1
|
Hu W, Wang Y, Han J, Zhang W, Chen J, Li X, Wang L. Microfluidic organ-on-a-chip models for the gut-liver axis: from structural mimicry to functional insights. Biomater Sci 2025; 13:1624-1656. [PMID: 40019226 DOI: 10.1039/d4bm01273a] [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: 03/01/2025]
Abstract
The gut-liver axis plays a crucial role in maintaining metabolic balance and overall human health. It orchestrates various processes, such as blood flow, nutrient transfer, metabolite processing, and immune cell communication between the two organs. Traditional methods, such as animal models and two-dimensional (2D) cell cultures, are insufficient in fully replicating the intricate functions of the gut-liver axis. The emergence of microfluidic technology has revolutionized this field, facilitating the development of organ-on-a-chip (OOC) systems. These systems are capable of mimicking the complex structures and dynamic environments of the gut and liver in vitro and incorporating sensors for real-time monitoring. In this article, we review the latest progress in gut-on-a-chip (GOC) and liver-on-a-chip (LOC) systems, as well as the integrated gut-liver-on-a-chip (GLOC) models. Our focus lies in the simulation of physiological parameters, three-dimensional (3D) structural mimicry, microbiome integration, and multicellular co-culture. All these aspects are essential for constructing accurate in vitro models of the gut and liver. Furthermore, we explore the current applications of OOC technology in the study of the gut and liver, including its use in disease modeling, toxicity testing, and drug screening. Finally, we discuss the challenges that remain and outline potential future directions for advancing GOC and LOC development in vitro.
Collapse
Affiliation(s)
- Wanlin Hu
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
2
|
Lin R, Huang Z, Liu Y, Zhou Y. Analysis of Personalized Cardiovascular Drug Therapy: From Monitoring Technologies to Data Integration and Future Perspectives. BIOSENSORS 2025; 15:191. [PMID: 40136988 PMCID: PMC11940481 DOI: 10.3390/bios15030191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/09/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025]
Abstract
Cardiovascular diseases have long been a major challenge to human health, and the treatment differences caused by individual variability remain unresolved. In recent years, personalized cardiovascular drug therapy has attracted widespread attention. This paper reviews the strategies for achieving personalized cardiovascular drug therapy through traditional dynamic monitoring and multidimensional data integration and analysis. It focuses on key technologies for dynamic monitoring, dynamic monitoring based on individual differences, and multidimensional data integration and analysis. By systematically reviewing the relevant literature, the main challenges in current research and the proposed potential directions for future studies were summarized.
Collapse
Affiliation(s)
| | | | | | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa 999078, Macau
| |
Collapse
|
3
|
Fukunaga I, Takebe T. In vitro liver models for toxicological research. Drug Metab Pharmacokinet 2025; 62:101478. [PMID: 40203632 DOI: 10.1016/j.dmpk.2025.101478] [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: 12/19/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Drug-induced liver injury (DILI) presents a major challenge not only in new drug development but also in post-marketing withdrawals and the safety of food, cosmetics, and chemicals. Experimental model organisms such as the rodents have been widely used for preclinical toxicological testing. However, the tension exists associated with the ethical and sustainable use of animals in part because animals do not necessarily inform the human-specific ADME (adsorption, dynamics, metabolism and elimination) profiling. To establish alternative models in humans, in vitro hepatic tissue models have been proposed, ranging from primary hepatocytes, immortal hepatocytes, to the development of new cell resources such as stem cell-derived hepatocytes. Given the evolving number of novel alternative methods, understanding possible combinations of cell sources and culture methods will be crucial to develop the context-of-use assays. This review primarily focuses on 3D liver organoid models for conducting. We will review the relevant cell sources, bioengineering methods, selection of training compounds, and biomarkers towards the rationale design of in vitro toxicology testing.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
4
|
Ebrahimi A, Ghorbanpoor H, Apaydın E, Demir Cevizlidere B, Özel C, Tüfekçioğlu E, Koç Y, Topal AE, Tomsuk Ö, Güleç K, Abdullayeva N, Kaya M, Ghorbani A, Şengel T, Benzait Z, Uysal O, Eker Sarıboyacı A, Doğan Güzel F, Singh H, Hassan S, Ankara H, Pat S, Atalay E, Avci H. Convenient rapid prototyping microphysiological niche for mimicking liver native basement membrane: Liver sinusoid on a chip. Colloids Surf B Biointerfaces 2024; 245:114292. [PMID: 39383580 DOI: 10.1016/j.colsurfb.2024.114292] [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: 07/08/2024] [Revised: 09/24/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Liver is responsible for the metabolization processes of up to 90 % of compounds and toxins in the body. Therefore liver-on-a-chip systems, as an in vitro promising cell culture platform, have great importance for fundamental science and drug development. In most of the liver-on-a-chip studies, seeding cells on both sides of a porous membrane, which represents the basement membrane, fail to resemble the native characteristics of biochemical, biophysical, and mechanical properties. In this study, polycarbonate (PC) and polyethylene terephthalate (PET) membranes were coated with gelatin to address this issue by accurately mimicking the native basement membrane present in the space of Disse. Various coating methods were used, including doctor blade, gel micro-injection, electrospinning, and spin coating. Spin coating was demonstrated to be the most effective technique owing to the ability to produce thin gel thickness with desirable surface roughness for cell interactions on both sides of the membrane. HepG2 and EA.HY926 cells were seeded on the upper and bottom sides of the gelatin-coated PET membrane and cultured on-chip for 7 days. Cell viability increased from 90 % to 95 %, while apoptotic index decreased. Albumin secretion notably rose between days 1-7 and 4-7, while GST-α secretion decreased from day 1 to day 7. In conclusion, the optimized spin coating process reported here can effectively modify the membranes to better mimic the native basement membrane niche characteristics.
Collapse
Affiliation(s)
- Aliakbar Ebrahimi
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Hamed Ghorbanpoor
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biomedical Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Elif Apaydın
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Bahar Demir Cevizlidere
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ceren Özel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Emre Tüfekçioğlu
- Department of Industrial Design/Department of Industrial Design, Faculty of Architecture and Design, Eskisehir Technical University, Eskisehir, Türkiye
| | - Yücel Koç
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ahmet Emin Topal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Biochemistry, School of Pharmacy, Bahçeşehir University, Istanbul, Türkiye
| | - Özlem Tomsuk
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Mechanical Engineering, Middle East Technical University, Ankara 06800, Türkiye
| | - Kadri Güleç
- Department of Analytical Chemistry, Institute of Health Sciences, Anadolu University, Eskisehir, Türkiye
| | - Nuran Abdullayeva
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Murat Kaya
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Aynaz Ghorbani
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Tayfun Şengel
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Central Research Laboratory Research and Application Center (ARUM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Zineb Benzait
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Ayla Eker Sarıboyacı
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye
| | - Fatma Doğan Güzel
- Department of Biomedical Engineering, Ankara Yildirim Beyazit University, Ankara, Türkiye
| | - Hemant Singh
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Shabir Hassan
- Department of Biological Sciences, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Center for Biotechnology, Khalifa University, Main Campus, Abu Dhabi, United Arab Emirates; Functional Biomaterials Group, Khalifa University, San Campus, Abu Dhabi, United Arab Emirates
| | - Hüseyin Ankara
- Mining Engineering Department, Engineering-Architecture Faculty, Eskisehir Osmangazi University, Meşelik Campus, Eskisehir 26480, Türkiye
| | - Suat Pat
- Eskisehir Osmangazi University, Faculty of Science, Department of Physics, Eskisehir TR-26040, Türkiye
| | - Eray Atalay
- Department of Ophthalmology, Faculty of Medicine, Eskisehir Osmangazi University, Eskisehir 26040, Türkiye
| | - Huseyin Avci
- Cellular Therapy and Stem Cell Production Application and Research Center (ESTEM), Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Türkiye; Department of Metallurgical and Materials Engineering, Eskisehir Osmangazi University, Eskisehir, Türkiye; Translational Medicine Research and Clinical Center (TATUM), Eskisehir Osmangazi University, Eskisehir, Türkiye.
| |
Collapse
|
5
|
Deng Q, Yang Y, Liu Y, Zou M, Huang G, Yang S, Li L, Qu Y, Luo Y, Zhang X. Assessing immune hepatotoxicity of troglitazone with a versatile liver-immune-microphysiological-system. Front Pharmacol 2024; 15:1335836. [PMID: 38873410 PMCID: PMC11169855 DOI: 10.3389/fphar.2024.1335836] [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: 11/09/2023] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Drug-induced liver injury is a prevalent adverse event associated with pharmaceutical agents. More significantly, there are certain drugs that present severe hepatotoxicity only during the clinical phase, consequently leading to the termination of drug development during clinical trials or the withdrawal from the market after approval. The establishment of an evaluation model that can sensitively manifest such hepatotoxicity has always been a challenging aspect in drug development. In this study, we build a liver-immune-microphysiological-system (LIMPS) to fully demonstrate the liver injury triggered by troglitazone (TGZ), a drug that was withdrawn from the market due to hepatotoxicity. Leveraging the capabilities of organ-on-chip technology allows for the dynamic modulation of cellular immune milieu, as well as the synergistic effects between drugs, hepatocytes and multiple immune cells. Through the LIMPS, we discovered that 1) TGZ can promote neutrophils to adhered hepatocytes, 2) the presence of TGZ enhances the crosstalk between macrophages and neutrophils, 3) the induction of damage in hepatocytes by TGZ at clinically relevant blood concentrations not observed in other in vitro experiments, 4) no hepatotoxicity was observed in LIMPS when exposed to rosiglitazone and pioglitazone, structurally similar analogs of TGZ, even at the higher multiples of blood drug concentration levels. As an immune-mediated liver toxicity assessment method, LIMPS is simple to operate and can be used to test multiple drug candidates to detect whether they will cause severe liver toxicity in clinical settings as early as possible.
Collapse
Affiliation(s)
- Quanfeng Deng
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Youlong Yang
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Yuangui Liu
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Mengting Zou
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Guiyuan Huang
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Shiqi Yang
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Lingyu Li
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Yueyang Qu
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning Province, China
| | - Xiuli Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Disease and College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu Province, China
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, Hunan Province, China
| |
Collapse
|
6
|
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
|
7
|
Moon HR, Surianarayanan N, Singh T, Han B. Microphysiological systems as reliable drug discovery and evaluation tools: Evolution from innovation to maturity. BIOMICROFLUIDICS 2023; 17:061504. [PMID: 38162229 PMCID: PMC10756708 DOI: 10.1063/5.0179444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Microphysiological systems (MPSs), also known as organ-on-chip or disease-on-chip, have recently emerged to reconstitute the in vivo cellular microenvironment of various organs and diseases on in vitro platforms. These microfluidics-based platforms are developed to provide reliable drug discovery and regulatory evaluation testbeds. Despite recent emergences and advances of various MPS platforms, their adoption of drug discovery and evaluation processes still lags. This delay is mainly due to a lack of rigorous standards with reproducibility and reliability, and practical difficulties to be adopted in pharmaceutical research and industry settings. This review discusses the current and potential use of MPS platforms in drug discovery processes while considering the context of several key steps during drug discovery processes, including target identification and validation, preclinical evaluation, and clinical trials. Opportunities and challenges are also discussed for the broader dissemination and adoption of MPSs in various drug discovery and regulatory evaluation steps. Addressing these challenges will transform long and expensive drug discovery and evaluation processes into more efficient discovery, screening, and approval of innovative drugs.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | | | - Tarun Singh
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Bumsoo Han
- Author to whom correspondence should be addressed:. Tel: +1-765-494-5626
| |
Collapse
|
8
|
Zhang X, Luo Y, Wang Q. Editorial: Organ mimicking technologies and their applications in drug discovery. Front Bioeng Biotechnol 2023; 11:1341153. [PMID: 38107621 PMCID: PMC10722410 DOI: 10.3389/fbioe.2023.1341153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 12/19/2023] Open
Affiliation(s)
- Xiuli Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Engineering, School of Chemical Engineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| |
Collapse
|
9
|
Li C, Holman JB, Shi Z, Qiu B, Ding W. On-chip modeling of tumor evolution: Advances, challenges and opportunities. Mater Today Bio 2023; 21:100724. [PMID: 37483380 PMCID: PMC10359640 DOI: 10.1016/j.mtbio.2023.100724] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/16/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
Tumor evolution is the accumulation of various tumor cell behaviors from tumorigenesis to tumor metastasis and is regulated by the tumor microenvironment (TME). However, the mechanism of solid tumor progression has not been completely elucidated, and thus, the development of tumor therapy is still limited. Recently, Tumor chips constructed by culturing tumor cells and stromal cells on microfluidic chips have demonstrated great potential in modeling solid tumors and visualizing tumor cell behaviors to exploit tumor progression. Herein, we review the methods of developing engineered solid tumors on microfluidic chips in terms of tumor types, cell resources and patterns, the extracellular matrix and the components of the TME, and summarize the recent advances of microfluidic chips in demonstrating tumor cell behaviors, including proliferation, epithelial-to-mesenchymal transition, migration, intravasation, extravasation and immune escape of tumor cells. We also outline the combination of tumor organoids and microfluidic chips to elaborate tumor organoid-on-a-chip platforms, as well as the practical limitations that must be overcome.
Collapse
Affiliation(s)
- Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Joseph Benjamin Holman
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Bensheng Qiu
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui, 230027, China
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui, 230027, China
| | - Weiping Ding
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| |
Collapse
|
10
|
Tabatabaei Rezaei N, Kumar H, Liu H, Lee SS, Park SS, Kim K. Recent Advances in Organ-on-Chips Integrated with Bioprinting Technologies for Drug Screening. Adv Healthc Mater 2023; 12:e2203172. [PMID: 36971091 PMCID: PMC11469032 DOI: 10.1002/adhm.202203172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Currently, the demand for more reliable drug screening devices has made scientists and researchers develop novel potential approaches to offer an alternative to animal studies. Organ-on-chips are newly emerged platforms for drug screening and disease metabolism investigation. These microfluidic devices attempt to recapitulate the physiological and biological properties of different organs and tissues using human-derived cells. Recently, the synergistic combination of additive manufacturing and microfluidics has shown a promising impact on improving a wide array of biological models. In this review, different methods are classified using bioprinting to achieve the relevant biomimetic models in organ-on-chips, boosting the efficiency of these devices to produce more reliable data for drug investigations. In addition to the tissue models, the influence of additive manufacturing on microfluidic chip fabrication is discussed, and their biomedical applications are reviewed.
Collapse
Affiliation(s)
- Nima Tabatabaei Rezaei
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Hitendra Kumar
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
- Department of Pathology and Laboratory MedicineCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Hongqun Liu
- Liver UnitCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Samuel S. Lee
- Liver UnitCumming School of MedicineUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Simon S. Park
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| | - Keekyoung Kim
- Department of Mechanical and Manufacturing EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
- Department of Biomedical EngineeringUniversity of CalgaryCalgaryAlbertaT2N 1N4Canada
| |
Collapse
|
11
|
Li S, Li C, Khan MI, Liu J, Shi Z, Gao D, Qiu B, Ding W. Microneedle array facilitates hepatic sinusoid construction in a large-scale liver-acinus-chip microsystem. MICROSYSTEMS & NANOENGINEERING 2023; 9:75. [PMID: 37303831 PMCID: PMC10247758 DOI: 10.1038/s41378-023-00544-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 06/13/2023]
Abstract
Hepatic sinusoids play a key role in maintaining high activities of liver cells in the hepatic acinus. However, the construction of hepatic sinusoids has always been a challenge for liver chips, especially for large-scale liver microsystems. Herein, we report an approach for the construction of hepatic sinusoids. In this approach, hepatic sinusoids are formed by demolding a self-developed microneedle array from a photocurable cell-loaded matrix in a large-scale liver-acinus-chip microsystem with a designed dual blood supply. Primary sinusoids formed by demolded microneedles and spontaneously self-organized secondary sinusoids can be clearly observed. Benefiting from significantly enhanced interstitial flows by formed hepatic sinusoids, cell viability is witnessed to be considerably high, liver microstructure formation occurs, and hepatocyte metabolism is enhanced. In addition, this study preliminarily demonstrates the effects of the resulting oxygen and glucose gradients on hepatocyte functions and the application of the chip in drug testing. This work paves the way for the biofabrication of fully functionalized large-scale liver bioreactors.
Collapse
Affiliation(s)
- Shibo Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| | - Chengpan Li
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Muhammad Imran Khan
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Jing Liu
- School of Biology, Food and Environment, Hefei University, Hefei, Anhui 230601 China
| | - Zhengdi Shi
- Department of Electronic Engineering and Information Science, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Dayong Gao
- Department of Mechanical Engineering, University of Washington, Seattle, WA 98195 USA
| | - Bensheng Qiu
- Center for Biomedical Imaging, University of Science and Technology of China, Hefei, Anhui 230027 China
| | - Weiping Ding
- Department of Oncology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001 China
| |
Collapse
|
12
|
McDuffie D, Barr D, Helm M, Baumert T, Agarwal A, Thomas E. Physiomimetic In Vitro Human Models for Viral Infection in the Liver. Semin Liver Dis 2023; 43:31-49. [PMID: 36402129 PMCID: PMC10005888 DOI: 10.1055/a-1981-5944] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Viral hepatitis is a leading cause of liver morbidity and mortality globally. The mechanisms underlying acute infection and clearance, versus the development of chronic infection, are poorly understood. In vitro models of viral hepatitis circumvent the high costs and ethical considerations of animal models, which also translate poorly to studying the human-specific hepatitis viruses. However, significant challenges are associated with modeling long-term infection in vitro. Differentiated hepatocytes are best able to sustain chronic viral hepatitis infection, but standard two-dimensional models are limited because they fail to mimic the architecture and cellular microenvironment of the liver, and cannot maintain a differentiated hepatocyte phenotype over extended periods. Alternatively, physiomimetic models facilitate important interactions between hepatocytes and their microenvironment by incorporating liver-specific environmental factors such as three-dimensional ECM interactions and co-culture with non-parenchymal cells. These physiologically relevant interactions help maintain a functional hepatocyte phenotype that is critical for sustaining viral hepatitis infection. In this review, we provide an overview of distinct, novel, and innovative in vitro liver models and discuss their functionality and relevance in modeling viral hepatitis. These platforms may provide novel insight into mechanisms that regulate viral clearance versus progression to chronic infections that can drive subsequent liver disease.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Madeline Helm
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Thomas Baumert
- Inserm Research Institute for Viral and Liver Diseases, University of Strasbourg, Strasbourg, France
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
13
|
Advances in cell coculture membranes recapitulating in vivo microenvironments. Trends Biotechnol 2023; 41:214-227. [PMID: 36030108 DOI: 10.1016/j.tibtech.2022.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/05/2022] [Accepted: 07/25/2022] [Indexed: 01/24/2023]
Abstract
Porous membranes play a critical role in in vitro heterogeneous cell coculture systems because they recapitulate the in vivo microenvironment to mediate physical and biochemical crosstalk between cells. While the conventionally available Transwell® system has been widely used for heterogeneous cell coculture, there are drawbacks to precise control over cell-cell interactions and separation for implantation. The size and numbers of the pores and the thickness of the porous membranes are crucial in determining the efficiency of paracrine signaling and direct junctions between cocultured cells, and significantly impact on the performance of heterogeneous cell cultures. These opportunities and challenges have motivated the design of advanced coculture platforms through improvement of the structural and functional properties of porous membranes.
Collapse
|
14
|
Qiu L, Kong B, Kong T, Wang H. Recent advances in liver-on-chips: Design, fabrication, and applications. SMART MEDICINE 2023; 2:e20220010. [PMID: 39188562 PMCID: PMC11235950 DOI: 10.1002/smmd.20220010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/20/2022] [Indexed: 08/28/2024]
Abstract
The liver is a multifunctional organ and the metabolic center of the human body. Most drugs and toxins are metabolized in the liver, resulting in varying degrees of hepatotoxicity. The damage of liver will seriously affect human health, so it is very important to study the prevention and treatment of liver diseases. At present, there are many research studies in this field. However, most of them are based on animal models, which are limited by the time-consuming processes and species difference between human and animals. In recent years, liver-on-chips have emerged and developed rapidly and are expected to replace animal models. Liver-on-chips refer to the use of a small number of liver cells on the chips to simulate the liver microenvironment and ultrastructure in vivo. They hold extensive applications in multiple fields by reproducing the unique physiological functions of the liver in vitro. In this review, we first introduced the physiology and pathology of liver and then described the cell system of liver-on-chips, the chip-based liver models, and the applications of liver-on-chips in liver transplantation, drug screening, and metabolic evaluation. Finally, we discussed the currently encountered challenges and future trends in liver-on-chips.
Collapse
Affiliation(s)
- Linjie Qiu
- The Eighth Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
- School of MedicineSun Yat‐Sen UniversityShenzhenChina
| | - Bin Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingDepartment of Biomedical EngineeringSchool of MedicineShenzhen UniversityShenzhenChina
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound ImagingDepartment of Biomedical EngineeringSchool of MedicineShenzhen UniversityShenzhenChina
| | - Huan Wang
- The Eighth Affiliated HospitalSun Yat‐Sen UniversityShenzhenChina
| |
Collapse
|
15
|
Kaur I, Vasudevan A, Rawal P, Tripathi DM, Ramakrishna S, Kaur S, Sarin SK. Primary Hepatocyte Isolation and Cultures: Technical Aspects, Challenges and Advancements. Bioengineering (Basel) 2023; 10:131. [PMID: 36829625 PMCID: PMC9952008 DOI: 10.3390/bioengineering10020131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Hepatocytes are differentiated cells that account for 80% of the hepatic volume and perform all major functions of the liver. In vivo, after an acute insult, adult hepatocytes retain their ability to proliferate and participate in liver regeneration. However, in vitro, prolonged culture and proliferation of viable and functional primary hepatocytes have remained the major and the most challenging goal of hepatocyte-based cell therapies and liver tissue engineering. The first functional cultures of rat primary hepatocytes between two layers of collagen gel, also termed as the "sandwich cultures", were reported in 1989. Since this study, several technical developments including choice of hydrogels, type of microenvironment, growth factors and culture conditions, mono or co-cultures of hepatocytes along with other supporting cell types have evolved for both rat and human primary hepatocytes in recent years. All these improvements have led to a substantial improvement in the number, life-span and hepatic functions of these cells in vitro for several downstream applications. In the current review, we highlight the details, limitations and prospects of different technical strategies being used in primary hepatocyte cultures. We discuss the use of newer biomaterials as scaffolds for efficient culture of primary hepatocytes. We also describe the derivation of mature hepatocytes from other cellular sources such as induced pluripotent stem cells, bone marrow stem cells and 3D liver organoids. Finally, we also explain the use of perfusion-based bioreactor systems and bioengineering strategies to support the long-term function of hepatocytes in 3D conditions.
Collapse
Affiliation(s)
- Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Preety Rawal
- School of Biotechnology, Gautam Buddha University, Greater Noida 201312, India
| | - Dinesh M. Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Shiv K. Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
16
|
Advances in application and innovation of microfluidic platforms for pharmaceutical analysis. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
17
|
Zhao W, Yao Y, Zhang T, Lu H, Zhang X, Zhao L, Chen X, Zhu J, Sui G, Zhao W. Primary exploration of host-microorganism interaction and enteritis treatment with an embedded membrane microfluidic chip of the human intestinal-vascular microsystem. Front Bioeng Biotechnol 2022; 10:1035647. [PMID: 36561041 PMCID: PMC9763581 DOI: 10.3389/fbioe.2022.1035647] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal flora plays a crucial role in the host's intestinal health. Imbalances in the intestinal flora, when accompanied by inflammation, affect the host's intestinal barrier function. Understanding it requires studying how living cells and tissues work in the context of living organs, but it is difficult to form the three-dimensional microstructure intestinal-vascular system by monolayer cell or co-culture cell models, and animal models are costly and slow. The use of microfluidic-based organ chips is a fast, simple, and high-throughput method that not only solves the affinity problem of animal models but the lack of microstructure problem of monolayer cells. In this study, we designed an embedded membrane chip to generate an in vitro gut-on-a-chip model. Human umbilical vein endothelial cells and Caco-2 were cultured in the upper and lower layers of the culture chambers in the microfluidic chip, respectively. The human peripheral blood mononuclear cells were infused into the capillary side at a constant rate using an external pump to simulate the in vitro immune system and the shear stress of blood in vivo. The model exhibited intestine morphology and function after only 5 days of culture, which is significantly less than the 21 days required for static culture in the Transwell® chamber. Furthermore, it was observed that drug-resistant bacteria triggered barrier function impairment and inflammation, resulting in enteritis, whereas probiotics (Lactobacillus rhamnosus GG) improved only partially. The use of Amikacin for enteritis is effective, whereas other antibiotic therapies do not work, which are consistent with clinical test results. This model may be used to explore intestinal ecology, host and intestinal flora interactions, and medication assessment.
Collapse
Affiliation(s)
- Wei Zhao
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Yuhan Yao
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Tong Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Huijun Lu
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Xinlian Zhang
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Linlin Zhao
- Shanghai Changhai Hospital Department of Gastroenterology, Shanghai, China
| | - Xi Chen
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Jinhui Zhu
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Guodong Sui
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| | - Wang Zhao
- Shanghai Key Laboratory of Atmospheric Particle Pollution Prevention (LAP3), Department of Environmental Science and Engineering, Fudan University, Shanghai, China
| |
Collapse
|
18
|
Shen P, Jia Y, Shi S, Sun J, Han X. Analytical and biomedical applications of microfluidics in traditional Chinese medicine research. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
19
|
Etxeberria L, Messelmani T, Badiola JH, Llobera A, Fernandez L, Vilas-Vilela JL, Leclerc E, Legallais C, Jellali R, Zaldua AM. Validation of HepG2/C3A Cell Cultures in Cyclic Olefin Copolymer Based Microfluidic Bioreactors. Polymers (Basel) 2022; 14:polym14214478. [PMID: 36365472 PMCID: PMC9655789 DOI: 10.3390/polym14214478] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Organ-on-chip (OoC) technology is one of the most promising in vitro tools to replace the traditional animal experiment-based paradigms of risk assessment. However, the use of OoC in drug discovery and toxicity studies remain still limited by the low capacity for high-throughput production and the incompatibility with standard laboratory equipment. Moreover, polydimethylsiloxanes, the material of choice for OoC, has several drawbacks, particularly the high absorption of drugs and chemicals. In this work, we report the development of a microfluidic device, using a process adapted for mass production, to culture liver cell line in dynamic conditions. The device, made of cyclic olefin copolymers, was manufactured by injection moulding and integrates Luer lock connectors compatible with standard medical and laboratory instruments. Then, the COC device was used for culturing HepG2/C3a cells. The functionality and behaviour of cultures were assessed by albumin secretion, cell proliferation, viability and actin cytoskeleton development. The cells in COC device proliferated well and remained functional for 9 days of culture. Furthermore, HepG2/C3a cells in the COC biochips showed similar behaviour to cells in PDMS biochips. The present study provides a proof-of-concept for the use of COC biochip in liver cells culture and illustrate their potential to develop OoC.
Collapse
Affiliation(s)
- Leire Etxeberria
- Leartiker S. Coop., Xemein Etorbidea 12, 48270 Markina-Xemein, Spain
- microLIQUID S.L, Goiru 9, 20500 Arrasate-Mondragon, Spain
- Macromolecular Chemistry Research Group (labquimac), Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Correspondence: (L.E.); (R.J.)
| | - Taha Messelmani
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
| | - Jon Haitz Badiola
- Leartiker S. Coop., Xemein Etorbidea 12, 48270 Markina-Xemein, Spain
| | - Andreu Llobera
- microLIQUID S.L, Goiru 9, 20500 Arrasate-Mondragon, Spain
| | - Luis Fernandez
- microLIQUID S.L, Goiru 9, 20500 Arrasate-Mondragon, Spain
| | - José Luis Vilas-Vilela
- Macromolecular Chemistry Research Group (labquimac), Department of Physical Chemistry, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- BC Materials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, 48940 Leioa, Spain
| | - Eric Leclerc
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Tokyo 153-8505, Japan
| | - Cécile Legallais
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
| | - Rachid Jellali
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
- Correspondence: (L.E.); (R.J.)
| | - Ane Miren Zaldua
- Leartiker S. Coop., Xemein Etorbidea 12, 48270 Markina-Xemein, Spain
| |
Collapse
|
20
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [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: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
21
|
Messelmani T, Le Goff A, Souguir Z, Maes V, Roudaut M, Vandenhaute E, Maubon N, Legallais C, Leclerc E, Jellali R. Development of Liver-on-Chip Integrating a Hydroscaffold Mimicking the Liver’s Extracellular Matrix. Bioengineering (Basel) 2022; 9:bioengineering9090443. [PMID: 36134989 PMCID: PMC9495334 DOI: 10.3390/bioengineering9090443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/23/2022] [Accepted: 08/31/2022] [Indexed: 12/12/2022] Open
Abstract
The 3Rs guidelines recommend replacing animal testing with alternative models. One of the solutions proposed is organ-on-chip technology in which liver-on-chip is one of the most promising alternatives for drug screening and toxicological assays. The main challenge is to achieve the relevant in vivo-like functionalities of the liver tissue in an optimized cellular microenvironment. Here, we investigated the development of hepatic cells under dynamic conditions inside a 3D hydroscaffold embedded in a microfluidic device. The hydroscaffold is made of hyaluronic acid and composed of liver extracellular matrix components (galactosamine, collagen I/IV) with RGDS (Arg-Gly-Asp-Ser) sites for cell adhesion. The HepG2/C3A cell line was cultured under a flow rate of 10 µL/min for 21 days. After seeding, the cells formed aggregates and proliferated, forming 3D spheroids. The cell viability, functionality, and spheroid integrity were investigated and compared to static cultures. The results showed a 3D aggregate organization of the cells up to large spheroid formations, high viability and albumin production, and an enhancement of HepG2 cell functionalities. Overall, these results highlighted the role of the liver-on-chip model coupled with a hydroscaffold in the enhancement of cell functions and its potential for engineering a relevant liver model for drug screening and disease study.
Collapse
Affiliation(s)
- Taha Messelmani
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
| | - Anne Le Goff
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
- Correspondence: (A.L.G.); (R.J.)
| | - Zied Souguir
- HCS Pharma, 250 rue Salvador Allende, Biocentre Fleming Bâtiment A, 59120 Loos, France
| | - Victoria Maes
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
- HCS Pharma, 250 rue Salvador Allende, Biocentre Fleming Bâtiment A, 59120 Loos, France
| | - Méryl Roudaut
- HCS Pharma, 250 rue Salvador Allende, Biocentre Fleming Bâtiment A, 59120 Loos, France
| | - Elodie Vandenhaute
- HCS Pharma, 250 rue Salvador Allende, Biocentre Fleming Bâtiment A, 59120 Loos, France
| | - Nathalie Maubon
- HCS Pharma, 250 rue Salvador Allende, Biocentre Fleming Bâtiment A, 59120 Loos, France
| | - Cécile Legallais
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
| | - Eric Leclerc
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Rachid Jellali
- CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu-CS 60319, Université de Technologie de Compiègne, 60203 Compiègne, France
- Correspondence: (A.L.G.); (R.J.)
| |
Collapse
|
22
|
Kanabekova P, Kadyrova A, Kulsharova G. Microfluidic Organ-on-a-Chip Devices for Liver Disease Modeling In Vitro. MICROMACHINES 2022; 13:428. [PMID: 35334720 PMCID: PMC8950395 DOI: 10.3390/mi13030428] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022]
Abstract
Mortality from liver disease conditions continues to be very high. As liver diseases manifest and progress silently, prompt measures after diagnosis are essential in the treatment of these conditions. Microfluidic organs-on-chip platforms have significant potential for the study of the pathophysiology of liver diseases in vitro. Different liver-on-a-chip microphysiological platforms have been reported to study cell-signaling pathways such as those activating stellate cells within liver diseases. Moreover, the drug efficacy for liver conditions might be evaluated on a cellular metabolic level. Here, we present a comprehensive review of microphysiological platforms used for modelling liver diseases. First, we briefly introduce the concept and importance of organs-on-a-chip in studying liver diseases in vitro, reflecting on existing reviews of healthy liver-on-a-chip platforms. Second, the techniques of cell cultures used in the microfluidic devices, including 2D, 3D, and spheroid cells, are explained. Next, the types of liver diseases (NAFLD, ALD, hepatitis infections, and drug injury) on-chip are explained for a further comprehensive overview of the design and methods of developing liver diseases in vitro. Finally, some challenges in design and existing solutions to them are reviewed.
Collapse
Affiliation(s)
- Perizat Kanabekova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Adina Kadyrova
- Department of Biological Sciences, School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Gulsim Kulsharova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| |
Collapse
|
23
|
From organ-on-chip to body-on-chip: The next generation of microfluidics platforms for in vitro drug efficacy and toxicity testing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:41-91. [PMID: 35094781 DOI: 10.1016/bs.pmbts.2021.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The high failure rate in drug development is often attributed to the lack of accurate pre-clinical models that may lead to false discoveries and inconclusive data when the compounds are eventually tested in clinical phase. With the evolution of cell culture technologies, drug testing systems have widely improved, and today, with the emergence of microfluidics devices, drug screening seems to be at the dawn of an important revolution. An organ-on-chip allows the culture of living cells in continuously perfused microchambers to reproduce physiological functions of a particular tissue or organ. The advantages of such systems are not only their ability to recapitulate the complex biochemical interactions between different human cell types but also to incorporate physical forces, including shear stress and mechanical stretching or compression. To improve this model, and to reproduce the absorption, distribution, metabolism, and elimination process of an exogenous compound, organ-on-chips can even be linked fluidically to mimic physiological interactions between different organs, leading to the development of body-on-chips. Although these technologies are still at a young age and need to address a certain number of limitations, they already demonstrated their relevance to study the effect of drugs or toxins on organs, displaying a similar response to what is observed in vivo. The purpose of this review is to present the evolution from organ-on-chip to body-on-chip, examine their current use for drug testing and discuss their advantages and future challenges they will face in order to become an essential pillar of pharmaceutical research.
Collapse
|
24
|
Lee SY, Kim D, Lee SH, Sung JH. Microtechnology-based in vitro models: Mimicking liver function and pathophysiology. APL Bioeng 2021; 5:041505. [PMID: 34703969 PMCID: PMC8520487 DOI: 10.1063/5.0061896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
The liver plays important roles in drug metabolism and homeostasis. The metabolism and biotransformation can not only affect the efficacy of drugs but also result in hepatotoxicity and drug-induced liver injury. Understanding the complex physiology of the liver and the pathogenetic mechanisms of liver diseases is essential for drug development. Conventional in vitro models have limitations in the ability to predict drug effects, due to the lack of physiological relevance. Recently, the liver-on-a-chip platform has been developed to reproduce the microarchitecture and in vivo environment of the liver. These efforts have improved the physiological relevance of the liver tissue used in the platform and have demonstrated its applicability to drug screening and disease models. In this review, we summarize the recent development of liver-on-a-chip models that closely mimic the in vivo liver environments and liver diseases.
Collapse
Affiliation(s)
- Seung Yeon Lee
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| | - Donghyun Kim
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, South Korea
| | - Seung Hwan Lee
- Department of Bionano Engineering, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 15588, South Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul 04066, South Korea
| |
Collapse
|
25
|
Nguyen N, Thurgood P, Sekar NC, Chen S, Pirogova E, Peter K, Baratchi S, Khoshmanesh K. Microfluidic models of the human circulatory system: versatile platforms for exploring mechanobiology and disease modeling. Biophys Rev 2021; 13:769-786. [PMID: 34777617 DOI: 10.1007/s12551-021-00815-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human circulatory system is a marvelous fluidic system, which is very sensitive to biophysical and biochemical cues. The current animal and cell culture models do not recapitulate the functional properties of the human circulatory system, limiting our ability to fully understand the complex biological processes underlying the dysfunction of this multifaceted system. In this review, we discuss the unique ability of microfluidic systems to recapitulate the biophysical, biochemical, and functional properties of the human circulatory system. We also describe the remarkable capacity of microfluidic technologies for exploring the complex mechanobiology of the cardiovascular system, mechanistic studying of cardiovascular diseases, and screening cardiovascular drugs with the additional benefit of reducing the need for animal models. We also discuss opportunities for further advancement in this exciting field.
Collapse
Affiliation(s)
- Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Sheng Chen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiometabolic Health, The University of Melbourne, Parkville, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | | |
Collapse
|
26
|
Panwar A, Das P, Tan LP. 3D Hepatic Organoid-Based Advancements in LIVER Tissue Engineering. Bioengineering (Basel) 2021; 8:185. [PMID: 34821751 PMCID: PMC8615121 DOI: 10.3390/bioengineering8110185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Liver-associated diseases and tissue engineering approaches based on in vitro culture of functional Primary human hepatocytes (PHH) had been restricted by the rapid de-differentiation in 2D culture conditions which restricted their usability. It was proven that cells growing in 3D format can better mimic the in vivo microenvironment, and thus help in maintaining metabolic activity, phenotypic properties, and longevity of the in vitro cultures. Again, the culture method and type of cell population are also recognized as important parameters for functional maintenance of primary hepatocytes. Hepatic organoids formed by self-assembly of hepatic cells are microtissues, and were able to show long-term in vitro maintenance of hepato-specific characteristics. Thus, hepatic organoids were recognized as an effective tool for screening potential cures and modeling liver diseases effectively. The current review summarizes the importance of 3D hepatic organoid culture over other conventional 2D and 3D culture models and its applicability in Liver tissue engineering.
Collapse
Affiliation(s)
- Amit Panwar
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore;
- Faculty of Biotechnology, Institute of Bio-Sciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road Barabanki, Uttar Pradesh 225003, India
| | - Prativa Das
- The Henry Samueli School of Engineering, University of California, Irvine, CA 92617, USA;
| | - Lay Poh Tan
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore;
- Singapore Centre for 3D Printing (SC3DP), Singapore 639798, Singapore
| |
Collapse
|
27
|
Kulsharova G, Kurmangaliyeva A, Darbayeva E, Rojas-Solórzano L, Toxeitova G. Development of a Hybrid Polymer-Based Microfluidic Platform for Culturing Hepatocytes towards Liver-on-a-Chip Applications. Polymers (Basel) 2021; 13:polym13193215. [PMID: 34641031 PMCID: PMC8513053 DOI: 10.3390/polym13193215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
The drug development process can greatly benefit from liver-on-a-chip platforms aiming to recapitulate the physiology, mechanisms, and functionalities of liver cells in an in vitro environment. The liver is the most important organ in drug metabolism investigation. Here, we report the development of a hybrid cyclic olefin copolymer (COC) and polydimethylsiloxane (PDMS) microfluidic (HCP) platform to culture a Huh7 hepatoma cell line in dynamic conditions towards the development of a liver-on-a-chip system. The microfluidic platform is comprised of a COC bottom layer with a microchannel and PDMS-based flat top layer sandwiched together. The HCP device was applied for culturing Huh7 cells grown on a collagen-coated microchannel. A computational fluid dynamics modeling study was conducted for the HCP device design revealing the presence of air volume fraction in the chamber and methods for optimizing experimental handling of the device. The functionality and metabolic activity of perfusion culture were assessed by the secretion rates of albumin, urea, and cell viability visualization. The HCP device hepatic culture remained functional and intact for 24 h, as assessed by resulting levels of biomarkers similar to published studies on other in vitro and 2D cell models. The present results provide a proof-of-concept demonstration of the hybrid COC–PDMS microfluidic chip for successfully culturing a Huh7 hepatoma cell line, thus paving the path towards developing a liver-on-a-chip platform.
Collapse
Affiliation(s)
- Gulsim Kulsharova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (E.D.); (L.R.-S.); (G.T.)
- Correspondence:
| | - Akbota Kurmangaliyeva
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Elvira Darbayeva
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (E.D.); (L.R.-S.); (G.T.)
| | - Luis Rojas-Solórzano
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (E.D.); (L.R.-S.); (G.T.)
| | - Galiya Toxeitova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (E.D.); (L.R.-S.); (G.T.)
| |
Collapse
|
28
|
Ferrari E, Rasponi M. Liver-Heart on chip models for drug safety. APL Bioeng 2021; 5:031505. [PMID: 34286172 PMCID: PMC8282347 DOI: 10.1063/5.0048986] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022] Open
Abstract
Current pre-clinical models to evaluate drug safety during the drug development process (DDP) mainly rely on traditional two-dimensional cell cultures, considered too simplistic and often ineffective, or animal experimentations, which are costly, time-consuming, and not truly representative of human responses. Their clinical translation thus remains limited, eventually causing attrition and leading to high rates of failure during clinical trials. These drawbacks can be overcome by the recently developed Organs-on-Chip (OoC) technology. OoC are sophisticated in vitro systems capable of recapitulating pivotal architecture and functionalities of human organs. OoC are receiving increasing attention from the stakeholders of the DDP, particularly concerning drug screening and safety applications. When a drug is administered in the human body, it is metabolized by the liver and the resulting compound may cause unpredicted toxicity on off-target organs such as the heart. In this sense, several liver and heart models have been widely adopted to assess the toxicity of new or recalled drugs. Recent advances in OoC technology are making available platforms encompassing multiple organs fluidically connected to efficiently assess and predict the systemic effects of compounds. Such Multi-Organs-on-Chip (MOoC) platforms represent a disruptive solution to study drug-related effects, which results particularly useful to predict liver metabolism on off-target organs to ultimately improve drug safety testing in the pre-clinical phases of the DDP. In this review, we focus on recently developed liver and heart on chip systems for drug toxicity testing. In addition, MOoC platforms encompassing connected liver and heart tissues have been further reviewed and discussed.
Collapse
Affiliation(s)
- Erika Ferrari
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milano, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, 20133 Milano, Italy
| |
Collapse
|
29
|
Lu S, Zhang J, Lin S, Zheng D, Shen Y, Qin J, Li Y, Wang S. Recent advances in the development of in vitro liver models for hepatotoxicity testing. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00142-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
30
|
Akarapipad P, Kaarj K, Liang Y, Yoon JY. Environmental Toxicology Assays Using Organ-on-Chip. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2021; 14:155-183. [PMID: 33974806 DOI: 10.1146/annurev-anchem-091620-091335] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Adverse effects of environmental toxicants to human health have traditionally been assayed using in vitro assays. Organ-on-chip (OOC) is a new platform that can bridge the gaps between in vitro assays (or 3D cell culture) and animal tests. Microenvironments, physical and biochemical stimuli, and adequate sensing and biosensing systems can be integrated into OOC devices to better recapitulate the in vivo tissue and organ behavior and metabolism. While OOCs have extensively been studied for drug toxicity screening, their implementation in environmental toxicology assays is minimal and has limitations. In this review, recent attempts of environmental toxicology assays using OOCs, including multiple-organs-on-chip, are summarized and compared with OOC-based drug toxicity screening. Requirements for further improvements are identified and potential solutions are suggested.
Collapse
Affiliation(s)
- Patarajarin Akarapipad
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721, USA;
| | - Kattika Kaarj
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona 85721, USA
| | - Yan Liang
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, USA
| | - Jeong-Yeol Yoon
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85721, USA;
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona 85721, USA
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, USA
| |
Collapse
|
31
|
Kulsharova G, Kurmangaliyeva A. Liver microphysiological platforms for drug metabolism applications. Cell Prolif 2021; 54:e13099. [PMID: 34291515 PMCID: PMC8450120 DOI: 10.1111/cpr.13099] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/21/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Drug development is a costly and lengthy process with low success rates. To improve the efficiency of drug development, there has been an increasing need in developing alternative methods able to eliminate toxic compounds early in the drug development pipeline. Drug metabolism plays a key role in determining the efficacy of a drug and its potential side effects. Since drug metabolism occurs mainly in the liver, liver cell‐based alternative engineering platforms have been growing in the last decade. Microphysiological liver cell‐based systems called liver‐on‐a‐chip platforms can better recapitulate the environment for human liver cells in laboratory settings and have the potential to reduce the number of animal models used in drug development by predicting the response of the liver to a drug in vitro. In this review, we discuss the liver microphysiological platforms from the perspective of drug metabolism studies. We highlight the stand‐alone liver‐on‐a‐chip platforms and multi‐organ systems integrating liver‐on‐a‐chip devices used for drug metabolism mimicry in vitro and review the state‐of‐the‐art platforms reported in the last few years. With the development of more robust and reproducible liver cell‐based microphysiological platforms, the drug development field has the potential of reducing the costs and lengths associated with currently existing drug testing methods.
Collapse
Affiliation(s)
- Gulsim Kulsharova
- School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | | |
Collapse
|
32
|
A Microfluidic Array Device for Single Cell Capture and Intracellular Ca2+ Response Analysis Induced by Dynamic Biochemical Stimulus. Biosci Rep 2021; 41:229251. [PMID: 34269374 PMCID: PMC8319492 DOI: 10.1042/bsr20210719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/29/2021] [Accepted: 07/13/2021] [Indexed: 01/09/2023] Open
Abstract
A microfluidic array was constructed for trapping single cell and loading identical dynamic biochemical stimulation for gain a better understanding of Ca2+ signalling in single cells by applying extracellular dynamic biochemical stimulus. This microfluidic array consists of multiple radially aligned flow channels with equal intersection angles, which was designed by a combination of stagnation point flow and physical barrier. Numerical simulation results and trajectory analysis shown the effectiveness of this single cell trapping device. Fluorescent experiment results demonstrated the effects of flow rate and frequency of dynamic stimulus on the profiles of biochemical concentration which exposed on captured cells. In this array chip, the captured single cells in each trapping channels were able to receive identical extracellular dynamic biochemical stimuli which being transmitted from the entrance at the middle of the microfluidic array. Besides, after loading dynamic Adenosine Triphosphate (ATP) stimulation on captured cells by this device, consistent average intracellular Ca2+ dynamics phase and cellular heterogeneity were observed in captured single K562 cells. Furthermore, this device is able to be used for investigating cellular respond in single cells to temporally varying environments by modulating the stimulation signal in terms of concentration, pattern, and duration of exposure.
Collapse
|
33
|
Kumar M, Toprakhisar B, Van Haele M, Antoranz A, Boon R, Chesnais F, De Smedt J, Tricot T, Idoype TI, Canella M, Tilliole P, De Boeck J, Bajaj M, Ranga A, Bosisio FM, Roskams T, van Grunsven LA, Verfaillie CM. A fully defined matrix to support a pluripotent stem cell derived multi-cell-liver steatohepatitis and fibrosis model. Biomaterials 2021; 276:121006. [PMID: 34304139 DOI: 10.1016/j.biomaterials.2021.121006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/01/2021] [Indexed: 01/12/2023]
Abstract
Chronic liver injury, as observed in non-alcoholic steatohepatitis (NASH), progressive fibrosis, and cirrhosis, remains poorly treatable. Steatohepatitis causes hepatocyte loss in part by a direct lipotoxic insult, which is amplified by derangements in the non-parenchymal cellular (NPC) interactive network wherein hepatocytes reside, including, hepatic stellate cells, liver sinusoidal endothelial cells and liver macrophages. To create an in vitro culture model encompassing all these cells, that allows studying liver steatosis, inflammation and fibrosis caused by NASH, we here developed a fully defined hydrogel microenvironment, termed hepatocyte maturation (HepMat) gel, that supports maturation and maintenance of pluripotent stem cell (PSC) derived hepatocyte- and NPC-like cells for at least one month. The HepMat-based co-culture system modeled key molecular and functional features of TGFβ-induced liver fibrosis and fatty-acid induced inflammation and fibrosis better than monocultures of its constituent cell populations. The novel co-culture system should open new avenues for studying mechanisms underlying liver steatosis, inflammation and fibrosis as well as for assessing drugs counteracting these effects.
Collapse
Affiliation(s)
- Manoj Kumar
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium.
| | - Burak Toprakhisar
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Matthias Van Haele
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Ruben Boon
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Francois Chesnais
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Jonathan De Smedt
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Tine Tricot
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Teresa Izuel Idoype
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Marco Canella
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Pierre Tilliole
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Jolan De Boeck
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Manmohan Bajaj
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium
| | - Adrian Ranga
- Biomechanics, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Francesca Maria Bosisio
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Tania Roskams
- Translational Cell & Tissue Research, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Leo A van Grunsven
- Liver Cell Biology Research Group, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catherine M Verfaillie
- Stem Cell Institute, Department of Stem Cell and Developmental Biology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
34
|
Innovation in bioanalytical strategies and in vitro drug-drug interaction study approaches in drug discovery. Bioanalysis 2021; 13:513-532. [PMID: 33682424 DOI: 10.4155/bio-2021-0001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Failure to evaluate actual toxicities of investigational molecules in drug discovery is majorly due to inadequate evaluation of their pharmacokinetics. Limitation of conventional drug metabolism profiling procedure demands advancement of existing approaches. Various techniques such as 3D cell culture system, bio microfluidic OoC model, sandwich culture model is in pipeline to be employed at their full potential in drug discovery phase. Although they outweigh the conventional techniques in various aspects, a more detailed exploration of applicability in terms of automation and high throughput analysis is required. This review extensively discusses various ongoing innovations in bioanalytical techniques. The review also proposed various scientific strategies to be adopted for prior assessment of interaction possibilities in translational drug discovery research.
Collapse
|
35
|
Serras AS, Rodrigues JS, Cipriano M, Rodrigues AV, Oliveira NG, Miranda JP. A Critical Perspective on 3D Liver Models for Drug Metabolism and Toxicology Studies. Front Cell Dev Biol 2021; 9:626805. [PMID: 33732695 PMCID: PMC7957963 DOI: 10.3389/fcell.2021.626805] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
The poor predictability of human liver toxicity is still causing high attrition rates of drug candidates in the pharmaceutical industry at the non-clinical, clinical, and post-marketing authorization stages. This is in part caused by animal models that fail to predict various human adverse drug reactions (ADRs), resulting in undetected hepatotoxicity at the non-clinical phase of drug development. In an effort to increase the prediction of human hepatotoxicity, different approaches to enhance the physiological relevance of hepatic in vitro systems are being pursued. Three-dimensional (3D) or microfluidic technologies allow to better recapitulate hepatocyte organization and cell-matrix contacts, to include additional cell types, to incorporate fluid flow and to create gradients of oxygen and nutrients, which have led to improved differentiated cell phenotype and functionality. This comprehensive review addresses the drug-induced hepatotoxicity mechanisms and the currently available 3D liver in vitro models, their characteristics, as well as their advantages and limitations for human hepatotoxicity assessment. In addition, since toxic responses are greatly dependent on the culture model, a comparative analysis of the toxicity studies performed using two-dimensional (2D) and 3D in vitro strategies with recognized hepatotoxic compounds, such as paracetamol, diclofenac, and troglitazone is performed, further highlighting the need for harmonization of the respective characterization methods. Finally, taking a step forward, we propose a roadmap for the assessment of drugs hepatotoxicity based on fully characterized fit-for-purpose in vitro models, taking advantage of the best of each model, which will ultimately contribute to more informed decision-making in the drug development and risk assessment fields.
Collapse
Affiliation(s)
- Ana S. Serras
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana S. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Madalena Cipriano
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Armanda V. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno G. Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
36
|
Son J, Kim HH, Lee JH, Jeong WI, Park JK. Assembly and Disassembly of the Micropatterned Collagen Sheets Containing Cells for Location-Based Cellular Function Analysis. BIOCHIP JOURNAL 2021. [DOI: 10.1007/s13206-021-00007-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Farooqi HMU, Kang B, Khalid MAU, Salih ARC, Hyun K, Park SH, Huh D, Choi KH. Real-time monitoring of liver fibrosis through embedded sensors in a microphysiological system. NANO CONVERGENCE 2021; 8:3. [PMID: 33528697 PMCID: PMC7855143 DOI: 10.1186/s40580-021-00253-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/18/2021] [Indexed: 05/03/2023]
Abstract
Hepatic fibrosis is a foreshadowing of future adverse events like liver cirrhosis, liver failure, and cancer. Hepatic stellate cell activation is the main event of liver fibrosis, which results in excessive extracellular matrix deposition and hepatic parenchyma's disintegration. Several biochemical and molecular assays have been introduced for in vitro study of the hepatic fibrosis progression. However, they do not forecast real-time events happening to the in vitro models. Trans-epithelial electrical resistance (TEER) is used in cell culture science to measure cell monolayer barrier integrity. Herein, we explored TEER measurement's utility for monitoring fibrosis development in a dynamic cell culture microphysiological system. Immortal HepG2 cells and fibroblasts were co-cultured, and transforming growth factor β1 (TGF-β1) was used as a fibrosis stimulus to create a liver fibrosis-on-chip model. A glass chip-based embedded TEER and reactive oxygen species (ROS) sensors were employed to gauge the effect of TGF-β1 within the microphysiological system, which promotes a positive feedback response in fibrosis development. Furthermore, albumin, Urea, CYP450 measurements, and immunofluorescent microscopy were performed to correlate the following data with embedded sensors responses. We found that chip embedded electrochemical sensors could be used as a potential substitute for conventional end-point assays for studying fibrosis in microphysiological systems.
Collapse
Affiliation(s)
| | - Bohye Kang
- Department of Mechatronics Engineering, Jeju National University, Jeju-si, Republic of Korea
| | | | | | - Kinam Hyun
- Department of Mechatronics Engineering, Jeju National University, Jeju-si, Republic of Korea
| | - Sung Hyuk Park
- Department of Mechatronics Engineering, Jeju National University, Jeju-si, Republic of Korea
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, USA
| | - Kyung Hyun Choi
- Department of Mechatronics Engineering, Jeju National University, Jeju-si, Republic of Korea.
| |
Collapse
|
38
|
Deng J, Cong Y, Han X, Wei W, Lu Y, Liu T, Zhao W, Lin B, Luo Y, Zhang X. A liver-on-a-chip for hepatoprotective activity assessment. BIOMICROFLUIDICS 2020; 14:064107. [PMID: 33312328 PMCID: PMC7710384 DOI: 10.1063/5.0024767] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/14/2020] [Indexed: 05/02/2023]
Abstract
Hepatoprotectant is critical for the treatment of liver disease. This study first reported the application of a liver chip in the hepatoprotective effect assessment. We first established a biomimetic sinusoid-on-a-chip by laminating four types of hepatic cell lines (HepG2, HUVEC, LX-2, and U937 cells) in a single microchannel with the help of laminar flow in the microchannel and some micro-fences. This chip was straightforward to fabricate and operate and was able to be long-term cultured. It also demonstrated better hepatic activity (cell viability, albumin synthesis, urea secretion, and cytochrome P450 enzyme activities) over the traditional planar cell culture model. Then, we loaded three hepatoprotectants (tiopronin, bifendatatum, and glycyrrhizinate) into the chip followed by the addition of acetaminophen as a toxin. We successfully observed the hepatoprotective effect of these hepatoprotectants in the chip, and we also found that bifendatatum predominantly reduced alanine transaminase secretion, tiopronin predominantly reduced lactate dehydrogenase secretion, and glycyrrhizinate predominantly reduced aspartate transaminase secretion, which revealed the different mechanisms of these hepatoprotectants and provided a clue for following molecular biological study of the protecting mechanism.
Collapse
Affiliation(s)
| | - Ye Cong
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, 116024 Dalian, China
| | - Xiahe Han
- College of Pharmaceutical Science, Soochow University, 215123 Suzhou, China
| | - Wenbo Wei
- Shenzhen Institute of Geriatrics & Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 518000 Shenzhen, China
| | - Yao Lu
- Biotechnologhy Division, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, 116044 Dalian, China
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, 116024 Dalian, China
| | - Bingcheng Lin
- Biotechnologhy Division, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Pharmaceutical Sciences, School of Chemical Engineering, Dalian University of Technology, 116024 Dalian, China
- Authors to whom correspondence should be addressed: and
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, 215123 Suzhou, China
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
39
|
Lohasz C, Bonanini F, Hoelting L, Renggli K, Frey O, Hierlemann A. Predicting Metabolism-Related Drug-Drug Interactions Using a Microphysiological Multitissue System. ACTA ACUST UNITED AC 2020; 4:e2000079. [PMID: 33073544 DOI: 10.1002/adbi.202000079] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 09/30/2020] [Indexed: 12/20/2022]
Abstract
Drug-drug interactions (DDIs) occur when the pharmacological activity of one drug is altered by a second drug. As multimorbidity and polypharmacotherapy are becoming more common due to the increasing age of the population, the risk of DDIs is massively increasing. Therefore, in vitro testing methods are needed to capture such multiorgan events. Here, a scalable, gravity-driven microfluidic system featuring 3D microtissues (MTs) that represent different organs for the prediction of drug-drug interactions is used. Human liver microtissues (hLiMTs) are combined with tumor microtissues (TuMTs) and treated with drug combinations that are known to cause DDIs in vivo. The testing system is able to capture and quantify DDIs upon co-administration of the anticancer prodrugs cyclophosphamide or ifosfamide with the antiretroviral drug ritonavir. Dosage of ritonavir inhibits hepatic metabolization of the two prodrugs to different extents and decreases their efficacy in acting on TuMTs. The flexible MT compartment design of the system, the use of polystyrene as chip material, and the assembly of several chips in stackable plates offer the potential to significantly advance preclinical substance testing. The possibility of testing a broad variety of drug combinations to identify possible DDIs will improve the drug development process and increase patient safety.
Collapse
Affiliation(s)
- Christian Lohasz
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| | - Flavio Bonanini
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| | | | - Kasper Renggli
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| | | | - Andreas Hierlemann
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, 4058, Switzerland
| |
Collapse
|
40
|
Moradi E, Jalili-Firoozinezhad S, Solati-Hashjin M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomater 2020; 116:67-83. [PMID: 32890749 DOI: 10.1016/j.actbio.2020.08.041] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/08/2023]
Abstract
The liver is the largest internal organ of the body with complex microarchitecture and function that plays critical roles in drug metabolism. Hepatotoxicity and drug-induced liver injury (DILI) caused by various drugs is the main reason for late-stage drug failures. Moreover, liver diseases are among the leading causes of death in the world, with the number of new cases arising each year. Although animal models have been used to understand human drug metabolism and toxicity before clinical trials, tridimensional microphysiological systems, such as liver-on-a-chip (Liver Chip) platforms, could better recapitulate features of human liver physiology and pathophysiology and thus, are often more predictive of human outcome. Liver Chip devices have shown promising results in mimicking in vivo condition by recapitulating the sinusoidal structure of the liver, maintaining high cell viability and cellular phenotypes, and emulating native liver functions. Here, we first review the cellular constituents and physiology of the liver and then critically discuss the state-of-the-art chip-based liver models and their applications in drug screening, disease modeling, and regenerative medicine. We finally address the pending issues of existing platforms and touch upon future directions for developing new, advanced on-chip models.
Collapse
Affiliation(s)
- Ehsanollah Moradi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Mehran Solati-Hashjin
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran.
| |
Collapse
|
41
|
3D In Vitro Human Organ Mimicry Devices for Drug Discovery, Development, and Assessment. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/6187048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The past few decades have shown significant advancement as complex in vitro humanized systems have substituted animal trials and 2D in vitro studies. 3D humanized platforms mimic the organs of interest with their stimulations (physical, electrical, chemical, and mechanical). Organ-on-chip devices, including in vitro modelling of 3D organoids, 3D microfabrication, and 3D bioprinted platforms, play an essential role in drug discovery, testing, and assessment. In this article, a thorough review is provided of the latest advancements in the area of organ-on-chip devices targeting liver, kidney, lung, gut, heart, skin, and brain mimicry devices for drug discovery, development, and/or assessment. The current strategies, fabrication methods, and the specific application of each device, as well as the advantages and disadvantages, are presented for each reported platform. This comprehensive review also provides some insights on the challenges and future perspectives for the further advancement of each organ-on-chip device.
Collapse
|
42
|
Meng D, Lei X, Li Y, Kong Y, Huang D, Zhang G. Three dimensional polyvinyl alcohol scaffolds modified with collagen for HepG2 cell culture. J Biomater Appl 2020; 35:459-470. [DOI: 10.1177/0885328220933505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The creation of in vitro functional hepatic tissue simulating micro environmental niche of the native liver is a keen area of research due to its demand in bioartificial liver. However, it is still unclear how to maintain benign cell function while achieving the sufficient cell quantity. In this work, we aim to prepare a novel scaffold for the culture of HepG2 cells, a liver cell line, by modifying polyvinyl alcohol (PVA) scaffold with collagen (COL). PVA is a kind of synthetic biostable polymer with high hydrophilicity in the human body, has been widely used in the biomedical field. However, the use of PVA is limited in cell cultures due to lack of biologically active functional groups. In this study, amino silane (KH-550), glutaraldehyde and native type I collagen were used to modify three-dimensional PVA scaffold to establish a suitable composite scaffold for hepatocyte culture. Three types of composite scaffolds were prepared for different collagen content, named as PVA/COL (0.2%), PVA/COL (0.5%) and PVA/COL (0.8%), respectively. The composite scaffolds were characterized by SEM, XPS, FTIR, MS, porosity estimation and water contact angle measurement. The PVA/COL (0.8%) scaffolds had the highest collagen content of 12.13%. The composite scaffold showed high porosity with interconnected pores. Furthermore, the biocompatibility between HepG2 cells and scaffolds was evaluated by the ability of cell proliferation, albumin secretion, as well as urea synthesis. The coating of collagen on PVA scaffolds promoted hydrophilicity and HepG2 cell adhesion. Additionally, enhanced cell proliferation, increased albumin secretion and urea synthesis were observed in HepG2 cells growing on collagen-coated three-dimensional PVA scaffolds.
Collapse
Affiliation(s)
- Di Meng
- College of Life Sciences, Nankai University, Tianjin, China
| | - Xiongxin Lei
- Institute of Process Engineering, National Key Laboratory of Biochemical Engineering, Beijing, China
| | - Yang Li
- Institute of Process Engineering, National Key Laboratory of Biochemical Engineering, Beijing, China
- Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yingjun Kong
- Institute of Process Engineering, National Key Laboratory of Biochemical Engineering, Beijing, China
| | - Dawei Huang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Guifeng Zhang
- Institute of Process Engineering, National Key Laboratory of Biochemical Engineering, Beijing, China
| |
Collapse
|
43
|
Cong Y, Han X, Wang Y, Chen Z, Lu Y, Liu T, Wu Z, Jin Y, Luo Y, Zhang X. Drug Toxicity Evaluation Based on Organ-on-a-chip Technology: A Review. MICROMACHINES 2020; 11:E381. [PMID: 32260191 PMCID: PMC7230535 DOI: 10.3390/mi11040381] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Abstract
Organ-on-a-chip academic research is in its blossom. Drug toxicity evaluation is a promising area in which organ-on-a-chip technology can apply. A unique advantage of organ-on-a-chip is the ability to integrate drug metabolism and drug toxic processes in a single device, which facilitates evaluation of toxicity of drug metabolites. Human organ-on-a-chip has been fabricated and used to assess drug toxicity with data correlation with the clinical trial. In this review, we introduced the microfluidic chip models of liver, kidney, heart, nerve, and other organs and multiple organs, highlighting the application of these models in drug toxicity detection. Some biomarkers of toxic injury that have been used in organ chip platforms or have potential for use on organ chip platforms are summarized. Finally, we discussed the goals and future directions for drug toxicity evaluation based on organ-on-a-chip technology.
Collapse
Affiliation(s)
- Ye Cong
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116023, China;
| | - Xiahe Han
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; (X.H.); (Y.W.)
| | - Youping Wang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; (X.H.); (Y.W.)
| | - Zongzheng Chen
- Health Science Center, Shenzhen University, Shenzhen 518060, China; (Z.C.); (Z.W.); (Y.J.)
| | - Yao Lu
- Biotechnologhy Division, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Tingjiao Liu
- College of Stomatology, Dalian Medical University, Dalian 116011, China;
| | - Zhengzhi Wu
- Health Science Center, Shenzhen University, Shenzhen 518060, China; (Z.C.); (Z.W.); (Y.J.)
| | - Yu Jin
- Health Science Center, Shenzhen University, Shenzhen 518060, China; (Z.C.); (Z.W.); (Y.J.)
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116023, China;
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China; (X.H.); (Y.W.)
| |
Collapse
|
44
|
da Silva Morais A, Vieira S, Zhao X, Mao Z, Gao C, Oliveira JM, Reis RL. Advanced Biomaterials and Processing Methods for Liver Regeneration: State-of-the-Art and Future Trends. Adv Healthc Mater 2020; 9:e1901435. [PMID: 31977159 DOI: 10.1002/adhm.201901435] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Liver diseases contribute markedly to the global burden of mortality and disease. The limited organ disposal for orthotopic liver transplantation results in a continuing need for alternative strategies. Over the past years, important progress has been made in the field of tissue engineering (TE). Many of the early trials to improve the development of an engineered tissue construct are based on seeding cells onto biomaterial scaffolds. Nowadays, several TE approaches have been developed and are applied to one vital organ: the liver. Essential elements must be considered in liver TE-cells and culturing systems, bioactive agents or growth factors (GF), and biomaterials and processing methods. The potential of hepatocytes, mesenchymal stem cells, and others as cell sources is demonstrated. They need engineered biomaterial-based scaffolds with perfect biocompatibility and bioactivity to support cell proliferation and hepatic differentiation as well as allowing extracellular matrix deposition and vascularization. Moreover, they require a microenvironment provided using conventional or advanced processing technologies in order to supply oxygen, nutrients, and GF. Herein the biomaterials and the conventional and advanced processing technologies, including cell-sheets process, 3D bioprinting, and microfluidic systems, as well as the future trends in these major fields are discussed.
Collapse
Affiliation(s)
- Alain da Silva Morais
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Sílvia Vieira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
| | - Xinlian Zhao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and FunctionalizationDepartment of Polymer Science and EngineeringZhejiang University Hangzhou 310027 China
| | - Joaquim M. Oliveira
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| | - Rui L. Reis
- 3B's Research GroupI3Bs – Research Institute on Biomaterials, Biodegradables and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine 4805‐017 Barco Guimarães Portugal
- ICVS/3B's–PT Government Associate Laboratory Braga/ Guimarães Portugal
- The Discoveries Centre for Regenerative and Precision MedicineUniversity of Minho 4805‐017 Barco Guimarães Portugal
| |
Collapse
|
45
|
Argentati C, Tortorella I, Bazzucchi M, Morena F, Martino S. Harnessing the Potential of Stem Cells for Disease Modeling: Progress and Promises. J Pers Med 2020; 10:E8. [PMID: 32041088 PMCID: PMC7151621 DOI: 10.3390/jpm10010008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/18/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
Ex vivo cell/tissue-based models are an essential step in the workflow of pathophysiology studies, assay development, disease modeling, drug discovery, and development of personalized therapeutic strategies. For these purposes, both scientific and pharmaceutical research have adopted ex vivo stem cell models because of their better predictive power. As matter of a fact, the advancing in isolation and in vitro expansion protocols for culturing autologous human stem cells, and the standardization of methods for generating patient-derived induced pluripotent stem cells has made feasible to generate and investigate human cellular disease models with even greater speed and efficiency. Furthermore, the potential of stem cells on generating more complex systems, such as scaffold-cell models, organoids, or organ-on-a-chip, allowed to overcome the limitations of the two-dimensional culture systems as well as to better mimic tissues structures and functions. Finally, the advent of genome-editing/gene therapy technologies had a great impact on the generation of more proficient stem cell-disease models and on establishing an effective therapeutic treatment. In this review, we discuss important breakthroughs of stem cell-based models highlighting current directions, advantages, and limitations and point out the need to combine experimental biology with computational tools able to describe complex biological systems and deliver results or predictions in the context of personalized medicine.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| |
Collapse
|
46
|
Deng J, Wei W, Chen Z, Lin B, Zhao W, Luo Y, Zhang X. Engineered Liver-on-a-Chip Platform to Mimic Liver Functions and Its Biomedical Applications: A Review. MICROMACHINES 2019; 10:E676. [PMID: 31591365 PMCID: PMC6843249 DOI: 10.3390/mi10100676] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/03/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Hepatology and drug development for liver diseases require in vitro liver models. Typical models include 2D planar primary hepatocytes, hepatocyte spheroids, hepatocyte organoids, and liver-on-a-chip. Liver-on-a-chip has emerged as the mainstream model for drug development because it recapitulates the liver microenvironment and has good assay robustness such as reproducibility. Liver-on-a-chip with human primary cells can potentially correlate clinical testing. Liver-on-a-chip can not only predict drug hepatotoxicity and drug metabolism, but also connect other artificial organs on the chip for a human-on-a-chip, which can reflect the overall effect of a drug. Engineering an effective liver-on-a-chip device requires knowledge of multiple disciplines including chemistry, fluidic mechanics, cell biology, electrics, and optics. This review first introduces the physiological microenvironments in the liver, especially the cell composition and its specialized roles, and then summarizes the strategies to build a liver-on-a-chip via microfluidic technologies and its biomedical applications. In addition, the latest advancements of liver-on-a-chip technologies are discussed, which serve as a basis for further liver-on-a-chip research.
Collapse
Affiliation(s)
- Jiu Deng
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Wenbo Wei
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Zongzheng Chen
- Integrated Chinese and Western Medicine Postdoctoral research station, Jinan University, Guangzhou 510632, China;
| | - Bingcheng Lin
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
47
|
Deng J, Chen Z, Zhang X, Luo Y, Wu Z, Lu Y, Liu T, Zhao W, Lin B. A liver-chip-based alcoholic liver disease model featuring multi-non-parenchymal cells. Biomed Microdevices 2019; 21:57. [DOI: 10.1007/s10544-019-0414-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|