
Abstract
The cancer-immunity cycle is a fundamental framework for understanding how the immune system interacts with cancer cells, balancing T cell recognition and elimination of tumors while avoiding autoimmune reactions. Despite advancements in immunotherapy, there remains a critical need to dissect each phase of the cycle, particularly the interactions among the tumor, vasculature, and immune system within the tumor microenvironment (TME). Innovative platforms such as organ-on-a-chip, organoids, and bioprinting within microphysiological systems (MPS) are increasingly utilized to enhance the understanding of these interactions. These systems meticulously replicate crucial aspects of the TME and immune responses, providing robust platforms to study cancer progression, immune evasion, and therapeutic interventions with greater physiological relevance. This review explores the latest advancements in MPS technologies for modeling various stages of the cancer-immune cycle, critically evaluating their applications and limitations in advancing the understanding of cancer-immune dynamics and guiding the development of next-generation immunotherapeutic strategies.
Collapse
Affiliation(s)
- Yujin Lee
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jaehong Min
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Solbin Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
| | - Wooju Park
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Jihoon Ko
- Department of BioNano Technology, Gachon University, Seongnam-si, Gyeonggi-do, 13120, Republic of Korea
| | - Noo Li Jeon
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea
- Department of Mechanical Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Advanced Machines and Design, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
- Qureator, Inc., San Diego, CA, 92110, USA
| |
Collapse
|
12
|
Geng Q, Xu Y, Hu Y, Wang L, Wang Y, Fan Z, Kong D. Progress in the Application of Organoids-On-A-Chip in Diseases. Organogenesis 2024; 20:2386727. [PMID: 39126669 PMCID: PMC11318694 DOI: 10.1080/15476278.2024.2386727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
With the rapid development of the field of life sciences, traditional 2D cell culture and animal models have long been unable to meet the urgent needs of modern biomedical research and new drug development. Establishing a new generation of experimental models and research models is of great significance for deeply understanding human health and disease processes, and developing effective treatment measures. As is well known, long research and development cycles, high risks, and high costs are the "three mountains" facing the development of new drugs today. Organoids and organ-on-chips technology can highly simulate and reproduce the human physiological environment and complex reactions in vitro, greatly improving the accuracy of drug clinical efficacy prediction, reducing drug development costs, and avoiding the defects of drug testing animal models. Therefore, organ-on-chips have enormous potential in medical diagnosis and treatment.
Collapse
Affiliation(s)
- Qiao Geng
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yanyan Xu
- Department of Anoenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Hu
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Wang
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi Wang
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhimin Fan
- Department of colorectal surgery, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Tonk M, Gupta V, Dhwaj A, Sachdeva M. Current developments and advancements of 3-dimensional printing in personalized medication and drug screening. Drug Metab Pers Ther 2024; 39:167-182. [PMID: 39331538 DOI: 10.1515/dmpt-2024-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/06/2024] [Indexed: 09/29/2024]
Abstract
INTRODUCTION 3-Dimensional printing (3DP) is an additive manufacturing (AM) technique that is expanding quickly because of its low cost and excellent efficiency. The 3D printing industry grew by 19.5 % in 2021 in spite of the COVID-19 epidemic, and by 2026, the worldwide market is expected to be valued up to 37.2 billion US dollars. CONTENT Science Direct, Scopus, MEDLINE, EMBASE, PubMed, DOAJ, and other academic databases provide evidence of the increased interest in 3DP technology and innovative drug delivery approaches in recent times. SUMMARY In this review four main 3DP technologies that are appropriate for pharmaceutical applications: extrusion-based, powder-based, liquid-based, and sheet lamination-based systems are discussed. This study is focused on certain 3DP technologies that may be used to create dosage forms, pharmaceutical goods, and other items with broad regulatory acceptance and technological viability for use in commercial manufacturing. It also discusses pharmaceutical applications of 3DP in drug delivery and drug screening. OUTLOOK The pharmaceutical sector has seen the prospect of 3D printing in risk assessment, medical personalisation, and the manufacture of complicated dose formulas at a reasonable cost. AM has great promise to revolutionise the manufacturing and use of medicines, especially in the field of personalized medicine. The need to understand more about the potential applications of 3DP in medical and pharmacological contexts has grown over time.
Collapse
Affiliation(s)
- Megha Tonk
- Raj Kumar Goel Institute of Technology (Pharmacy), Ghaziabad, Uttar Pradesh, India
| | - Vishal Gupta
- Raj Kumar Goel Institute of Technology (Pharmacy), Ghaziabad, Uttar Pradesh, India
| | | | - Monika Sachdeva
- Raj Kumar Goel Institute of Technology (Pharmacy), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
14
|
Camara Dit Pinto S, Cherkaoui J, Ghosh D, Cazaubon V, Benzeroual KE, Levine SM, Cherkaoui M, Sood GK, Anandasabapathy S, Dhingra S, Vierling JM, Gallo NR. A virtual scalable model of the Hepatic Lobule for acetaminophen hepatotoxicity prediction. NPJ Digit Med 2024; 7:340. [PMID: 39604584 PMCID: PMC11603025 DOI: 10.1038/s41746-024-01349-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
Addressing drug-induced liver injury is crucial in drug development, often causing Phase III trial failures and market withdrawals. Traditional animal models fail to predict human liver toxicity accurately. Virtual twins of human organs present a promising solution. We introduce the Virtual Hepatic Lobule, a foundational element of the Living Liver, a multi-scale liver virtual twin. This model integrates blood flow dynamics and an acetaminophen-induced injury model to predict hepatocyte injury patterns specific to patients. By incorporating metabolic zonation, our predictions align with clinical zonal hepatotoxicity observations. This methodology advances the development of a human liver virtual twin, aiding in the prediction and validation of drug-induced liver injuries.
Collapse
Affiliation(s)
- Stelian Camara Dit Pinto
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Jalal Cherkaoui
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
- Institut National des Sciences Appliquées, Lyon, France
| | - Debarshi Ghosh
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Valentine Cazaubon
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Kenza E Benzeroual
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA
| | | | - Mohammed Cherkaoui
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA
| | - Gagan K Sood
- Division of Gastroenterology, Baylor College of Medicine, Houston, TX, USA
| | | | - Sadhna Dhingra
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - John M Vierling
- Departments of Medicine and Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Nicolas R Gallo
- Department of Computer Science, Digital Engineering and Artificial Intelligence, Long Island University, Brooklyn, NY, USA.
- Arnold & Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, NY, USA.
| |
Collapse
|
15
|
Guo K, van den Beucken T. Advances in drug-induced liver injury research: in vitro models, mechanisms, omics and gene modulation techniques. Cell Biosci 2024; 14:134. [PMID: 39488681 PMCID: PMC11531151 DOI: 10.1186/s13578-024-01317-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Drug-induced liver injury (DILI) refers to drug-mediated damage to the structure and function of the liver, ranging from mild elevation of liver enzymes to severe hepatic insufficiency, and in some cases, progressing to liver failure. The mechanisms and clinical symptoms of DILI are diverse due to the varying combination of drugs, making clinical treatment and prevention complex. DILI has significant public health implications and is the primary reason for post-marketing drug withdrawals. The search for reliable preclinical models and validated biomarkers to predict and investigate DILI can contribute to a more comprehensive understanding of adverse effects and drug safety. In this review, we examine the progress of research on DILI, enumerate in vitro models with potential benefits, and highlight cellular molecular perturbations that may serve as biomarkers. Additionally, we discuss omics approaches frequently used to gather comprehensive datasets on molecular events in response to drug exposure. Finally, three commonly used gene modulation techniques are described, highlighting their application in identifying causal relationships in DILI. Altogether, this review provides a thorough overview of ongoing work and approaches in the field of DILI.
Collapse
Affiliation(s)
- Kaidi Guo
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands.
| | - Twan van den Beucken
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
16
|
Zhao Y, Yin N, Yang R, Faiola F. Recent advances in environmental toxicology: Exploring gene editing, organ-on-a-chip, chimeric animals, and in silico models. Food Chem Toxicol 2024; 193:115022. [PMID: 39326696 DOI: 10.1016/j.fct.2024.115022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/05/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
In our daily life, we are exposed to various environmental pollutants in multiple ways. At present, we mainly rely on animal models and two-dimensional cell culture models to evaluate the toxicity of environmental pollutants. Nevertheless, results in animal models do not always apply to humans because of differences between species, while two-dimensional cell culture models cannot replicate the in vivo microenvironments, making it difficult to predict the true toxic response of environmental pollutants in humans. The development of various high-end technologies in recent years has provided new opportunities for environmental toxicology research. The application of these high-end technologies in environmental toxicology can complement the limitations of traditional environmental toxicology screening and more accurately predict the toxicity of environmental pollutants. In this review, we first introduce the advantages and disadvantages of traditional environmental toxicology methods, then review the principles and development of four high-end technologies, such as gene editing, organ-on-a-chip, chimeric animals, and in silico models, summarize their application in toxicity testing, and finally emphasize their importance/potential in environmental toxicology.
Collapse
Affiliation(s)
- Yanyi Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
17
|
Man Y, Liu Y, Chen Q, Zhang Z, Li M, Xu L, Tan Y, Liu Z. Organoids-On-a-Chip for Personalized Precision Medicine. Adv Healthc Mater 2024:e2401843. [PMID: 39397335 DOI: 10.1002/adhm.202401843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 08/25/2024] [Indexed: 10/15/2024]
Abstract
The development of personalized precision medicine has become a pivotal focus in modern healthcare. Organoids-on-a-Chip (OoCs), a groundbreaking fusion of organoid culture and microfluidic chip technology, has emerged as a promising approach to advancing patient-specific treatment strategies. In this review, the diverse applications of OoCs are explored, particularly their pivotal role in personalized precision medicine, and their potential as a cutting-edge technology is highlighted. By utilizing patient-derived organoids, OoCs offer a pathway to optimize treatments, create precise disease models, investigate disease mechanisms, conduct drug screenings, and individualize therapeutic strategies. The emphasis is on the significance of this technological fusion in revolutionizing healthcare and improving patient outcomes. Furthermore, the transformative potential of personalized precision medicine, future prospects, and ongoing advancements in the field, with a focus on genomic medicine, multi-omics integration, and ethical frameworks are discussed. The convergence of these innovations can empower patients, redefine treatment approaches, and shape the future of healthcare.
Collapse
Affiliation(s)
- Yunqi Man
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Zhirou Zhang
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| |
Collapse
|
18
|
Dey S, Bhat A, Janani G, Shandilya V, Gupta R, Mandal BB. Microfluidic human physiomimetic liver model as a screening platform for drug induced liver injury. Biomaterials 2024; 310:122627. [PMID: 38823194 DOI: 10.1016/j.biomaterials.2024.122627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/19/2024] [Indexed: 06/03/2024]
Abstract
The pre-clinical animal models often fail to predict intrinsic and idiosyncratic drug induced liver injury (DILI), thus contributing to drug failures in clinical trials, black box warnings and withdrawal of marketed drugs. This suggests a critical need for human-relevant in vitro models to predict diverse DILI phenotypes. In this study, a porcine liver extracellular matrix (ECM) based biomaterial ink with high printing fidelity, biocompatibility and tunable rheological and mechanical properties is formulated for supporting both parenchymal and non-parenchymal cells. Further, we applied 3D printing and microfluidic technology to bioengineer a human physiomimetic liver acinus model (HPLAM), recapitulating the radial hepatic cord-like structure with functional sinusoidal microvasculature network, biochemical and biophysical properties of native liver acinus. Intriguingly, the human derived hepatic cells incorporated HPLAM cultured under physiologically relevant microenvironment, acts as metabolic biofactories manifesting enhanced hepatic functionality, secretome levels and biomarkers expression over several weeks. We also report that the matured HPLAM reproduces dose- and time-dependent hepatotoxic response of human clinical relevance to drugs typically recognized for inducing diverse DILI phenotypes as compared to conventional static culture. Overall, the developed HPLAM emulates in vivo like functions and may provide a useful platform for DILI risk assessment to better determine safety and human risk.
Collapse
Affiliation(s)
- Souradeep Dey
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Amritha Bhat
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - G Janani
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Vartik Shandilya
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Raghvendra Gupta
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Department of Chemical Engineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Biman B Mandal
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India; Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
19
|
Majer J, Alex A, Shi J, Chaney EJ, Mukherjee P, Spillman DR, Marjanovic M, Newman CF, Groseclose RM, Watson PD, Boppart SA, Hood SR. Multimodal imaging of a liver-on-a-chip model using labelled and label-free optical microscopy techniques. LAB ON A CHIP 2024; 24:4594-4608. [PMID: 39258913 PMCID: PMC12013482 DOI: 10.1039/d4lc00504j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
A liver-on-a-chip model is an advanced complex in vitro model (CIVM) that incorporates different cell types and extracellular matrix to mimic the microenvironment of the human liver in a laboratory setting. Given the heterogenous and complex nature of liver-on-a-chip models, brightfield and fluorescence-based imaging techniques are widely utilized for assessing the changes occurring in these models with different treatment and environmental conditions. However, the utilization of optical microscopy techniques for structural and functional evaluation of the liver CIVMs have been limited by the reduced light penetration depth and lack of 3D information obtained using these imaging techniques. In this study, the potential of both labelled as well as label-free multimodal optical imaging techniques for visualization and characterization of the cellular and sub-cellular features of a liver-on-a-chip model was investigated. (1) Cellular uptake and distribution of Alexa 488 (A488)-labelled non-targeted and targeted antisense oligonucleotides (ASO and ASO-GalNAc) in the liver-on-a-chip model was determined using multiphoton microscopy. (2) Hyperspectral stimulated Raman scattering (SRS) microscopy of the C-H region was used to determine the heterogeneity of chemical composition of circular and cuboidal hepatocytes in the liver-on-a-chip model in a label-free manner. Additionally, the spatial overlap between the intracellular localization of ASO and lipid droplets was explored using simultaneous hyperspectral SRS and fluorescence microscopy. (3) The capability of light sheet fluorescence microscopy (LSFM) for full-depth 3D visualization of sub-cellular distribution of A488-ASO and cellular phenotypes in the liver-on-a-chip model was demonstrated. In summary, multimodal optical microscopy is a promising platform that can be utilized for visualization and quantification of 3D cellular organization, drug distribution and functional changes occurring in liver-on-a-chip models, and can provide valuable insights into liver biology and drug uptake mechanisms by enabling better characterization of these liver models.
Collapse
Affiliation(s)
- Jan Majer
- Pre-Clinical Sciences, Research Technologies, GSK, Stevenage, UK.
- School of Biosciences, Cardiff University, Cardiff, UK.
| | - Aneesh Alex
- Pre-Clinical Sciences, Research Technologies, GSK, Collegeville, PA, USA
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Jindou Shi
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Eric J Chaney
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Prabuddha Mukherjee
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Darold R Spillman
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- NIH/NIBIB P41 Center for Label-Free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Marina Marjanovic
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- NIH/NIBIB P41 Center for Label-Free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Carla F Newman
- Pre-Clinical Sciences, Research Technologies, GSK, Stevenage, UK.
| | - Reid M Groseclose
- Pre-Clinical Sciences, Research Technologies, GSK, Collegeville, PA, USA
| | | | - Stephen A Boppart
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Electrical and Computer Engineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- NIH/NIBIB P41 Center for Label-Free Imaging and Multiscale Biophotonics (CLIMB), University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Interdisciplinary Health Sciences Institute, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Steve R Hood
- Pre-Clinical Sciences, Research Technologies, GSK, Stevenage, UK.
- GSK Center for Optical Molecular Imaging, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
20
|
Kim J, Ro J, Cho YK. Vascularized platforms for investigating cell communication via extracellular vesicles. BIOMICROFLUIDICS 2024; 18:051504. [PMID: 39323481 PMCID: PMC11421861 DOI: 10.1063/5.0220840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
The vascular network plays an essential role in the maintenance of all organs in the body via the regulated delivery of oxygen and nutrients, as well as tissue communication via the transfer of various biological signaling molecules. It also serves as a route for drug administration and affects pharmacokinetics. Due to this importance, engineers have sought to create physiologically relevant and reproducible vascular systems in tissue, considering cell-cell and extracellular matrix interaction with structural and physical conditions in the microenvironment. Extracellular vesicles (EVs) have recently emerged as important carriers for transferring proteins and genetic material between cells and organs, as well as for drug delivery. Vascularized platforms can be an ideal system for studying interactions between blood vessels and EVs, which are crucial for understanding EV-mediated substance transfer in various biological situations. This review summarizes recent advances in vascularized platforms, standard and microfluidic-based techniques for EV isolation and characterization, and studies of EVs in vascularized platforms. It provides insights into EV-related (patho)physiological regulations and facilitates the development of EV-based therapeutics.
Collapse
|
21
|
Park SY, Koh WG, Lee HJ. Enhanced hepatotoxicity assessment through encapsulated HepG2 spheroids in gelatin hydrogel matrices: Bridging the gap from 2D to 3D culture. Eur J Pharm Biopharm 2024; 202:114417. [PMID: 39013493 DOI: 10.1016/j.ejpb.2024.114417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/07/2024] [Accepted: 07/13/2024] [Indexed: 07/18/2024]
Abstract
Conventional 2D drug screening often fails to accurately predict clinical outcomes. We present an innovative approach to improve hepatotoxicity assessment by encapsulating HepG2 spheroids in gelatin hydrogel matrices with different mechanical properties. Encapsulated spheroids exhibit sustained liver-specific functionality, enhanced expression of drug-metabolizing enzymes, and increased drug sensitivity compared to 2D cultures. The platform detects critical variations in drug response, with significant differences in IC50 values between 2D and spheroid cultures ranging from 1.3-fold to > 13-fold, particularly for acetaminophen. Furthermore, drug-metabolizing enzyme expression varies across hydrogel concentrations, suggesting a role for matrix mechanical properties in modulating hepatocyte function. This novel spheroid-hydrogel platform offers a transformative approach to hepatotoxicity assessment, providing increased sensitivity, improved prediction, and a more physiologically relevant environment. The use of such advanced in vitro models can accelerate drug development, reduce animal testing, and contribute to improved patient safety and clinical outcomes.
Collapse
Affiliation(s)
- Se Yeon Park
- School of Chemical, Biological and Battery Engineering, Gachon University, 1342 Seongnam-daero, Seongnam-si, Gyeonggi-do 13120, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Hyun Jong Lee
- School of Chemical, Biological and Battery Engineering, Gachon University, 1342 Seongnam-daero, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| |
Collapse
|
22
|
Kang S, Chen EC, Cifuentes H, Co JY, Cole G, Graham J, Hsia R, Kiyota T, Klein JA, Kroll KT, Nieves Lopez LM, Norona LM, Peiris H, Potla R, Romero-Lopez M, Roth JG, Tseng M, Fullerton AM, Homan KA. Complex in vitromodels positioned for impact to drug testing in pharma: a review. Biofabrication 2024; 16:042006. [PMID: 39189069 DOI: 10.1088/1758-5090/ad6933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.
Collapse
Affiliation(s)
- Serah Kang
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Eugene C Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Helen Cifuentes
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julia Y Co
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Gabrielle Cole
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica Graham
- Product Quality & Occupational Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of Americaica
| | - Rebecca Hsia
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Tomomi Kiyota
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica A Klein
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Katharina T Kroll
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Lenitza M Nieves Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Leah M Norona
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Heshan Peiris
- Human Genetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Ratnakar Potla
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Monica Romero-Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julien G Roth
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Min Tseng
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Aaron M Fullerton
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Kimberly A Homan
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| |
Collapse
|
23
|
Mankhong S, Den-Udom T, Tanawattanasuntorn T, Suriyun T, Muta K, Kitiyakara C, Ketsawatsomkron P. The microbial metabolite p-cresol compromises the vascular barrier and induces endothelial cytotoxicity and inflammation in a 3D human vessel-on-a-chip. Sci Rep 2024; 14:18553. [PMID: 39122790 PMCID: PMC11316076 DOI: 10.1038/s41598-024-69124-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Increased protein-bound uremic toxins (PBUTs) in patients with chronic kidney disease (CKD) are associated with cardiovascular diseases (CVDs); however, whether retention of PBUTs causes CVD remains unclear. Previous studies assessing the impacts of PBUTs on the vasculature have relied on 2D cell cultures lacking in vivo microenvironments. Here, we investigated the impact of various PBUTs (p-cresol (PC), indoxyl sulfate (IS), and p-cresyl sulfate (PCS)) on microvascular function using an organ-on-a-chip (OOC). Human umbilical vein endothelial cells were used to develop 3D vessels. Chronic exposure to PC resulted in significant vascular leakage compared with controls, whereas IS or PCS treatment did not alter the permeability of 3D vessels. Increased permeability induced by PC was correlated with derangement of cell adherens junction complex, vascular endothelial (VE)-cadherin and filamentous (F)-actin. Additionally, PC decreased endothelial viability in a concentration-dependent manner with a lower IC50 in 3D vessels than in 2D cultures. IS slightly decreased cell viability, while PCS did not affect viability. PC induced inflammatory responses by increasing monocyte adhesion to endothelial surfaces of 3D vessels and IL-6 production. In conclusion, this study leveraged an OOC to determine the diverse effects of PBUTs, demonstrating that PC accumulation is detrimental to ECs during kidney insufficiency.
Collapse
Affiliation(s)
- Sakulrat Mankhong
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Moo 14, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Thittaya Den-Udom
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Moo 14, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Tanotnon Tanawattanasuntorn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Moo 14, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Thunwarat Suriyun
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Moo 14, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Kenjiro Muta
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Moo 14, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Chagriya Kitiyakara
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Pimonrat Ketsawatsomkron
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Moo 14, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand.
| |
Collapse
|
24
|
Buron N, Porceddu M, Loyant R, Martel C, Allard JA, Fromenty B, Borgne-Sanchez A. Drug-induced impairment of mitochondrial fatty acid oxidation and steatosis: assessment of causal relationship with 45 pharmaceuticals. Toxicol Sci 2024; 200:369-381. [PMID: 38676573 DOI: 10.1093/toxsci/kfae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2024] Open
Abstract
Drug-induced liver injury (DILI) represents a major issue for pharmaceutical companies, being a potential cause of black-box warnings on marketed pharmaceuticals, or drug withdrawal from the market. Lipid accumulation in the liver also referred to as steatosis, may be secondary to impaired mitochondrial fatty acid oxidation (mtFAO). However, an overall causal relationship between drug-induced mtFAO inhibition and the occurrence of steatosis in patients has not yet been established with a high number of pharmaceuticals. Hence, 32 steatogenic and 13 nonsteatogenic drugs were tested for their ability to inhibit mtFAO in isolated mouse liver mitochondria. To this end, mitochondrial respiration was measured with palmitoyl-l-carnitine, palmitoyl-CoA + l-carnitine, or octanoyl- l-carnitine. This mtFAO tri-parametric assay was able to predict the occurrence of steatosis in patients with a sensitivity and positive predictive value above 88%. To get further information regarding the mechanism of drug-induced mtFAO impairment, mitochondrial respiration was also measured with malate/glutamate or succinate. Drugs such as diclofenac, methotrexate, and troglitazone could inhibit mtFAO secondary to an impairment of the mitochondrial respiratory chain, whereas dexamethasone, olanzapine, and zidovudine appeared to impair mtFAO directly. Mitochondrial swelling, transmembrane potential, and production of reactive oxygen species were also assessed for all compounds. Only the steatogenic drugs amiodarone, ketoconazole, lovastatin, and toremifene altered all these 3 mitochondrial parameters. In conclusion, our tri-parametric mtFAO assay could be useful in predicting the occurrence of steatosis in patients. The combination of this assay with other mitochondrial parameters could also help to better understand the mechanism of drug-induced mtFAO inhibition.
Collapse
Affiliation(s)
- Nelly Buron
- MITOLOGICS S.A.S., Faculté de Médecine, Créteil 94000, France
| | | | - Roxane Loyant
- MITOLOGICS S.A.S., Faculté de Médecine, Créteil 94000, France
| | - Cécile Martel
- MITOLOGICS S.A.S., Faculté de Médecine, Créteil 94000, France
| | - Julien A Allard
- INSERM, INRAE, Univ Rennes, Institut NUMECAN, UMR_S1317, Rennes 35000, France
| | - Bernard Fromenty
- INSERM, INRAE, Univ Rennes, Institut NUMECAN, UMR_S1317, Rennes 35000, France
| | | |
Collapse
|
25
|
Mehta V, Karnam G, Madgula V. Liver-on-chips for drug discovery and development. Mater Today Bio 2024; 27:101143. [PMID: 39070097 PMCID: PMC11279310 DOI: 10.1016/j.mtbio.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Recent FDA modernization act 2.0 has led to increasing industrial R&D investment in advanced in vitro 3D models such as organoids, spheroids, organ-on-chips, 3D bioprinting, and in silico approaches. Liver-related advanced in vitro models remain the prime area of interest, as liver plays a central role in drug clearance of compounds. Growing evidence indicates the importance of recapitulating the overall liver microenvironment to enhance hepatocyte maturity and culture longevity using liver-on-chips (LoC) in vitro. Hence, pharmaceutical industries have started exploring LoC assays in the two of the most challenging areas: accurate in vitro-in vivo extrapolation (IVIVE) of hepatic drug clearance and drug-induced liver injury. We examine the joint efforts of commercial chip manufacturers and pharmaceutical companies to present an up-to-date overview of the adoption of LoC technology in the drug discovery. Further, several roadblocks are identified to the rapid adoption of LoC assays in the current drug development framework. Finally, we discuss some of the underexplored application areas of LoC models, where conventional 2D hepatic models are deemed unsuitable. These include clearance prediction of metabolically stable compounds, immune-mediated drug-induced liver injury (DILI) predictions, bioavailability prediction with gut-liver systems, hepatic clearance prediction of drugs given during pregnancy, and dose adjustment studies in disease conditions. We conclude the review by discussing the importance of PBPK modeling with LoC, digital twins, and AI/ML integration with LoC.
Collapse
Affiliation(s)
- Viraj Mehta
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Guruswamy Karnam
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Vamsi Madgula
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| |
Collapse
|
26
|
Kim J, Yoon T, Lee S, Kim PJ, Kim Y. Reconstitution of human tissue barrier function for precision and personalized medicine. LAB ON A CHIP 2024; 24:3347-3366. [PMID: 38895863 DOI: 10.1039/d4lc00104d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Tissue barriers in a body, well known as tissue-to-tissue interfaces represented by endothelium of the blood vessels or epithelium of organs, are essential for maintaining physiological homeostasis by regulating molecular and cellular transports. It is crucial for predicting drug response to understand physiology of tissue barriers through which drugs are absorbed, distributed, metabolized and excreted. Since the FDA Modernization Act 2.0, which prompts the inception of alternative technologies for animal models, tissue barrier chips, one of the applications of organ-on-a-chip or microphysiological system (MPS), have only recently been utilized in the context of drug development. Recent advancements in stem cell technology have brightened the prospects for the application of tissue barrier chips in personalized medicine. In past decade, designing and engineering these microfluidic devices, and demonstrating the ability to reconstitute tissue functions were main focus of this field. However, the field is now advancing to the next level of challenges: validating their utility in drug evaluation and creating personalized models using patient-derived cells. In this review, we briefly introduce key design parameters to develop functional tissue barrier chip, explore the remarkable recent progress in the field of tissue barrier chips and discuss future perspectives on realizing personalized medicine through the utilization of tissue barrier chips.
Collapse
Affiliation(s)
- Jaehoon Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Taehee Yoon
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sungryeong Lee
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Paul J Kim
- Department of Psychiatry & Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - YongTae Kim
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
27
|
Jadalannagari S, Ewart L. Beyond the hype and toward application: liver complex in vitro models in preclinical drug safety. Expert Opin Drug Metab Toxicol 2024; 20:607-619. [PMID: 38465923 DOI: 10.1080/17425255.2024.2328794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Drug induced Liver-Injury (DILI) is a leading cause of drug attrition and complex in vitro models (CIVMs), including three dimensional (3D) spheroids, 3D bio printed tissues and flow-based systems, could improve preclinical prediction. Although CIVMs have demonstrated good sensitivity and specificity in DILI detection their adoption remains limited. AREAS COVERED This article describes DILI, the challenges with its prediction and the current strategies and models that are being used. It reviews data from industry-FDA collaborations and strategic partnerships and finishes with an outlook of CIVMs in preclinical toxicity testing. Literature searches were performed using PubMed and Google Scholar while product information was collected from manufacturer websites. EXPERT OPINION Liver CIVMs are promising models for predicting DILI although, a decade after their introduction, routine use by the pharmaceutical industry is limited. To accelerate their adoption, several industry-regulator-developer partnerships or consortia have been established to guide the development and qualification. Beyond this, liver CIVMs should continue evolving to capture greater immunological mimicry while partnering with computational approaches to deliver systems that change the paradigm of predicting DILI.
Collapse
Affiliation(s)
| | - Lorna Ewart
- Department of Bioinnovations, Emulate Inc, Boston, MA, USA
| |
Collapse
|
28
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
29
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
30
|
Henriquez JE, Badwaik VD, Bianchi E, Chen W, Corvaro M, LaRocca J, Lunsman TD, Zu C, Johnson KJ. From Pipeline to Plant Protection Products: Using New Approach Methodologies (NAMs) in Agrochemical Safety Assessment. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10710-10724. [PMID: 38688008 DOI: 10.1021/acs.jafc.4c00958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The human population will be approximately 9.7 billion by 2050, and food security has been identified as one of the key issues facing the global population. Agrochemicals are an important tool available to farmers that enable high crop yields and continued access to healthy foods, but the average new agrochemical active ingredient takes more than ten years, 350 million dollars, and 20,000 animals to develop and register. The time, monetary, and animal costs incentivize the use of New Approach Methodologies (NAMs) in early-stage screening to prioritize chemical candidates. This review outlines NAMs that are currently available or can be adapted for use in early-stage screening agrochemical programs. It covers new in vitro screens that are on the horizon in key areas of regulatory concern. Overall, early-stage screening with NAMs enables the prioritization of development for agrochemicals without human and environmental health concerns through a more directed, agile, and iterative development program before animal-based regulatory testing is even considered.
Collapse
Affiliation(s)
| | - Vivek D Badwaik
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Enrica Bianchi
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Wei Chen
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | | | - Jessica LaRocca
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | | | - Chengli Zu
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| | - Kamin J Johnson
- Corteva Agriscience, Indianapolis, Indiana 46268, United States
| |
Collapse
|
31
|
AlShmmari SK, Fardous RS, Shinwari Z, Cialla-May D, Popp J, Ramadan Q, Zourob M. Hepatic spheroid-on-a-chip: Fabrication and characterization of a spheroid-based in vitro model of the human liver for drug screening applications. BIOMICROFLUIDICS 2024; 18:034105. [PMID: 38817733 PMCID: PMC11136519 DOI: 10.1063/5.0210955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
The integration of microfabrication and microfluidics techniques into cell culture technology has significantly transformed cell culture conditions, scaffold architecture, and tissue biofabrication. These tools offer precise control over cell positioning and enable high-resolution analysis and testing. Culturing cells in 3D systems, such as spheroids and organoids, enables recapitulating the interaction between cells and the extracellular matrix, thereby allowing the creation of human-based biomimetic tissue models that are well-suited for pre-clinical drug screening. Here, we demonstrate an innovative microfluidic device for the formation, culture, and testing of hepatocyte spheroids, which comprises a large array of patterned microwells for hosting hepatic spheroid culture in a reproducible and organized format in a dynamic fluidic environment. The device allows maintaining and characterizing different spheroid sizes as well as exposing to various drugs in parallel enabling high-throughput experimentation. These liver spheroids exhibit physiologically relevant hepatic functionality, as evidenced by their ability to produce albumin and urea at levels comparable to in vivo conditions and the capability to distinguish the toxic effects of selected drugs. This highlights the effectiveness of the microenvironment provided by the chip in maintaining the functionality of hepatocyte spheroids. These data support the notion that the liver-spheroid chip provides a favorable microenvironment for the maintenance of hepatocyte spheroid functionality.
Collapse
Affiliation(s)
| | | | - Zakia Shinwari
- Cell Therapy and Immunology Department, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | | | | | - Qasem Ramadan
- College of Science & General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Mohammed Zourob
- College of Science & General Studies, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
32
|
Feng L, Wang Y, Fu Y, Li T, He G. Stem Cell-Based Strategies: The Future Direction of Bioartificial Liver Development. Stem Cell Rev Rep 2024; 20:601-616. [PMID: 38170319 DOI: 10.1007/s12015-023-10672-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Acute liver failure (ALF) results from severe liver damage or end-stage liver disease. It is extremely fatal and causes serious health and economic burdens worldwide. Once ALF occurs, liver transplantation (LT) is the only definitive and recommended treatment; however, LT is limited by the scarcity of liver grafts. Consequently, the clinical use of bioartificial liver (BAL) has been proposed as a treatment strategy for ALF. Human primary hepatocytes are an ideal cell source for these methods. However, their high demand and superior viability prevent their widespread use. Hence, finding alternatives that meet the seed cell quality and quantity requirements is imperative. Stem cells with self-renewing, immunogenic, and differentiative capacities are potential cell sources. MSCs and its secretomes encompass a spectrum of beneficial properties, such as anti-inflammatory, immunomodulatory, anti-ROS (reactive oxygen species), anti-apoptotic, pro-metabolomic, anti-fibrogenesis, and pro-regenerative attributes. This review focused on the recent status and future directions of stem cell-based strategies in BAL for ALF. Additionally, we discussed the opportunities and challenges associated with promoting such strategies for clinical applications.
Collapse
Affiliation(s)
- Lei Feng
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550000, Guizhou, China.
| | - Yi Wang
- Shanxi Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, Shanxi, China
| | - Yu Fu
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ting Li
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, Guangdong, China.
| | - Guolin He
- Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
33
|
Olaizola-Rodrigo C, Palma-Florez S, Ranđelović T, Bayona C, Ashrafi M, Samitier J, Lagunas A, Mir M, Doblaré M, Ochoa I, Monge R, Oliván S. Tuneable hydrogel patterns in pillarless microfluidic devices. LAB ON A CHIP 2024; 24:2094-2106. [PMID: 38444329 DOI: 10.1039/d3lc01082a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Organ-on-chip (OOC) technology has recently emerged as a powerful tool to mimic physiological or pathophysiological conditions through cell culture in microfluidic devices. One of its main goals is bypassing animal testing and encouraging more personalized medicine. The recent incorporation of hydrogels as 3D scaffolds into microfluidic devices has changed biomedical research since they provide a biomimetic extracellular matrix to recreate tissue architectures. However, this technology presents some drawbacks such as the necessity for physical structures as pillars to confine these hydrogels, as well as the difficulty in reaching different shapes and patterns to create convoluted gradients or more realistic biological structures. In addition, pillars can also interfere with the fluid flow, altering the local shear forces and, therefore, modifying the mechanical environment in the OOC model. In this work, we present a methodology based on a plasma surface treatment that allows building cell culture chambers with abutment-free patterns capable of producing precise shear stress distributions. Therefore, pillarless devices with arbitrary geometries are needed to obtain more versatile, reliable, and biomimetic experimental models. Through computational simulation studies, these shear stress changes are demonstrated in different designed and fabricated geometries. To prove the versatility of this new technique, a blood-brain barrier model has been recreated, achieving an uninterrupted endothelial barrier that emulates part of the neurovascular network of the brain. Finally, we developed a new technology that could avoid the limitations mentioned above, allowing the development of biomimetic OOC models with complex and adaptable geometries, with cell-to-cell contact if required, and where fluid flow and shear stress conditions could be controlled.
Collapse
Affiliation(s)
- Claudia Olaizola-Rodrigo
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
- BEOnChip S.L., Zaragoza, Spain.
| | - Sujey Palma-Florez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Teodora Ranđelović
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- CIBER-BBN, ISCIII, Spain
| | - Clara Bayona
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
| | - Mehran Ashrafi
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
| | - Josep Samitier
- CIBER-BBN, ISCIII, Spain
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Anna Lagunas
- CIBER-BBN, ISCIII, Spain
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Spain
| | - Mònica Mir
- CIBER-BBN, ISCIII, Spain
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| | - Manuel Doblaré
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- CIBER-BBN, ISCIII, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- CIBER-BBN, ISCIII, Spain
| | | | - Sara Oliván
- Tissue Microenvironment (TME), Lab. Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain.
- Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
| |
Collapse
|
34
|
An J, Zhang S, Wu J, Chen H, Xu G, Hou Y, Liu R, Li N, Cui W, Li X, Du Y, Gu Q. Assessing bioartificial organ function: the 3P model framework and its validation. LAB ON A CHIP 2024; 24:1586-1601. [PMID: 38362645 DOI: 10.1039/d3lc01020a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
The rapid advancement in the fabrication and culture of in vitro organs has marked a new era in biomedical research. While strides have been made in creating structurally diverse bioartificial organs, such as the liver, which serves as the focal organ in our study, the field lacks a uniform approach for the predictive assessment of liver function. Our research bridges this gap with the introduction of a novel, machine-learning-based "3P model" framework. This model draws on a decade of experimental data across diverse culture platform studies, aiming to identify critical fabrication parameters affecting liver function, particularly in terms of albumin and urea secretion. Through meticulous statistical analysis, we evaluated the functional sustainability of the in vitro liver models. Despite the diversity of research methodologies and the consequent scarcity of standardized data, our regression model effectively captures the patterns observed in experimental findings. The insights gleaned from our study shed light on optimizing culture conditions and advance the evaluation of the functional maintenance capacity of bioartificial livers. This sets a precedent for future functional evaluations of bioartificial organs using machine learning.
Collapse
Affiliation(s)
- Jingmin An
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| | - Shuyu Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
| | - Juan Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Haolin Chen
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Guoshi Xu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Yifan Hou
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Ruoyu Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
| | - Na Li
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100864, P.R. China.
| | - Wenjuan Cui
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100864, P.R. China.
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Xin Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Yi Du
- Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100864, P.R. China.
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| | - Qi Gu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chaoyang District, Beijing, 100101, P. R. China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, P. R. China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 100149, P. R. China
| |
Collapse
|
35
|
Ko J, Song J, Choi N, Kim HN. Patient-Derived Microphysiological Systems for Precision Medicine. Adv Healthc Mater 2024; 13:e2303161. [PMID: 38010253 PMCID: PMC11469251 DOI: 10.1002/adhm.202303161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Patient-derived microphysiological systems (P-MPS) have emerged as powerful tools in precision medicine that provide valuable insight into individual patient characteristics. This review discusses the development of P-MPS as an integration of patient-derived samples, including patient-derived cells, organoids, and induced pluripotent stem cells, into well-defined MPSs. Emphasizing the necessity of P-MPS development, its significance as a nonclinical assessment approach that bridges the gap between traditional in vitro models and clinical outcomes is highlighted. Additionally, guidance is provided for engineering approaches to develop microfluidic devices and high-content analysis for P-MPSs, enabling high biological relevance and high-throughput experimentation. The practical implications of the P-MPS are further examined by exploring the clinically relevant outcomes obtained from various types of patient-derived samples. The construction and analysis of these diverse samples within the P-MPS have resulted in physiologically relevant data, paving the way for the development of personalized treatment strategies. This study describes the significance of the P-MPS in precision medicine, as well as its unique capacity to offer valuable insights into individual patient characteristics.
Collapse
Affiliation(s)
- Jihoon Ko
- Department of BioNano TechnologyGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
| | - Jiyoung Song
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
| | - Nakwon Choi
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- KU‐KIST Graduate School of Converging Science and TechnologyKorea UniversitySeoul02841Republic of Korea
| | - Hong Nam Kim
- Brain Science InstituteKorea Institute of Science and Technology (KIST)Seoul02792Republic of Korea
- Division of Bio‐Medical Science & TechnologyKIST SchoolSeoul02792Republic of Korea
- School of Mechanical EngineeringYonsei UniversitySeoul03722Republic of Korea
- Yonsei‐KIST Convergence Research InstituteYonsei UniversitySeoul03722Republic of Korea
| |
Collapse
|
36
|
Bai H, Olson KNP, Pan M, Marshall T, Singh H, Ma J, Gilbride P, Yuan Y, McCormack J, Si L, Maharjan S, Huang D, Qian X, Livermore C, Zhang YS, Xie X. Rapid Prototyping of Thermoplastic Microfluidic 3D Cell Culture Devices by Creating Regional Hydrophilicity Discrepancy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304332. [PMID: 38032118 PMCID: PMC10870023 DOI: 10.1002/advs.202304332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/02/2023] [Indexed: 12/01/2023]
Abstract
Microfluidic 3D cell culture devices that enable the recapitulation of key aspects of organ structures and functions in vivo represent a promising preclinical platform to improve translational success during drug discovery. Essential to these engineered devices is the spatial patterning of cells from different tissue types within a confined microenvironment. Traditional fabrication strategies lack the scalability, cost-effectiveness, and rapid prototyping capabilities required for industrial applications, especially for processes involving thermoplastic materials. Here, an approach to pattern fluid guides inside microchannels is introduced by establishing differential hydrophilicity using pressure-sensitive adhesives as masks and a subsequent selective coating with a biocompatible polymer. Optimal coating conditions are identified using polyvinylpyrrolidone, which resulted in rapid and consistent hydrogel flow in both the open-chip prototype and the fully bonded device containing additional features for medium perfusion. The suitability of the device for dynamic 3D cell culture is tested by growing human hepatocytes in the device under controlled fluid flow for a 14-day period. Additionally, the study demonstrated the potential of using the device for pharmaceutical high-throughput screening applications, such as predicting drug-induced liver injury. The approach offers a facile strategy of rapid prototyping thermoplastic microfluidic organ chips with varying geometries, microstructures, and substrate materials.
Collapse
Affiliation(s)
| | | | - Ming Pan
- Xellar BiosystemsCambridgeMA02458USA
| | | | | | | | | | | | | | - Longlong Si
- CAS Key Laboratory of Quantitative Engineering BiologyShenzhen Institute of Synthetic BiologyShenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055P. R. China
- University of Chinese Academy of SciencesBeijing100049P. R. China
| | - Sushila Maharjan
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMA02142USA
| | - Di Huang
- Research Center for Nano‐biomaterials & Regenerative MedicineCollege of Biomedical EngineeringTaiyuan University of TechnologyTaiyuan030024P. R. China
| | | | - Carol Livermore
- Department of Mechanical and Industrial EngineeringNortheastern UniversityBostonMA02115USA
| | - Yu Shrike Zhang
- Division of Engineering in MedicineDepartment of MedicineBrigham and Women's HospitalHarvard Medical SchoolCambridgeMA02142USA
| | - Xin Xie
- Xellar BiosystemsCambridgeMA02458USA
| |
Collapse
|
37
|
Hajam MI, Khan MM. Microfluidics: a concise review of the history, principles, design, applications, and future outlook. Biomater Sci 2024; 12:218-251. [PMID: 38108438 DOI: 10.1039/d3bm01463k] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Microfluidic technologies have garnered significant attention due to their ability to rapidly process samples and precisely manipulate fluids in assays, making them an attractive alternative to conventional experimental methods. With the potential for revolutionary capabilities in the future, this concise review provides readers with insights into the fascinating world of microfluidics. It begins by introducing the subject's historical background, allowing readers to familiarize themselves with the basics. The review then delves into the fundamental principles, discussing the underlying phenomena at play. Additionally, it highlights the different aspects of microfluidic device design, classification, and fabrication. Furthermore, the paper explores various applications, the global market, recent advancements, and challenges in the field. Finally, the review presents a positive outlook on trends and draws lessons to support the future flourishing of microfluidic technologies.
Collapse
Affiliation(s)
- Mohammad Irfan Hajam
- Department of Mechanical Engineering, National Institute of Technology Srinagar, India.
| | - Mohammad Mohsin Khan
- Department of Mechanical Engineering, National Institute of Technology Srinagar, India.
| |
Collapse
|
38
|
Leal F, Zeiringer S, Jeitler R, Costa PF, Roblegg E. A comprehensive overview of advanced dynamic in vitro intestinal and hepatic cell culture models. Tissue Barriers 2024; 12:2163820. [PMID: 36680530 PMCID: PMC10832944 DOI: 10.1080/21688370.2022.2163820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 12/22/2022] [Indexed: 01/22/2023] Open
Abstract
Orally administered drugs pass through the gastrointestinal tract before being absorbed in the small intestine and metabolised in the liver. To test the efficacy and toxicity of drugs, animal models are often employed; however, they are not suitable for investigating drug-tissue interactions and making reliable predictions, since the human organism differs drastically from animals in terms of absorption, distribution, metabolism and excretion of substances. Likewise, simple static in vitro cell culture systems currently used in preclinical drug screening often do not resemble the native characteristics of biological barriers. Dynamic models, on the other hand, provide in vivo-like cell phenotypes and functionalities that offer great potential for safety and efficacy prediction. Herein, current microfluidic in vitro intestinal and hepatic models are reviewed, namely single- and multi-tissue micro-bioreactors, which are associated with different methods of cell cultivation, i.e., scaffold-based versus scaffold-free.
Collapse
Affiliation(s)
- Filipa Leal
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Scarlett Zeiringer
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| | - Ramona Jeitler
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| | - Pedro F. Costa
- BIOFABICS, Rua Alfredo Allen 455, 4200-135 Porto, Portugal
| | - Eva Roblegg
- Department of Pharmaceutical Technology and Biopharmacy, University of Graz, Institute of Pharmaceutical Sciences, Universitaetsplatz 1, Graz, Austria
| |
Collapse
|
39
|
Rispoli P, Scandiuzzi Piovesan T, Decorti G, Stocco G, Lucafò M. iPSCs as a groundbreaking tool for the study of adverse drug reactions: A new avenue for personalized therapy. WIREs Mech Dis 2024; 16:e1630. [PMID: 37770042 DOI: 10.1002/wsbm.1630] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/10/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023]
Abstract
Induced pluripotent stem cells (iPSCs), obtained by reprogramming different somatic cell types, represent a promising tool for the study of drug toxicities, especially in the context of personalized medicine. Indeed, these cells retain the same genetic heritage of the donor, allowing the development of personalized models. In addition, they represent a useful tool for the study of adverse drug reactions (ADRs) in special populations, such as pediatric patients, which are often poorly represented in clinical trials due to ethical issues. Particularly, iPSCs can be differentiated into any tissue of the human body, following several protocols which use different stimuli to induce specific differentiation processes. Differentiated cells also maintain the genetic heritage of the donor, and therefore are suitable for personalized pharmacological studies; moreover, iPSC-derived differentiated cells are a valuable tool for the investigation of the mechanisms underlying the physiological differentiation processes. iPSCs-derived organoids represent another important tool for the study of ADRs. Precisely, organoids are in vitro 3D models which better represent the native organ, both from a structural and a functional point of view. Moreover, in the same way as iPSC-derived 2D models, iPSC-derived organoids are appropriate personalized models since they retain the genetic heritage of the donor. In comparison to other in vitro models, iPSC-derived organoids present advantages in terms of versatility, patient-specificity, and ethical issues. This review aims to provide an updated report of the employment of iPSCs, and 2D and 3D models derived from these, for the study of ADRs. This article is categorized under: Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- Paola Rispoli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | | | - Giuliana Decorti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Gabriele Stocco
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
- Institute for Maternal and Child Health IRCCS Burlo Garofolo, Trieste, Italy
| | - Marianna Lucafò
- Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
40
|
Xie R, Pal V, Yu Y, Lu X, Gao M, Liang S, Huang M, Peng W, Ozbolat IT. A comprehensive review on 3D tissue models: Biofabrication technologies and preclinical applications. Biomaterials 2024; 304:122408. [PMID: 38041911 PMCID: PMC10843844 DOI: 10.1016/j.biomaterials.2023.122408] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
The limitations of traditional two-dimensional (2D) cultures and animal testing, when it comes to precisely foreseeing the toxicity and clinical effectiveness of potential drug candidates, have resulted in a notable increase in the rate of failure during the process of drug discovery and development. Three-dimensional (3D) in-vitro models have arisen as substitute platforms with the capacity to accurately depict in-vivo conditions and increasing the predictivity of clinical effects and toxicity of drug candidates. It has been found that 3D models can accurately represent complex tissue structure of human body and can be used for a wide range of disease modeling purposes. Recently, substantial progress in biomedicine, materials and engineering have been made to fabricate various 3D in-vitro models, which have been exhibited better disease progression predictivity and drug effects than convention models, suggesting a promising direction in pharmaceutics. This comprehensive review highlights the recent developments in 3D in-vitro tissue models for preclinical applications including drug screening and disease modeling targeting multiple organs and tissues, like liver, bone, gastrointestinal tract, kidney, heart, brain, and cartilage. We discuss current strategies for fabricating 3D models for specific organs with their strengths and pitfalls. We expand future considerations for establishing a physiologically-relevant microenvironment for growing 3D models and also provide readers with a perspective on intellectual property, industry, and regulatory landscape.
Collapse
Affiliation(s)
- Renjian Xie
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Vaibhav Pal
- Department of Chemistry, Pennsylvania State University, University Park, PA, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Yanrong Yu
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Xiaolu Lu
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Mengwei Gao
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Shijie Liang
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Miao Huang
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Weijie Peng
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China; School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China.
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA; Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Materials Research Institute, Pennsylvania State University, University Park, PA, USA; Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State University, Hershey, PA, 17033, USA; Department of Medical Oncology, Cukurova University, Adana, 01130, Turkey; Biotechnology Research and Application Center, Cukurova University, Adana, 01130, Turkey.
| |
Collapse
|
41
|
Cherradi S, Taulet N, Duong HT. An original donor-dependent spheroid system for the prediction of idiosyncratic drug-induced liver injury risk. IN VITRO MODELS 2023; 2:281-295. [PMID: 39872500 PMCID: PMC11756448 DOI: 10.1007/s44164-023-00057-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 01/30/2025]
Abstract
One major drawback of preclinical models to test drug-induced liver injury (DILI) is their inability to predict the interindividual difference of DILI effect in a population. Consequently, a high number of molecules that passed preclinical phases, fail clinical trials, and many FDA-approved drugs were removed from the market due to idiosyncratic DILI. We use a proprietary-depleted human serum-based cell educating technology to generate donor-dependent spheroids with distinct morphology and functionality. We demonstrate that educated spheroids could capture the large variations in susceptibility to drug-induced liver injury between donors. We show that the model could predict clinical apparent DILI risk with a high specificity and sensitivity. We provide evidence that the model could address non-genetic factor-associated DILI risk and severity such as age or sex. Our study supports the benefit of using donor-dependent educated spheroids for hepatotoxicity evaluation in preclinical phase or in an exploratory study clinical trial phase 2 to provide a robust safety profile to a drug.
Collapse
Affiliation(s)
- Sara Cherradi
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, Grabels, France
| | - Nicolas Taulet
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, Grabels, France
| | - Hong Tuan Duong
- PredictCan Biotechnologies SAS, Biopôle Euromédecine, Grabels, France
| |
Collapse
|
42
|
Solan ME, Schackmuth B, Bruce ED, Pradhan S, Sayes CM, Lavado R. Effects of short-chain per- and polyfluoroalkyl substances (PFAS) on toxicologically relevant gene expression profiles in a liver-on-a-chip model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 337:122610. [PMID: 37742859 DOI: 10.1016/j.envpol.2023.122610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/23/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023]
Abstract
Short-chain per- and polyfluoroalkyl substances (PFAS) are highly stable and widely used environmental contaminants that pose potential health risks to humans. Aggregating reliable mechanistic information for safety assessments necessitates physiologically relevant high-throughput screening approaches. Here, we demonstrated the utility of a liver-on-a-chip model to investigate the effects of five short-chain PFAS at low (1 nM) and high (1 μM) concentrations on toxicologically-relevant gene expression profiles using the QuantiGene® Plex Assay. We found that the short-chain PFAS tested in this study modulated the expression of ABCG2, a gene encoding for the breast cancer resistance protein (BCRP), with marked and significant upregulation (up to 4-fold) observed for all but one of the short-chain PFAS tested. PFBS and HFPO-DA repressed SLCO1B3 expression, a gene that encodes for an essential liver-specific organic anion transporter. High concentrations of PFBS, PFHxA, and PFHxS upregulated the expression of genes encCYP1A1,CYP2B6 and CYP2C19 with the same treatments resulting in the repression of the expression of the gene encoding CYP1A2. This dysregulation could have consequences for the clearance of endogenous compounds and xenobiotics. However, we acknowledge that increased expression of genes encoding for transporters and biotransformation enzymes may or may not indicate changes to their protein expression or activity. Overall, our study provides important insights into the effects of short-chain PFAS on liver function and their potential implications for human health. The use of the liver-on-a-chip model in combination with the QuantiGene® Plex Assay may be a valuable tool for future high-throughput screening and gene expression profiling in toxicology studies.
Collapse
Affiliation(s)
- Megan E Solan
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Bennett Schackmuth
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Erica D Bruce
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Sahar Pradhan
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Christie M Sayes
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA
| | - Ramon Lavado
- Department of Environmental Science, Baylor University, Waco, TX, 76798, USA.
| |
Collapse
|
43
|
Kim MK, Jeong W, Jeon S, Kang HW. 3D bioprinting of dECM-incorporated hepatocyte spheroid for simultaneous promotion of cell-cell and -ECM interactions. Front Bioeng Biotechnol 2023; 11:1305023. [PMID: 38026892 PMCID: PMC10679743 DOI: 10.3389/fbioe.2023.1305023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
The cell spheroid technology, which greatly enhances cell-cell interactions, has gained significant attention in the development of in vitro liver models. However, existing cell spheroid technologies still have limitations in improving hepatocyte-extracellular matrix (ECM) interaction, which have a significant impact on hepatic function. In this study, we have developed a novel bioprinting technology for decellularized ECM (dECM)-incorporated hepatocyte spheroids that could enhance both cell-cell and -ECM interactions simultaneously. To provide a biomimetic environment, a porcine liver dECM-based cell bio-ink was developed, and a spheroid printing process using this bio-ink was established. As a result, we precisely printed the dECM-incorporated hepatocyte spheroids with a diameter of approximately 160-220 μm using primary mouse hepatocyte (PMHs). The dECM materials were uniformly distributed within the bio-printed spheroids, and even after more than 2 weeks of culture, the spheroids maintained their spherical shape and high viability. The incorporation of dECM also significantly improved the hepatic function of hepatocyte spheroids. Compared to hepatocyte-only spheroids, dECM-incorporated hepatocyte spheroids showed approximately 4.3- and 2.5-fold increased levels of albumin and urea secretion, respectively, and a 2.0-fold increase in CYP enzyme activity. These characteristics were also reflected in the hepatic gene expression levels of ALB, HNF4A, CPS1, and others. Furthermore, the dECM-incorporated hepatocyte spheroids exhibited up to a 1.8-fold enhanced drug responsiveness to representative hepatotoxic drugs such as acetaminophen, celecoxib, and amiodarone. Based on these results, it can be concluded that the dECM-incorporated spheroid printing technology has great potential for the development of highly functional in vitro liver tissue models for drug toxicity assessment.
Collapse
Affiliation(s)
- Min Kyeong Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- Center for Scientific Instrumentation, Korea Basic Science Institute, Chungbuk, Republic of Korea
| | - Wonwoo Jeong
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Seunggyu Jeon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Hyun-Wook Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| |
Collapse
|
44
|
Macedo MH, Dias Neto M, Pastrana L, Gonçalves C, Xavier M. Recent Advances in Cell-Based In Vitro Models to Recreate Human Intestinal Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301391. [PMID: 37736674 PMCID: PMC10625086 DOI: 10.1002/advs.202301391] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/03/2023] [Indexed: 09/23/2023]
Abstract
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.
Collapse
Affiliation(s)
- Maria Helena Macedo
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Mafalda Dias Neto
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Lorenzo Pastrana
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Catarina Gonçalves
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| | - Miguel Xavier
- INL – International Iberian Nanotechnology LaboratoryAvenida Mestre José VeigaBraga4715‐330Portugal
| |
Collapse
|
45
|
Lim AY, Kato Y, Sakolish C, Valdiviezo A, Han G, Bajaj P, Stanko J, Ferguson SS, Villenave R, Hewitt P, Hardwick RN, Rusyn I. Reproducibility and Robustness of a Liver Microphysiological System PhysioMimix LC12 under Varying Culture Conditions and Cell Type Combinations. Bioengineering (Basel) 2023; 10:1195. [PMID: 37892925 PMCID: PMC10603899 DOI: 10.3390/bioengineering10101195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The liver is one of the key organs for exogenous and endogenous metabolism and is often a target for drug- and chemical-driven toxicity. A wide range of experimental approaches has been established to model and characterize the mechanisms of drug- and chemical-induced hepatotoxicity. A number of microfluidics-enabled in vitro models of the liver have been developed, but the unclear translatability of these platforms has hindered their adoption by the pharmaceutical industry; to achieve wide use for drug and chemical safety evaluation, demonstration of reproducibility and robustness under various contexts of use is required. One of these commercially available platforms is the PhysioMimix LC12, a microfluidic device where cells are seeded into a 3D scaffold that is continuously perfused with recirculating cell culture media to mimic liver sinusoids. Previous studies demonstrated this model's functionality and potential applicability to preclinical drug development. However, to gain confidence in PhysioMimix LC12's robustness and reproducibility, supplementary characterization steps are needed, including the assessment of various human hepatocyte sources, contribution of non-parenchymal cells (NPCs), and comparison to other models. In this study, we performed replicate studies averaging 14 days with either primary human hepatocytes (PHHs) or induced pluripotent stem cell (iPSC)-derived hepatocytes, with and without NPCs. Albumin and urea secretion, lactate dehydrogenase, CYP3A4 activity, and metabolism were evaluated to assess basal function and metabolic capacity. Model performance was characterized by different cell combinations under intra- and inter-experimental replication and compared to multi-well plates and other liver platforms. PhysioMimix LC12 demonstrated the highest metabolic function with PHHs, with or without THP-1 or Kupffer cells, for up to 10-14 days. iPSC-derived hepatocytes and PHHs co-cultured with additional NPCs demonstrated sub-optimal performance. Power analyses based on replicate experiments and different contexts of use will inform future study designs due to the limited throughput and high cell demand. Overall, this study describes a workflow for independent testing of a complex microphysiological system for specific contexts of use, which may increase end-user adoption in drug development.
Collapse
Affiliation(s)
- Alicia Y. Lim
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Yuki Kato
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
- Laboratory for Drug Discovery and Development, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Courtney Sakolish
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Alan Valdiviezo
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| | - Gang Han
- Department of Epidemiology and Biostatistics, Texas A&M University, College Station, TX 77843, USA
| | - Piyush Bajaj
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02141, USA
| | - Jason Stanko
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Stephen S. Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Remi Villenave
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293 Darmstadt, Germany
| | - Rhiannon N. Hardwick
- Discovery Toxicology, Pharmaceutical Candidate Optimization, Bristol Myers Squibb, San Diego, CA 92121, USA
| | - Ivan Rusyn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
46
|
Wiriyakulsit N, Keawsomnuk P, Thongin S, Ketsawatsomkron P, Muta K. A model of hepatic steatosis with declined viability and function in a liver-organ-on-a-chip. Sci Rep 2023; 13:17019. [PMID: 37813918 PMCID: PMC10562420 DOI: 10.1038/s41598-023-44198-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/04/2023] [Indexed: 10/11/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) begins with benign steatosis caused by ectopic storage of triacylglycerols in the liver. Persistent steatosis, in combination with other genetic and environmental factors, leads to nonalcoholic steatohepatitis (NASH) characterized by functional impairment, inflammation, and fibrosis. However, it remains unclear how persistent steatosis directly contributes to the progression of NAFLD, which may represent a therapeutic target. The organ-on-a-chip (OOC) has emerged as a new culture platform to recapitulate human pathological conditions under which drug candidates can be screened. Here, we developed a simple OOC steatosis model using the Mimetas OrganoPlate with a human liver cell line, HepG2. Treating the HepG2 OOCs with fatty acid overload induced steatosis within 24 h. Moreover, persistent steatosis for 6 days impaired OOC viability and hepatic function, as measured by a WST-8 assay and albumin production, respectively. Lastly, the HepG2 OOCs were exposed to drugs being tested in clinical trials for NAFLD/NASH during the 6-day period. Pioglitazone improved the OOC viability while elafibranor reduced the steatosis in association with reduced viability and albumin production. In conclusion, we show that the HepG2 steatosis OOC model is a useful tool on which the efficacy and toxicity of various therapeutic candidates can be tested.
Collapse
Affiliation(s)
- Natsupa Wiriyakulsit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Ploychanok Keawsomnuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Saowarose Thongin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Pimonrat Ketsawatsomkron
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Kenjiro Muta
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand.
| |
Collapse
|
47
|
Schmid R, Kaiser J, Willbold R, Walther N, Wittig R, Lindén M. Towards a simple in vitro surface chemistry pre-screening method for nanoparticles to be used for drug delivery to solid tumours. Biomater Sci 2023; 11:6287-6298. [PMID: 37551433 DOI: 10.1039/d3bm00966a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
An efficient nanoparticulate drug carrier intended for chemotherapy based on intravenous administration must exhibit a long enough blood circulation time, a good penetrability into the tumour volume, as well as an efficient uptake by cancer cells. Limiting factors for the therapeutic outcome in vivo are recognition of the nanoparticles as foreign objects, which triggers nanoparticle uptake by defence organs rich in macrophages, e.g. liver and spleen, on the time-scale of accumulation and uptake in/by the tumour. However, the development of nanomedicine towards efficient nanoparticle-based delivery to solid tumours is hampered by the lack of simple, reproducible, cheap, and predictive means for early identification of promising nanoparticle formulations. The surface chemistry of nanoparticles is known to be the most important determinant for the biological fate of nanoparticles, as it influences the extent of serum protein adsorption, and also the relative composition of the protein corona. Here we preliminarily evaluate an extremely simple screening method for nanoparticle surface chemistry pre-optimization based on nanoparticle uptake in vitro by PC-3 cancer cells and THP-1 macrophages. Only when both selectivity for the cancer cells as well as the extent of nanoparticle uptake are taken into consideration do the in vitro results mirror literature results obtained for small animal models. Furthermore, although not investigated here, the screening method does also lend itself to the study of actively targeted nanoparticles.
Collapse
Affiliation(s)
- Roman Schmid
- Inorganic Chemistry II, Albert-Einstein-Allee 11, Ulm University, 89081 Ulm, Germany.
| | - Juliane Kaiser
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Ramona Willbold
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Nomusa Walther
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Rainer Wittig
- Institute for Laser Technologies in Medicine & Metrology (ILM) at Ulm University, Helmholtzstrasse 12, 89081 Ulm, Germany.
| | - Mika Lindén
- Inorganic Chemistry II, Albert-Einstein-Allee 11, Ulm University, 89081 Ulm, Germany.
| |
Collapse
|
48
|
Freires IA, Morelo DFC, Soares LFF, Costa IS, de Araújo LP, Breseghello I, Abdalla HB, Lazarini JG, Rosalen PL, Pigossi SC, Franchin M. Progress and promise of alternative animal and non-animal methods in biomedical research. Arch Toxicol 2023; 97:2329-2342. [PMID: 37394624 DOI: 10.1007/s00204-023-03532-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/24/2023] [Indexed: 07/04/2023]
Abstract
Cell culture and invertebrate animal models reflect a significant evolution in scientific research by providing reliable evidence on the physiopathology of diseases, screening for new drugs, and toxicological tests while reducing the need for mammals. In this review, we discuss the progress and promise of alternative animal and non-animal methods in biomedical research, with a special focus on drug toxicity.
Collapse
Affiliation(s)
- Irlan Almeida Freires
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil.
| | - David Fernando Colon Morelo
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | | | | | | | | | - Henrique Ballassini Abdalla
- Laboratory of Neuroimmune Interface of Pain Research, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil
| | - Josy Goldoni Lazarini
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | - Pedro Luiz Rosalen
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
- Graduate Program in Biological Sciences, Federal University of Alfenas, Alfenas, Brazil
| | | | - Marcelo Franchin
- School of Dentistry, Federal University of Alfenas, Alfenas, Brazil
- Bioactivity and Applications Lab, Department of Biological Sciences, Faculty of Science and Engineering, School of Natural Sciences, University of Limerick, Limerick, Ireland
| |
Collapse
|
49
|
Gimondi S, Ferreira H, Reis RL, Neves NM. Microfluidic Devices: A Tool for Nanoparticle Synthesis and Performance Evaluation. ACS NANO 2023; 17:14205-14228. [PMID: 37498731 PMCID: PMC10416572 DOI: 10.1021/acsnano.3c01117] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
The use of nanoparticles (NPs) in nanomedicine holds great promise for the treatment of diseases for which conventional therapies present serious limitations. Additionally, NPs can drastically improve early diagnosis and follow-up of many disorders. However, to harness their full capabilities, they must be precisely designed, produced, and tested in relevant models. Microfluidic systems can simulate dynamic fluid flows, gradients, specific microenvironments, and multiorgan complexes, providing an efficient and cost-effective approach for both NPs synthesis and screening. Microfluidic technologies allow for the synthesis of NPs under controlled conditions, enhancing batch-to-batch reproducibility. Moreover, due to the versatility of microfluidic devices, it is possible to generate and customize endless platforms for rapid and efficient in vitro and in vivo screening of NPs' performance. Indeed, microfluidic devices show great potential as advanced systems for small organism manipulation and immobilization. In this review, first we summarize the major microfluidic platforms that allow for controlled NPs synthesis. Next, we will discuss the most innovative microfluidic platforms that enable mimicking in vitro environments as well as give insights into organism-on-a-chip and their promising application for NPs screening. We conclude this review with a critical assessment of the current challenges and possible future directions of microfluidic systems in NPs synthesis and screening to impact the field of nanomedicine.
Collapse
Affiliation(s)
- Sara Gimondi
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| | - Helena Ferreira
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| | - Nuno M. Neves
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters
of the European Institute of Excellence on Tissue Engineering and
Regenerative Medicine, AvePark, Parque
de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s−PT
Government Associate Laboratory, 4805-017 Braga, Guimarães, Portugal
| |
Collapse
|
50
|
McDuffie D, Alver CG, Suthar B, Helm M, Oliver D, Burgess RA, Barr D, Thomas E, Agarwal A. Acrylic-based culture plate format perfusion device to establish liver endothelial-epithelial interface. LAB ON A CHIP 2023; 23:3106-3119. [PMID: 37313651 PMCID: PMC10351567 DOI: 10.1039/d3lc00382e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microphysiological Systems (MPSs) or organs-on-chips, are microfluidic devices used to model human physiology in vitro. Polydimethylsiloxane (PDMS) is the most widely used material for organs-on-chips due to its established fabrication methods and biocompatibility properties. However, non-specific binding of small molecules limits PDMS for drug screening applications. Here, we designed a novel acrylic-based MPS to capture the physiological architecture that is observed universally in tissues across the body: the endothelial-epithelial interface (EEI). To reconstruct the EEI biology, we designed a membrane-based chip that features endothelial cells on the underside of the membrane exposed to mechanical shear from the path of media flow, and epithelial cells on the opposite side of the membrane protected from flow, as they are in vivo. We used a liver model with a hepatic progenitor cell line and human umbilical vein endothelial cells to assess the biological efficacy of the MPS. We computationally modeled the physics that govern the function of perfusion through the MPS. Empirically, efficacy was measured by comparing differentiation of the hepatic progenitor cells between the MPS and 2D culture conditions. We demonstrated that the MPS significantly improved hepatocyte differentiation, increased extracellular protein transport, and raised hepatocyte sensitivity to drug treatment. Our results strongly suggest that physiological perfusion has a profound effect on proper hepatocyte function, and the modular chip design motivates opportunities for future study of multi-organ interactions.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Bhumi Suthar
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Madeline Helm
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - David Oliver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - R Alan Burgess
- Department of Pathology & Laboratory Medicine, Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - David Barr
- Department of Pathology & Laboratory Medicine, Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Emmanuel Thomas
- Department of Pathology & Laboratory Medicine, Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, USA
| |
Collapse
|