1
|
Ramírez-González GA, Consumi-Tubito C, Vargas-Méndez E, Centeno-Cerdas C. Advancing Organ-on-a-Chip Systems: The Role of Scaffold Materials and Coatings in Engineering Cell Microenvironment. Polymers (Basel) 2025; 17:1263. [PMID: 40363048 PMCID: PMC12074455 DOI: 10.3390/polym17091263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
For organ-on-a-chip (OoC) engineering, the use of biocompatible coatings and materials is not only recommended but essential. Extracellular matrix (ECM) components are commonly used as coatings due to their effects on cell orientation, protein expression, differentiation, and adhesion. Among the most frequently used coatings are collagen, fibronectin, and Matrigel, according to the specific cell type and intended OoC application. Additionally, materials such as polydimethylsiloxane (PDMS), thermoplastics, chitosan, and alginate serve as scaffolding components due to their biomechanical properties and biocompatibility. Here, we discuss some of the most employed coating techniques, including SAMs, dip coating, spin coating, microcontact printing, and 3D bioprinting, each offering advantages and drawbacks. Current challenges comprise enhancing biocompatibility, exploring novel materials, and improving scalability and reproducibility.
Collapse
Affiliation(s)
- Guido Andrés Ramírez-González
- Master’s Program in Medical Device Engineering, School of Materials Science and Engineering, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
- Biotechnology Research Center, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
| | - Chiara Consumi-Tubito
- Biotechnology Research Center, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
| | - Ernesto Vargas-Méndez
- Department of Biochemistry, School of Medicine, University of Costa Rica, San Jose 11501-2060, Costa Rica;
| | - Carolina Centeno-Cerdas
- Biotechnology Research Center, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
| |
Collapse
|
2
|
Schellberg BG, Koppes AN, Koppes RA. In situ monitoring of barrier function on-chip via automated, non-invasive luminescence sensing. LAB ON A CHIP 2025. [PMID: 40181784 PMCID: PMC11969330 DOI: 10.1039/d4lc01090f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/28/2025] [Indexed: 04/05/2025]
Abstract
Over the past 30 years, organs-on-a-chip (OOCs) have emerged as a robust alternative to address the technological challenges associated with current in vitro and in vivo options. Although OOCs offer improved bio-relevance and controlled complexity, broad adoption has remained limited. Most approaches to characterize on-chip structure and function require human intervention, limiting device translation and feasibility. Here, we introduce a new fiber optic-based sensing platform that enables automated, temporal luminescence sensing on-chip, validated for real-time readout of epithelial and endothelial barrier function under cytokine-induced inflammation. Our platform, capable of at least 1 μM resolution, tracked paracellular transport in situ for 9 days of culture under perfusion on-chip. These results offer an alternative sensing approach for continuous, non-invasive luminescence monitoring in OOCs.
Collapse
Affiliation(s)
- Bryan G Schellberg
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
| | - Abigail N Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA.
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Truong N, Zahra A, Lintao RCV, Chauhan R, Bento GF, Vidal Jr. M, Kim S, Lam PY, Conrads T, Conrads K, Han A, Menon R, Richardson LS. Modeling reproductive and pregnancy-associated tissues using organ-on-chip platforms: challenges, limitations, and the high throughput data frontier. Front Bioeng Biotechnol 2025; 13:1568389. [PMID: 40236940 PMCID: PMC11996799 DOI: 10.3389/fbioe.2025.1568389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Over the past decade, organ-on-chip technology (microphysiological systems or tissue chips) has reshaped in-vitro physiological and pathological modeling and pharmaceutical drug assessment. FDA Modernization Act 2.0 allows for alternatives to animal testing or the use of appropriate non-animal models/new approach methods (NAMs), such as Organ-on-chips (OC) platforms or in silico simulation models, to generate pre-clinical in-vitro drug trial data for regulatory purposes primes the microfluidic field to have exponential growth in the coming years. The changes in the approaches of regulatory agencies could significantly impact the development of therapeutics for use during pregnancy. However, limitations of the devices and molecular and biochemical assay shortfalls hinder the progress of the OOC field. This review describes available reproductive and pregnancy-related OOC platforms, and the current methodologies utilized to generate endpoint datasets (e.g., microscopic imaging, immunocytochemistry, real-time polymerase chain reaction, cytokine multiplex analysis). Microfluidic platform limitations, such as fewer number of cells or low supernatant volumes and restrictions regarding fabrication materials, are described. Novel approaches (e.g., spatial transcriptomics, imaging cytometry by time of flight, exosomes analysis using Exoview) to overcome these challenges are described. OOC platforms are primed to provide biologically relevant and clinically translational data that can revolutionize in-vitro physiological modeling, drug discovery, and toxicologic risk assessment. However, engineering adaptations to increase the throughput of devices (i.e., device arrays) and biological advancements to improve data throughput are both needed for these platforms to reach their full potential.
Collapse
Affiliation(s)
- Nina Truong
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Abir Zahra
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Ryan C. V. Lintao
- Institute of Reproductive Health, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Rahul Chauhan
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Giovana Fernanda Bento
- Department of Pathology, Botucatu Medical School, São Paulo State University, São Paulo, Brazil
| | - Manuel Vidal Jr.
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- College of Medicine, San Beda University, Manila, Philippines
- Department of Chemistry, College of Science, De La Salle University Manila, Manila, Philippines
| | - Sungjin Kim
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Po Yi Lam
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Thomas Conrads
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, Falls Church, VA, United States
| | - Kelly Conrads
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, Falls Church, VA, United States
| | - Arum Han
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Lauren S. Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Wang H, Danoy M, Gong Y, Utami T, Arakawa H, Kato Y, Nishikawa M, Sakai Y, Leclerc E. Palmitic Acid Induced a Dedifferentiation Profile at the Transcriptome Level: A Collagen Synthesis but no Triglyceride Accumulation in Hepatocyte-Like Cells Derived From Human-Induced Pluripotent Stem Cells Cultivated Inside Organ on a Chip. J Appl Toxicol 2025; 45:460-471. [PMID: 39506029 DOI: 10.1002/jat.4714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 11/08/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the main causes of critical liver diseases leading to steatosis, steatohepatitis, fibrosis, and ultimately to liver cirrhosis and hepatic carcinoma. In this study, the effect of palmitic acid (PA), one of the most abundant dietary fatty acids, was investigated using an organ-on-a-chip (OoC) technology on hepatocyte-like cells derived from human-induced pluripotent stem cells (hiPSCs). After 1 week of hepatic maturation, followed by 1 week of exposure, the transcriptomic analysis showed lower liver transcription factor activity. It also revealed that 318 genes were differentially expressed between the control and 0.5-mM PA conditions. The 0.5-mM PA conditions were characterized by the downregulation of hepatic markers (liver transcription factors, phase I and phase II metabolism genes) of lipidic genes (metabolism and transport). In parallel, the 0.5-mM PA treatment upregulated several extracellular matrix genes (such as collagen genes). The physiopathological staining demonstrated no lipid accumulation in our model and confirmed the secretion of collagen in the 0.5-mM PA conditions. However, the production of albumin, the metabolic biotransformation by the cytochrome P450 enzymes, and the biliary acid concentrations were not altered by the PA treatments. Overall, our data illustrated the response to PA characterized by an early stage of dedifferentiation observed at the transcriptomic levels associated with a modification of the collagenic profile but without lipid accumulation. We believe that our model provides new insight of the onset of palmitic lipotoxicity in the early stage of NAFLD.
Collapse
Affiliation(s)
- Hanyuan Wang
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- CNRS/IIS IRL 2820; Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Mathieu Danoy
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Ya Gong
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Tia Utami
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
- CNRS/IIS IRL 2820; Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| | - Eric Leclerc
- CNRS/IIS IRL 2820; Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Silva B, Bragança J. Induced pluripotent stem cell-derived mesenchymal stem cells for modeling and treating metabolic associated fatty liver disease and metabolic associated steatohepatitis: Challenges and opportunities. World J Stem Cells 2025; 17:99331. [DOI: 10.4252/wjsc.v17.i2.99331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
The potential of induced pluripotent stem cells (iPSCs) for modeling and treating metabolic associated fatty liver disease (MAFLD) and metabolic associated steatohepatitis (MASH) is emerging. MAFLD is a growing global health concern, currently with limited treatment options. While primary mesenchymal stem cells hold promise, iPSCs offer a versatile alternative due to their ability to differentiate into various cell types, including iPSC-derived mesenchymal stem cells. However, challenges remain, including optimizing differentiation protocols, ensuring cell safety, and addressing potential tumorigenicity risks. In addition, iPSCs offer the possibility to generate complex cellular models, including three-dimensional organoid models, which are closer representations of the human disease than animal models. Those models would also be valuable for drug discovery and personalized medicine approaches. Overall, iPSCs and their derivatives offer new perspectives for advancing MAFLD/MASH research and developing novel therapeutic strategies. Further research is needed to overcome current limitations and translate this potential into effective clinical applications.
Collapse
Affiliation(s)
- Bárbara Silva
- Algarve Biomedical Center-Research Institute, University of Algarve, Faro 8005-139, Portugal
- Algarve Biomedical Center, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
| | - José Bragança
- Algarve Biomedical Center, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, Algarve Biomedical Center-Research Institute, University of Algarve, Faro 8005-139, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon 1000-001, Portugal
| |
Collapse
|
8
|
Perry AS, Hadad N, Chatterjee E, Jimenez-Ramos M, Farber-Eger E, Roshani R, Stolze LK, Betti MJ, Zhao S, Huang S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Vaitinadin NS, Freedman JE, Tanriverdi K, Alsop E, Van Keuren-Jensen K, Sauld JFK, Mahajan G, Khan SS, Colangelo L, Nayor M, Fisher-Hoch S, McCormick JB, North KE, Below JE, Wells QS, Abel ED, Kalhan R, Scott C, Guilliams M, Gamazon ER, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. Cell Rep Med 2024; 5:101871. [PMID: 39657669 PMCID: PMC11722105 DOI: 10.1016/j.xcrm.2024.101871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/06/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Hepatic steatosis is a central phenotype in multi-system metabolic dysfunction and is increasing in parallel with the obesity pandemic. We use a translational approach integrating clinical phenotyping and outcomes, circulating proteomics, and tissue transcriptomics to identify dynamic, functional biomarkers of hepatic steatosis. Using multi-modality imaging and broad proteomic profiling, we identify proteins implicated in the progression of hepatic steatosis that are largely encoded by genes enriched at the transcriptional level in the human liver. These transcripts are differentially expressed across areas of steatosis in spatial transcriptomics, and several are dynamic during stages of steatosis. Circulating multi-protein signatures of steatosis strongly associate with fatty liver disease and multi-system metabolic outcomes. Using a humanized "liver-on-a-chip" model, we induce hepatic steatosis, confirming cell-specific expression of prioritized targets. These results underscore the utility of this approach to identify a prognostic, functional, dynamic "liquid biopsy" of human liver, relevant to biomarker discovery and mechanistic research applications.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Niran Hadad
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Emeli Chatterjee
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Maria Jimenez-Ramos
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | | | - Rashedeh Roshani
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Michael J Betti
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shilin Zhao
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Shi Huang
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Liesbet Martens
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Timothy J Kendall
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK; Edinburgh Pathology, University of Edinburgh, Edinburgh, UK
| | - Tinne Thone
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | - Samuel Bailin
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Curtis L Gabriel
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Koethe
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - J Jeffrey Carr
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Jane E Freedman
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Eric Alsop
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | | | | | | | - Sadiya S Khan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Laura Colangelo
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Susan Fisher-Hoch
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Joseph B McCormick
- School of Public Health, The University of Texas Health Science Center at Houston, Brownsville, TX, USA
| | - Kari E North
- CVD Genetic Epidemiology Computational Laboratory, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quinn S Wells
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Ravi Kalhan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Charlotte Scott
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Myeloid Cell Biology in Tissue Homeostasis and Regeneration, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Eric R Gamazon
- Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| | - Ravi Shah
- Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
9
|
Campanile E, Colombi A, Bretti G. Two-step global sensitivity analysis of a non-local integro-differential model for Cancer-on-Chip experiments. Math Biosci 2024; 378:109330. [PMID: 39486639 DOI: 10.1016/j.mbs.2024.109330] [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: 05/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/04/2024]
Abstract
The present work focuses on a non-local integro-differential model reproducing Cancer-on-chip experiments where tumor cells, treated with chemotherapy drugs, secrete chemical signals stimulating the immune response. The reliability of the model in reproducing the phenomenon of interest is investigated through a global sensitivity analysis, rather than a local one, to have global information about the role of parameters, and by examining potential non-linear effects in greater detail. Focusing on a region in the parameter space, the effect of 13 model parameters on the in silico outcome is investigated by considering 11 different target outputs, properly selected to monitor the spatial distribution and the dynamics of immune cells along the period of observation. In order to cope with the large number of model parameters to be investigated and the computational cost of each numerical simulation, a two-step global sensitivity analysis is performed. First, the screening Morris method is applied to rank the effect of the 13 model parameters on each target output and it emerges that all the output targets are mainly affected by the same 6 parameters. The extended Fourier Amplitude Sensitivity Test (eFAST) method is then used to quantify the role of these 6 parameters. As a result, the proposed analysis highlights the feasibility of the considered space of parameters, and indicates that the most relevant parameters are those related to the chemical field and cell-substrate adhesion. In turn, it suggests how to possibly improve the model description as well as the calibration procedure, in order to better capture the observed phenomena and, at the same time, reduce the complexity of the simulation algorithm. On one hand, the model could be simplified by neglecting cell-cell alignment effects unless clear empirical evidences of their importance emerge. On the other hand, the best way to increase the accuracy and reliability of our model predictions would be to have experimental data/information to reduce the uncertainty of the more relevant parameters.
Collapse
Affiliation(s)
- Elio Campanile
- Fondazione the Microsoft Research, University of Trento, Centre for Computational and Systems Biology (COSBI), Piazza Manifattura 1, Rovereto, 38068, Italy; Department of Mathematics, University of Trento, Via Calepina, 14, Trento, 38122, Italy
| | - Annachiara Colombi
- Department of Mathematical Sciences (DISMA) Politecnico di Torino, DISMA, C.so Duca degli Abruzzi 24, Torino, 10129, Italy.
| | - Gabriella Bretti
- Istituto per le Applicazioni del Calcolo, CNR, Via dei Taurini 19, Rome, 00185, Italy
| |
Collapse
|
10
|
Chang M, Fu A, Liu B, Wang Y, Zeng H. Dynamic Liver Chip Based on Well-Coupled Microfluidics: An Accurate NASH Model for Drug Evaluation. Anal Chem 2024; 96:16280-16288. [PMID: 39356613 DOI: 10.1021/acs.analchem.4c03234] [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: 10/04/2024]
Abstract
The convenient liver model in vitro recapitulating the hepatic functions, metabolism, and even steatohepatitis to perform the accurate drug evaluation is still challenging because of the unattainable hominine physiological microenvironment in vitro. Here, the progressed stages of nonalcoholic steatohepatitis (NASH) disease were precisely modeled to accurately evaluate the performance of antilipemic based on the dynamic liver chip adopting the well-coupled microfluidics, which well recapitulated the normal and steatohepatitis of liver in vitro. In brief, the mild nutrient flow and sufficient oxygen supply for parenchymal liver cells could be well supplied through the endothelial cells layer that mimicked the real physiological barrier of endothelium, while the loading of drugs might be obtained by directly adding drug into the running nutrient flow to mimic the intravenously administrable. The progressed degree of steatohepatitis could be directly reflected by the amount of intramyocellular lipid content (IMLC) of the HepG2 cell hepatocyte layer in wells that were induced by different concentrations of free fatty acids (FFA). To prove the concept of the liver chip in drug evaluation, an accurate assessment of the performance of firsocostat, the acetyl-CoA carboxylase (ACC) inhibitor of hepatic mitochondria of hepatocytes, was carried out. The subtle time dependence of firsocostat treatment to different progressed stages of NASH was clearly figured out. Therefore, we prospect the liver chip that adopted well-coupled microfluidics could be an accurate and standard liver model in vitro to carry out the antilipemic evaluation and screening, which significantly enlightens the drug evaluation by liver on chip in vitro.
Collapse
Affiliation(s)
- Mingyang Chang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Anchen Fu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Bingqian Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Yongxiang Wang
- Department of Gynaecology and Obstetrics, Shuguang Hospital, Affiliated with Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Shanghai 201203, China
| | - Hulie Zeng
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| |
Collapse
|
11
|
Qi L, Groeger M, Sharma A, Goswami I, Chen E, Zhong F, Ram A, Healy K, Hsiao EC, Willenbring H, Stahl A. Adipocyte inflammation is the primary driver of hepatic insulin resistance in a human iPSC-based microphysiological system. Nat Commun 2024; 15:7991. [PMID: 39266553 PMCID: PMC11393072 DOI: 10.1038/s41467-024-52258-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
Interactions between adipose tissue, liver and immune system are at the center of metabolic dysfunction-associated steatotic liver disease and type 2 diabetes. To address the need for an accurate in vitro model, we establish an interconnected microphysiological system (MPS) containing white adipocytes, hepatocytes and proinflammatory macrophages derived from isogenic human induced pluripotent stem cells. Using this MPS, we find that increasing the adipocyte-to-hepatocyte ratio moderately affects hepatocyte function, whereas macrophage-induced adipocyte inflammation causes lipid accumulation in hepatocytes and MPS-wide insulin resistance, corresponding to initiation of metabolic dysfunction-associated steatotic liver disease. We also use our MPS to identify and characterize pharmacological intervention strategies for hepatic steatosis and systemic insulin resistance and find that the glucagon-like peptide-1 receptor agonist semaglutide improves hepatocyte function by acting specifically on adipocytes. These results establish our MPS modeling the adipose tissue-liver axis as an alternative to animal models for mechanistic studies or drug discovery in metabolic diseases.
Collapse
Affiliation(s)
- Lin Qi
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Marko Groeger
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Aditi Sharma
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ishan Goswami
- Department of Bioengineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Erzhen Chen
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Fenmiao Zhong
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Apsara Ram
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Kevin Healy
- Department of Bioengineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Materials Science and Engineering, College of Engineering, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Edward C Hsiao
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Holger Willenbring
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Eli and Edythe Broad Center for Regeneration Medicine, University of California San Francisco, San Francisco, CA, 94143, USA.
- Liver Center, University of California San Francisco, San Francisco, CA, 94143, USA.
| | - Andreas Stahl
- Department of Nutritional Science and Toxicology, College of Natural Resources, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
12
|
Razavi Z, Soltani M, Pazoki-Toroudi H, Dabagh M. Microfluidic systems for modeling digestive cancer: a review of recent progress. Biomed Phys Eng Express 2024; 10:052002. [PMID: 39142294 DOI: 10.1088/2057-1976/ad6f15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 08/14/2024] [Indexed: 08/16/2024]
Abstract
Purpose. This review aims to highlight current improvements in microfluidic devices designed for digestive cancer simulation. The review emphasizes the use of multicellular 3D tissue engineering models to understand the complicated biology of the tumor microenvironment (TME) and cancer progression. The purpose is to develop oncology research and improve digestive cancer patients' lives.Methods. This review analyzes recent research on microfluidic devices for mimicking digestive cancer. It uses tissue-engineered microfluidic devices, notably organs on a chip (OOC), to simulate human organ function in the lab. Cell cultivation on modern three-dimensional hydrogel platforms allows precise geometry, biological components, and physiological qualities. The review analyzes novel methodologies, key findings, and technical progress to explain this field's advances.Results. This study discusses current advances in microfluidic devices for mimicking digestive cancer. Micro physiological systems with multicellular 3D tissue engineering models are emphasized. These systems capture complex biochemical gradients, niche variables, and dynamic cell-cell interactions in the tumor microenvironment (TME). These models reveal stomach cancer biology and progression by duplicating the TME. Recent discoveries and technology advances have improved our understanding of gut cancer biology, as shown in the review.Conclusion. Microfluidic systems play a crucial role in modeling digestive cancer and furthering oncology research. These platforms could transform drug development and treatment by revealing the complex biology of the tumor microenvironment and cancer progression. The review provides a complete summary of recent advances and suggests future research for field professionals. The review's major goal is to further medical research and improve digestive cancer patients' lives.
Collapse
Affiliation(s)
- ZahraSadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran
- Biochemistry Research Center, Iran University Medical Sciences, Tehran, Iran
| | - Madjid Soltani
- Department of Mechanical Engineering, K N Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, Waterloo, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
- Centre for Sustainable Business, International Business University, Toronto, Canada
| | | | - Mahsa Dabagh
- Department of Biomedical Engineering, University of Wisconsin-Milwaukee, WI 53211, United States of America
| |
Collapse
|
13
|
Karnawat K, Parthasarathy R, Sakhrie M, Karthik H, Krishna KV, Balachander GM. Building in vitro models for mechanistic understanding of liver regeneration in chronic liver diseases. J Mater Chem B 2024; 12:7669-7691. [PMID: 38973693 DOI: 10.1039/d4tb00738g] [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: 07/09/2024]
Abstract
The liver has excellent regeneration potential and attains complete functional recovery from partial hepatectomy. The regenerative mechanisms malfunction in chronic liver diseases (CLDs), which fuels disease progression. CLDs account for 2 million deaths per year worldwide. Pathophysiological studies with clinical correlation have shown evidence of deviation of normal regenerative mechanisms and its contribution to fueling fibrosis and disease progression. However, we lack realistic in vitro models that can allow experimental manipulation for mechanistic understanding of liver regeneration in CLDs and testing of candidate drugs. In this review, we aim to provide the framework for building appropriate organotypic models for dissecting regenerative responses in CLDs, with the focus on non-alcoholic steatohepatitis (NASH). By drawing parallels with development and hepatectomy, we explain the selection of critical components such as cells, signaling, and, substrate-driven biophysical cues to build an appropriate CLD model. We highlight the organoid-based organotypic models available for NASH disease modeling, including organ-on-a-chip and 3D bioprinted models. With the focus on bioprinting as a fabrication method, we prescribe building in vitro CLD models and testing schemes for exploring the regenerative responses in the bioprinted model.
Collapse
Affiliation(s)
- Khushi Karnawat
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Rithika Parthasarathy
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Mesevilhou Sakhrie
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Harikeshav Karthik
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Konatala Vibhuvan Krishna
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Gowri Manohari Balachander
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| |
Collapse
|
14
|
Rainu SK, Singh N. 3D microscaffolds with triple-marker sensitive nanoprobes for studying fatty liver disease in vitro. NANOSCALE 2024; 16:10048-10063. [PMID: 38712552 DOI: 10.1039/d4nr00434e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a heterogeneous condition that encompasses a wide range of liver diseases that progresses from simple hepatic steatosis to the life-threatening state of cirrhosis. However, due to the heterogeneity of this disease, comprehensive analysis of several physicochemical and biological factors that drive its progression is necessary. Therefore, an in vitro platform is required that would enable real-time monitoring of these changes to better understand the progression of these diseases. The earliest stage of NAFLD, i.e. hepatic steatosis, is characterised by triglyceride accumulation in the form of lipid vacuoles in the cytosol of hepatocytes. This fatty acid accumulation is usually accompanied by hepatic inflammation, leading to tissue acidification and dysregulated expression of certain proteases such as matrix metalloproteinases (MMPs). Taking cues from the biological parameters of the disease, we report here a 3D in vitro GelMA/alginate microscaffold platform encapsulating a triple-marker (pH, MMP-3 and MMP-9) sensitive fluorescent nanoprobe for monitoring, and hence, distinguishing the fatty liver disease (hepatic steatosis) from healthy livers on the basis of pH change and MMP expression. The nanoprobe consists of a carbon nanoparticle (CNP) core, which exhibits intrinsic pH-dependent fluorescence properties, decorated either with an MMP-3 (NpMMP3) or MMP-9 (NpMMP9) sensitive peptide substrate. These peptide substrates are flanked with a fluorophore-quencher pair that separates on enzymatic cleavage, resulting in fluorescence emission. The cocktail of these nanoprobes generated multiple fluorescence signals corresponding to slightly acidic pH (blue) and overexpression of MMP-3 (green) and MMP-9 (red) enzymes in a 3D in vitro fatty liver model, whereas no/negligible fluorescence signals were observed in a healthy liver model. Moreover, this platform enabled us to mimic fatty liver disease in a more realistic manner. Therefore, this 3D in vitro platform encapsulating triple-marker sensitive fluorescent nanoprobes would facilitate the monitoring of the changes in pH and MMP expression, thereby enabling us to distinguish a healthy liver from a diseased liver and to study liver disease stages on the basis of these markers.
Collapse
Affiliation(s)
- Simran Kaur Rainu
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India.
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
15
|
Zhang L, Yu Y, Wang Q, Huang X, Feng Z, Li Z. Oridonin loaded peptide nanovesicles alleviate nonalcoholic fatty liver disease in mice. Pharm Dev Technol 2024; 29:123-130. [PMID: 38327230 DOI: 10.1080/10837450.2024.2315460] [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: 09/30/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
This study was to construct a nanovesicle delivery system to improve the loading efficiency and stability of ORI for the treatment of nonalcoholic fatty liver disease (NAFLD). This nanovesicles (NVs) exerted a narrow size distribution (195.6 ± 11.49 nm) and high entrapment efficiency (84.46 ± 1.34%). In vitro cell studies demonstrated that the NVs treatment enhanced the cellular uptake of ORI and reduced lipid over-accumulation and total cholesterol levels in NAFLD cell model. At the same time, in vivo study proved that, compared with the normal group, the model group mice showed a decrease in body weight, a significant increase in liver index (6.71 ± 0.62, p < 0.01), and symptoms of liver lipid accumulation, lipid vesicles, and liver tissue fibrosis. Compared with the model group, after high-dose ORI NVs intervention, mice gained weight, decreased liver index (4.69 ± 0.55, p < 0.01), reduced hepatic lipid droplet vacuoles, reduced lipid accumulation (reduced oil red area, p < 0.001), and alleviated the degree of liver fibrosis (reduced blue collagen area, p < 0.001). In conclusion, ORI/HP-β-CD/H9-HePC NVs showed specific liver accumulation and improved therapeutic effects, the nano drug loading system provides a promising strategy for the encapsulation of ORI to effectively alleviate the process of NAFLD.
Collapse
Affiliation(s)
- Lifen Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yao Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qi Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xi Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zheng Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhi Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou, China
| |
Collapse
|
16
|
Perry AS, Hadad N, Chatterjee E, Ramos MJ, Farber-Eger E, Roshani R, Stolze LK, Zhao S, Martens L, Kendall TJ, Thone T, Amancherla K, Bailin S, Gabriel CL, Koethe J, Carr JJ, Terry JG, Freedman J, Tanriverdi K, Alsop E, Keuren-Jensen KV, Sauld JFK, Mahajan G, Khan S, Colangelo L, Nayor M, Fisher-Hoch S, McCormick J, North KE, Below J, Wells Q, Abel D, Kalhan R, Scott C, Guilliams M, Fallowfield JA, Banovich NE, Das S, Shah R. A prognostic molecular signature of hepatic steatosis is spatially heterogeneous and dynamic in human liver. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.26.24301828. [PMID: 38352394 PMCID: PMC10863022 DOI: 10.1101/2024.01.26.24301828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) prevalence is increasing in parallel with an obesity pandemic, calling for novel strategies for prevention and treatment. We defined a circulating proteome of human MASLD across ≈7000 proteins in ≈5000 individuals from diverse, at-risk populations across the metabolic health spectrum, demonstrating reproducible diagnostic performance and specifying both known and novel metabolic pathways relevant to MASLD (central carbon and amino acid metabolism, hepatocyte regeneration, inflammation, fibrosis, insulin sensitivity). A parsimonious proteomic signature of MASLD was associated with a protection from MASLD and its related multi-system metabolic consequences in >26000 free-living individuals, with an additive effect to polygenic risk. The MASLD proteome was encoded by genes that demonstrated transcriptional enrichment in liver, with spatial transcriptional activity in areas of steatosis in human liver biopsy and dynamicity for select targets in human liver across stages of steatosis. We replicated several top relations from proteomics and spatial tissue transcriptomics in a humanized "liver-on-a-chip" model of MASLD, highlighting the power of a full translational approach to discovery in MASLD. Collectively, these results underscore utility of blood-based proteomics as a dynamic "liquid biopsy" of human liver relevant to clinical biomarker and mechanistic applications.
Collapse
|
17
|
Carvalho AM, Bansal R, Barrias CC, Sarmento B. The Material World of 3D-Bioprinted and Microfluidic-Chip Models of Human Liver Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307673. [PMID: 37961933 DOI: 10.1002/adma.202307673] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Biomaterials are extensively used to mimic cell-matrix interactions, which are essential for cell growth, function, and differentiation. This is particularly relevant when developing in vitro disease models of organs rich in extracellular matrix, like the liver. Liver disease involves a chronic wound-healing response with formation of scar tissue known as fibrosis. At early stages, liver disease can be reverted, but as disease progresses, reversion is no longer possible, and there is no cure. Research for new therapies is hampered by the lack of adequate models that replicate the mechanical properties and biochemical stimuli present in the fibrotic liver. Fibrosis is associated with changes in the composition of the extracellular matrix that directly influence cell behavior. Biomaterials could play an essential role in better emulating the disease microenvironment. In this paper, the recent and cutting-edge biomaterials used for creating in vitro models of human liver fibrosis are revised, in combination with cells, bioprinting, and/or microfluidics. These technologies have been instrumental to replicate the intricate structure of the unhealthy tissue and promote medium perfusion that improves cell growth and function, respectively. A comprehensive analysis of the impact of material hints and cell-material interactions in a tridimensional context is provided.
Collapse
Affiliation(s)
- Ana Margarida Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| |
Collapse
|
18
|
R N, Aggarwal A, Sravani AB, Mallya P, Lewis S. Organ-On-A-Chip: An Emerging Research Platform. Organogenesis 2023; 19:2278236. [PMID: 37965897 PMCID: PMC10653779 DOI: 10.1080/15476278.2023.2278236] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/27/2023] [Indexed: 11/16/2023] Open
Abstract
In drug development, conventional preclinical and clinical testing stages rely on cell cultures and animal experiments, but these methods may fall short of fully representing human biology. To overcome this limitation, the emergence of organ-on-a-chip (OOC) technology has sparked interest as a transformative approach in drug testing research. By closely replicating human organ responses to external signals, OOC devices hold immense potential in revolutionizing drug efficacy and safety predictions. This review focuses on the advancements, applications, and prospects of OOC devices in drug testing. Based on the latest advances in the field of OOC systems and their clinical applications, this review reflects the effectiveness of OOC devices in replacing human volunteers in certain clinical studies. This review underscores the critical role of OOC technology in transforming drug testing methodologies.
Collapse
Affiliation(s)
- Nithin R
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Ayushi Aggarwal
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Anne Boyina Sravani
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Pooja Mallya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
19
|
Wang E, Andrade MJ, Smith Q. Vascularized liver-on-a-chip model to investigate nicotine-induced dysfunction. BIOMICROFLUIDICS 2023; 17:064108. [PMID: 38155919 PMCID: PMC10754629 DOI: 10.1063/5.0172677] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
The development of physiologically relevant in vitro systems for simulating disease onset and progression and predicting drug metabolism holds tremendous value in reducing drug discovery time and cost. However, many of these platforms lack accuracy in replicating the tissue architecture and multicellular interactions. By leveraging three-dimensional cell culture, biomimetic soft hydrogels, and engineered stimuli, in vitro models have continued to progress. Nonetheless, the incorporation of the microvasculature has been met with many challenges, specifically with the addition of parenchymal cell types. Here, a systematic approach to investigating the initial seeding density of endothelial cells and its effects on interconnected networks was taken and combined with hepatic spheroids to form a liver-on-a-chip model. Leveraging this system, nicotine's effects on microvasculature and hepatic function were investigated. The findings indicated that nicotine led to interrupted adherens junctions, decreased guanosine triphosphate cyclohydrolase 1 expression, impaired angiogenesis, and lowered barrier function, all key factors in endothelial dysfunction. With the combination of the optimized microvascular networks, a vascularized liver-on-a-chip was formed, providing functional xenobiotic metabolism and synthesis of both albumin and urea. This system provides insight into potential hepatotoxicity caused by various drugs and allows for assessing vascular dysfunction in a high throughput manner.
Collapse
Affiliation(s)
- Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | - Melisa J. Andrade
- Department of Chemical and Biomolecular Engineering, University of California Irvine, Irvine, California 92697, USA
| | | |
Collapse
|
20
|
Wiriyakulsit N, Keawsomnuk P, Thongin S, Ketsawatsomkron P, Muta K. A model of hepatic steatosis with declined viability and function in a liver-organ-on-a-chip. Sci Rep 2023; 13:17019. [PMID: 37813918 PMCID: PMC10562420 DOI: 10.1038/s41598-023-44198-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/04/2023] [Indexed: 10/11/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) begins with benign steatosis caused by ectopic storage of triacylglycerols in the liver. Persistent steatosis, in combination with other genetic and environmental factors, leads to nonalcoholic steatohepatitis (NASH) characterized by functional impairment, inflammation, and fibrosis. However, it remains unclear how persistent steatosis directly contributes to the progression of NAFLD, which may represent a therapeutic target. The organ-on-a-chip (OOC) has emerged as a new culture platform to recapitulate human pathological conditions under which drug candidates can be screened. Here, we developed a simple OOC steatosis model using the Mimetas OrganoPlate with a human liver cell line, HepG2. Treating the HepG2 OOCs with fatty acid overload induced steatosis within 24 h. Moreover, persistent steatosis for 6 days impaired OOC viability and hepatic function, as measured by a WST-8 assay and albumin production, respectively. Lastly, the HepG2 OOCs were exposed to drugs being tested in clinical trials for NAFLD/NASH during the 6-day period. Pioglitazone improved the OOC viability while elafibranor reduced the steatosis in association with reduced viability and albumin production. In conclusion, we show that the HepG2 steatosis OOC model is a useful tool on which the efficacy and toxicity of various therapeutic candidates can be tested.
Collapse
Affiliation(s)
- Natsupa Wiriyakulsit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Ploychanok Keawsomnuk
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Saowarose Thongin
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Pimonrat Ketsawatsomkron
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand
| | - Kenjiro Muta
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111 Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand.
| |
Collapse
|
21
|
Li Z, Li Q, Zhou C, Lu K, Liu Y, Xuan L, Wang X. Organoid-on-a-chip: Current challenges, trends, and future scope toward medicine. BIOMICROFLUIDICS 2023; 17:051505. [PMID: 37900053 PMCID: PMC10613095 DOI: 10.1063/5.0171350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/12/2023] [Indexed: 10/31/2023]
Abstract
In vitro organoid models, typically defined as 3D multicellular aggregates, have been extensively used as a promising tool in drug screening, disease progression research, and precision medicine. Combined with advanced microfluidics technique, organoid-on-a-chip can flexibly replicate in vivo organs within the biomimetic physiological microenvironment by accurately regulating different parameters, such as fluid conditions and concentration gradients of biochemical factors. Since engineered organ reconstruction has opened a new paradigm in biomedicine, innovative approaches are increasingly required in micro-nano fabrication, tissue construction, and development of pharmaceutical products. In this Perspective review, the advantages and characteristics of organoid-on-a-chip are first introduced. Challenges in current organoid culture, extracellular matrix building, and device manufacturing techniques are subsequently demonstrated, followed by potential alternative approaches, respectively. The future directions and emerging application scenarios of organoid-on-a-chip are finally prospected to further satisfy the clinical demands.
Collapse
Affiliation(s)
- Zhangjie Li
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qinyu Li
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, 999077 Hong Kong, China
| | - Chenyang Zhou
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Kangyi Lu
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yijun Liu
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Lian Xuan
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaolin Wang
- Author to whom correspondence should be addressed:
| |
Collapse
|
22
|
Krylov D, Rodimova S, Karabut M, Kuznetsova D. Experimental Models for Studying Structural and Functional State of the Pathological Liver (Review). Sovrem Tekhnologii Med 2023; 15:65-82. [PMID: 38434194 PMCID: PMC10902899 DOI: 10.17691/stm2023.15.4.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Indexed: 03/05/2024] Open
Abstract
Liver pathologies remain one of the leading causes of mortality worldwide. Despite a high prevalence of liver diseases, the possibilities of diagnosing, prognosing, and treating non-alcoholic and alcoholic liver diseases still have a number of limitations and require the development of new methods and approaches. In laboratory studies, various models are used to reconstitute the pathological conditions of the liver, including cell cultures, spheroids, organoids, microfluidic systems, tissue slices. We reviewed the most commonly used in vivo, in vitro, and ex vivo models for studying non-alcoholic fatty liver disease and alcoholic liver disease, toxic liver injury, and fibrosis, described their advantages, limitations, and prospects for use. Great emphasis was placed on the mechanisms of development of pathological conditions in each model, as well as the assessment of the possibility of reconstructing various key aspects of pathogenesis for all these pathologies. There is currently no consensus on the choice of the most adequate model for studying liver pathology. The choice of a certain effective research model is determined by the specific purpose and objectives of the experiment.
Collapse
Affiliation(s)
- D.P. Krylov
- Laboratory Assistant, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Student, Institute of Biology and Biomedicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| | - S.A. Rodimova
- Junior Researcher, Laboratory of Regenerative Medicine, Scientific Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - M.M. Karabut
- Researcher, Laboratory of Genomics of Adaptive Antitumor Immunity, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - D.S. Kuznetsova
- Head of Laboratory of Molecular Biotechnologies, Research Institute of Experimental Oncology and Biomedical Technologies; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia; Head of the Research Laboratory for Molecular Genetic Researches, Institute of Clinical Medicine; National Research Lobachevsky State University of Nizhny Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603022, Russia
| |
Collapse
|
23
|
Fang J, Celton-Morizur S, Desdouets C. NAFLD-Related HCC: Focus on the Latest Relevant Preclinical Models. Cancers (Basel) 2023; 15:3723. [PMID: 37509384 PMCID: PMC10377912 DOI: 10.3390/cancers15143723] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and one of the deadliest cancers worldwide. Despite extensive research, the biological mechanisms underlying HCC's development and progression remain only partially understood. Chronic overeating and/or sedentary-lifestyle-associated obesity, which promote Non-Alcoholic Fatty Liver Disease (NAFLD), have recently emerged as worrying risk factors for HCC. NAFLD is characterized by excessive hepatocellular lipid accumulation (steatosis) and affects one quarter of the world's population. Steatosis progresses in the more severe inflammatory form, Non-Alcoholic Steatohepatitis (NASH), potentially leading to HCC. The incidence of NASH is expected to increase by up to 56% over the next 10 years. Better diagnoses and the establishment of effective treatments for NAFLD and HCC will require improvements in our understanding of the fundamental mechanisms of the disease's development. This review describes the pathogenesis of NAFLD and the mechanisms underlying the transition from NAFL/NASH to HCC. We also discuss a selection of appropriate preclinical models of NAFLD for research, from cellular models such as liver-on-a-chip models to in vivo models, focusing particularly on mouse models of dietary NAFLD-HCC.
Collapse
Affiliation(s)
- Jing Fang
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France
- Genomic Instability, Metabolism, Immunity and Liver Tumorigenesis Laboratory, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France
| | - Séverine Celton-Morizur
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France
- Genomic Instability, Metabolism, Immunity and Liver Tumorigenesis Laboratory, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France
| | - Chantal Desdouets
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, 75006 Paris, France
- Genomic Instability, Metabolism, Immunity and Liver Tumorigenesis Laboratory, Equipe Labellisée Ligue Contre le Cancer, 75005 Paris, France
| |
Collapse
|
24
|
Wang H, Shen H, Seo W, Hwang S. Experimental models of fatty liver diseases: Status and appraisal. Hepatol Commun 2023; 7:e00200. [PMID: 37378635 DOI: 10.1097/hc9.0000000000000200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Fatty liver diseases, including alcohol-associated liver disease (ALD) and nonalcoholic fatty liver disease nonalcoholic fatty liver disease (NAFLD), affect a large number of people worldwide and become one of the major causes of end-stage liver disease, such as liver cirrhosis and hepatocellular carcinoma (HCC). Unfortunately, there are currently no approved pharmacological treatments for ALD or NAFLD. This situation highlights the urgent need to explore new intervention targets and discover effective therapeutics for ALD and NAFLD. The lack of properly validated preclinical disease models is a major obstacle to the development of clinical therapies. ALD and NAFLD models have been in the development for decades, but there are still no models that recapitulate the full spectrum of ALD and NAFLD. Throughout this review, we summarize the current in vitro and in vivo models used for research on fatty liver diseases and discuss the advantages and limitations of these models.
Collapse
Affiliation(s)
- Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Wonhyo Seo
- Laboratory of Hepatotoxicity, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
25
|
Otumala AE, Hellen DJ, Luna CA, Delgado P, Dissanayaka A, Ugwumadu C, Oshinowo O, Islam MM, Shen L, Karpen SJ, Myers DR. Opportunities and considerations for studying liver disease with microphysiological systems on a chip. LAB ON A CHIP 2023; 23:2877-2898. [PMID: 37282629 DOI: 10.1039/d2lc00940d] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Advances in microsystem engineering have enabled the development of highly controlled models of the liver that better recapitulate the unique in vivo biological conditions. In just a few short years, substantial progress has been made in creating complex mono- and multi-cellular models that mimic key metabolic, structural, and oxygen gradients crucial for liver function. Here we review: 1) the state-of-the-art in liver-centric microphysiological systems and 2) the array of liver diseases and pressing biological and therapeutic challenges which could be investigated with these systems. The engineering community has unique opportunities to innovate with new liver-on-a-chip devices and partner with biomedical researchers to usher in a new era of understanding of the molecular and cellular contributors to liver diseases and identify and test rational therapeutic modalities.
Collapse
Affiliation(s)
- Adiya E Otumala
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dominick J Hellen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - C Alessandra Luna
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Priscilla Delgado
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Anjana Dissanayaka
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Chidozie Ugwumadu
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Oluwamayokun Oshinowo
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Md Mydul Islam
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Luyao Shen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Saul J Karpen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - David R Myers
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, 1760 Haygood Dr, Suite E-160, Rm E-156, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
26
|
Aasadollahei N, Rezaei N, Golroo R, Agarwal T, Vosough M, Piryaei A. Bioengineering liver microtissues for modeling non-alcoholic fatty liver disease. EXCLI JOURNAL 2023; 22:367-391. [PMID: 37223084 PMCID: PMC10201011 DOI: 10.17179/excli2022-5892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/16/2023] [Indexed: 05/25/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the world's most common chronic liver disease. However, due to the lack of reliable in vitro NAFLD models, drug development studies have faced many limitations, and there is no food and drug administration-approved medicine for NAFLD treatment. A functional biomimetic in vitro human liver model requires an optimized natural microenvironment using appropriate cellular composition, to provide constructive cell-cell interactions, and niche-specific bio-molecules to supply crucial cues as cell-matrix interplay. Such a suitable liver model could employ appropriate and desired biochemical, mechanical, and physical properties similar to native tissue. Moreover, bioengineered three-dimensional tissues, specially microtissues and organoids, and more recently using infusion-based cultivation systems such as microfluidics can mimic natural tissue conditions and facilitate the exchange of nutrients and soluble factors to improve physiological function in the in vitro generated constructs. This review highlights the key players involved in NAFLD initiation and progression and discussed the available cells and matrices for in vitro NAFLD modeling. The strategies for optimizing the liver microenvironment to generate a powerful and biomimetic in vitro NAFLD model were described as well. Finally, the current challenges and future perospective for promotion in this subject were discussed.
Collapse
Affiliation(s)
- Negar Aasadollahei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Reihaneh Golroo
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tarun Agarwal
- Department of Bio-Technology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Rezvani M, Vallier L, Guillot A. Modeling Nonalcoholic Fatty Liver Disease in the Dish Using Human-Specific Platforms: Strategies and Limitations. Cell Mol Gastroenterol Hepatol 2023; 15:1135-1145. [PMID: 36740045 PMCID: PMC10031472 DOI: 10.1016/j.jcmgh.2023.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a chronic liver disease affecting multiple cell types of the human liver. The high prevalence of NAFLD and the lack of approved therapies increase the demand for reliable models for the preclinical discovery of drug targets. In the last decade, multiple proof-of-principle studies have demonstrated human-specific NAFLD modeling in the dish. These systems have included technologies based on human induced pluripotent stem cell derivatives, liver tissue section cultures, intrahepatic cholangiocyte organoids, and liver-on-a-chip. These platforms differ in functional maturity, multicellularity, scalability, and spatial organization. Identifying an appropriate model for a specific NAFLD-related research question is challenging. Therefore, we review different platforms for their strengths and limitations in modeling NAFLD. To define the fidelity of the current human in vitro NAFLD models in depth, we define disease hallmarks within the NAFLD spectrum that range from steatosis to severe fibroinflammatory tissue injury. We discuss how the most common methods are efficacious in modeling genetic contributions and aspects of the early NAFLD-related tissue response. We also highlight the shortcoming of current models to recapitulate the complexity of inter-organ crosstalk and the chronic process of liver fibrosis-to-cirrhosis that usually takes decades in patients. Importantly, we provide methodological overviews and discuss implementation hurdles (eg, reproducibility or costs) to help choose the most appropriate NAFLD model for the individual research focus: hepatocyte injury, ductular reaction, cellular crosstalk, or other applications. In sum, we highlight current strategies and deficiencies to model NAFLD in the dish and propose a framework for the next generation of human-specific investigations.
Collapse
Affiliation(s)
- Milad Rezvani
- Charité Universitätsmedizin Berlin, Department of Pediatric Gastroenterology, Nephrology and Metabolic Medicine, Berlin, Germany; Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Berlin Institute of Health, Center for Regenerative Therapies (BCRT), Berlin, Germany; Berlin Institute of Health, Clinician-Scientist Program, Berlin, Germany
| | - Ludovic Vallier
- Berlin Institute of Health, Center for Regenerative Therapies (BCRT), Berlin, Germany; Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Adrien Guillot
- Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, Department of Hepatology & Gastroenterology, Berlin, Germany.
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Di-Iacovo N, Pieroni S, Piobbico D, Castelli M, Scopetti D, Ferracchiato S, Della-Fazia MA, Servillo G. Liver Regeneration and Immunity: A Tale to Tell. Int J Mol Sci 2023; 24:1176. [PMID: 36674692 PMCID: PMC9864482 DOI: 10.3390/ijms24021176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/28/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
The physiological importance of the liver is demonstrated by its unique and essential ability to regenerate following extensive injuries affecting its function. By regenerating, the liver reacts to hepatic damage and thus enables homeostasis to be restored. The aim of this review is to add new findings that integrate the regenerative pathway to the current knowledge. An optimal regeneration is achieved through the integration of two main pathways: IL-6/JAK/STAT3, which promotes hepatocyte proliferation, and PI3K/PDK1/Akt, which in turn enhances cell growth. Proliferation and cell growth are events that must be balanced during the three phases of the regenerative process: initiation, proliferation and termination. Achieving the correct liver/body weight ratio is ensured by several pathways as extracellular matrix signalling, apoptosis through caspase-3 activation, and molecules including transforming growth factor-beta, and cyclic adenosine monophosphate. The actors involved in the regenerative process are numerous and many of them are also pivotal players in both the immune and non-immune inflammatory process, that is observed in the early stages of hepatic regeneration. Balance of Th17/Treg is important in liver inflammatory process outcomes. Knowledge of liver regeneration will allow a more detailed characterisation of the molecular mechanisms that are crucial in the interplay between proliferation and inflammation.
Collapse
Affiliation(s)
- Nicola Di-Iacovo
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Stefania Pieroni
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Danilo Piobbico
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Marilena Castelli
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Damiano Scopetti
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Simona Ferracchiato
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Maria Agnese Della-Fazia
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
| | - Giuseppe Servillo
- Department of Medicine and Surgery, University of Perugia, Piazzale L. Severi 1, 06129 Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale (C.U.R.Ge.F.), University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
31
|
Cavarzerani E, Caligiuri I, Bartoletti M, Canzonieri V, Rizzolio F. 3D dynamic cultures of HGSOC organoids to model innovative and standard therapies. Front Bioeng Biotechnol 2023; 11:1135374. [PMID: 37143603 PMCID: PMC10151532 DOI: 10.3389/fbioe.2023.1135374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/24/2023] [Indexed: 05/06/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) needs new technologies for improving cancer diagnosis and therapy. It is a fatal disease with few options for the patients. In this context, dynamic culture systems coupling with patient-derived cancer 3D microstructures could offer a new opportunity for exploring novel therapeutic approaches. In this study, we optimized a passive microfluidic platform with 3D cancer organoids, which allows a standardized approach among different patients, a minimum requirement of samples, multiple interrogations of biological events, and a rapid response. The passive flow was optimized to improve the growth of cancer organoids, avoiding the disruption of the extracellular matrix (ECM). Under optimized conditions of the OrganoFlow (tilting angle of 15° and an interval of rocking every 8 min), the cancer organoids grow faster than when they are in static conditions and the number of dead cells is reduced over time. To calculate the IC 50 values of standard chemotherapeutic drugs (carboplatin, paclitaxel, and doxorubicin) and targeted drugs (ATRA), different approaches were utilized. Resazurin staining, ATP-based assay, and DAPI/PI colocalization assays were compared, and the IC 50 values were calculated. The results showed that in the passive flow, the IC 50 values are lower than in static conditions. FITC-labeled paclitaxel shows a better penetration of ECM under passive flow than in static conditions, and cancer organoids start to die after 48 h instead of 96 h, respectively. Cancer organoids are the last frontiers for ex vivo testing of drugs that replicate the response of patients in the clinic. For this study, organoids derived from ascites or tissues of patients with Ovarian Cancer have been used. In conclusion, it was possible to develop a protocol for organoid cultures in a passive microfluidic platform with a higher growth rate, faster drug response, and better penetration of drugs into ECM, maintaining the samples' vitals and collecting the data on the same plate for up to 16 drugs.
Collapse
Affiliation(s)
- Enrico Cavarzerani
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, Aviano, Italy
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice, Italy
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, Aviano, Italy
| | - Michele Bartoletti
- Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano, Italy
| | - Vincenzo Canzonieri
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Flavio Rizzolio
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, Aviano, Italy
- Department of Molecular Sciences and Nanosystems, Ca’ Foscari University of Venice, Venice, Italy
- *Correspondence: Flavio Rizzolio,
| |
Collapse
|
32
|
Deguchi S, Takayama K. State-of-the-art liver disease research using liver-on-a-chip. Inflamm Regen 2022; 42:62. [PMID: 36494740 PMCID: PMC9733013 DOI: 10.1186/s41232-022-00248-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
To understand disease pathophysiologies, models that recapitulate human functions are necessary. In vitro models that consist of human cells are preferred to ones using animal cells, because organ functions can vary from species to species. However, conventional in vitro models do not recapitulate human organ functions well. Organ-on-a-chip technology provides a reliable in vitro model of the functional units of human organs. Organ-on-a-chip technology uses microfluidic devices and their accessories to impart organ functions to human cells. Using microfluidic devices, we can co-culture multiple cell types that compose human organs. Moreover, we can culture human cells under physiologically relevant stresses, such as mechanical and shear stresses. Current organ-on-a-chip technology can reproduce the functions of several organs including the liver. Because it is difficult to maintain the function of human hepatocytes, which are the gold standard of in vitro liver models, under conventional culture conditions, the application of liver-on-a-chips to liver disease research is expected. This review introduces the current status and future prospects of liver-on-a-chips in liver disease research.
Collapse
Affiliation(s)
- Sayaka Deguchi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
- Department of Medical Science, Graduate School of Medicine, Kyoto University, Kyoto, 606-8507 Japan
| | - Kazuo Takayama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507 Japan
- AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, 100-0004 Japan
| |
Collapse
|
33
|
Yang Z, Liu X, Cribbin EM, Kim AM, Li JJ, Yong KT. Liver-on-a-chip: Considerations, advances, and beyond. BIOMICROFLUIDICS 2022; 16:061502. [PMID: 36389273 PMCID: PMC9646254 DOI: 10.1063/5.0106855] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 10/25/2022] [Indexed: 05/14/2023]
Abstract
The liver is the largest internal organ in the human body with largest mass of glandular tissue. Modeling the liver has been challenging due to its variety of major functions, including processing nutrients and vitamins, detoxification, and regulating body metabolism. The intrinsic shortfalls of conventional two-dimensional (2D) cell culture methods for studying pharmacokinetics in parenchymal cells (hepatocytes) have contributed to suboptimal outcomes in clinical trials and drug development. This prompts the development of highly automated, biomimetic liver-on-a-chip (LOC) devices to simulate native liver structure and function, with the aid of recent progress in microfluidics. LOC offers a cost-effective and accurate model for pharmacokinetics, pharmacodynamics, and toxicity studies. This review provides a critical update on recent developments in designing LOCs and fabrication strategies. We highlight biomimetic design approaches for LOCs, including mimicking liver structure and function, and their diverse applications in areas such as drug screening, toxicity assessment, and real-time biosensing. We capture the newest ideas in the field to advance the field of LOCs and address current challenges.
Collapse
Affiliation(s)
| | | | - Elise M. Cribbin
- School of Biomedical Engineering, University of Technology Sydney, New South Wales 2007, Australia
| | - Alice M. Kim
- School of Biomedical Engineering, University of Technology Sydney, New South Wales 2007, Australia
| | - Jiao Jiao Li
- Authors to whom correspondence should be addressed: and
| | - Ken-Tye Yong
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
34
|
Park Y, Thadasina D, Bolujo I, Isidan A, Cross-Najafi AA, Lopez K, Li P, Dahlem AM, Kennedy L, Sato K, Francis H, Alpini G, Zhang W, Ekser B. Three-Dimensional Organoids as a Model to Study Nonalcoholic Fatty Liver Disease. Semin Liver Dis 2022; 42:423-433. [PMID: 36044928 PMCID: PMC11567686 DOI: 10.1055/a-1934-5588] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Despite the rising prevalence of nonalcoholic fatty liver disease (NAFLD), the underlying disease pathophysiology remains unclear. There is a great need for an efficient and reliable "human" in vitro model to study NAFLD and the progression to nonalcoholic steatohepatitis (NASH), which will soon become the leading indication for liver transplantation. Here, we review the recent developments in the use of three-dimensional (3D) liver organoids as a model to study NAFLD and NASH pathophysiology and possible treatments. Various techniques that are currently used to make liver organoids are discussed, such as the use of induced pluripotent stem cells versus primary cell lines and human versus murine cells. Moreover, methods for inducing lipid droplet accumulation and fibrosis to model NAFLD are explored. Finally, the limitations specific to the 3D organoid model for NAFLD/NASH are reviewed, highlighting the need for further development of multilineage models to include hepatic nonparenchymal cells and immune cells. The ultimate goal is to be able to accurately recapitulate the complex liver microenvironment in which NAFLD develops and progresses to NASH.
Collapse
Affiliation(s)
- Yujin Park
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Deepthi Thadasina
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ifeoluwa Bolujo
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Abdulkadir Isidan
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Arthur A. Cross-Najafi
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kevin Lopez
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ping Li
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Andrew M. Dahlem
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Lindsey Kennedy
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, and Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Keisaku Sato
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, and Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Heather Francis
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, and Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Gianfranco Alpini
- Department of Medicine, Division of Gastroenterology and Hepatology, Indiana University School of Medicine, and Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Wenjun Zhang
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| | - Burcin Ekser
- Department of Surgery, Division of Transplant Surgery, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
35
|
Shi H, Qiao F, Lu W, Huang K, Wen Y, Ye L, Chen Y. Baicalin improved hepatic injury of NASH by regulating NRF2/HO-1/NRLP3 pathway. Eur J Pharmacol 2022; 934:175270. [DOI: 10.1016/j.ejphar.2022.175270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/01/2022] [Accepted: 09/08/2022] [Indexed: 11/03/2022]
|
36
|
Wang SX, Yan JS, Chan YS. Advancements in MAFLD Modeling with Human Cell and Organoid Models. Int J Mol Sci 2022; 23:11850. [PMID: 36233151 PMCID: PMC9569457 DOI: 10.3390/ijms231911850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolic (dysfunction) associated fatty liver disease (MAFLD) is one of the most prevalent liver diseases and has no approved therapeutics. The high failure rates witnessed in late-phase MAFLD drug trials reflect the complexity of the disease, and how the disease develops and progresses remains to be fully understood. In vitro, human disease models play a pivotal role in mechanistic studies to unravel novel disease drivers and in drug testing studies to evaluate human-specific responses. This review focuses on MAFLD disease modeling using human cell and organoid models. The spectrum of patient-derived primary cells and immortalized cell lines employed to model various liver parenchymal and non-parenchymal cell types essential for MAFLD development and progression is discussed. Diverse forms of cell culture platforms utilized to recapitulate tissue-level pathophysiology in different stages of the disease are also reviewed.
Collapse
Affiliation(s)
- Shi-Xiang Wang
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
| | - Ji-Song Yan
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yun-Shen Chan
- Guangzhou Laboratory, No. 9 Xing Dao Huan Bei Road, Guangzhou International Bio Island, Guangzhou 510005, China
| |
Collapse
|
37
|
Dalsbecker P, Beck Adiels C, Goksör M. Liver-on-a-chip devices: the pros and cons of complexity. Am J Physiol Gastrointest Liver Physiol 2022; 323:G188-G204. [PMID: 35819853 DOI: 10.1152/ajpgi.00346.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Physiologically relevant and broadly applicable liver cell culture platforms are of great importance in both drug development and disease modeling. Organ-on-a-chip systems offer a promising alternative to conventional, static two-dimensional (2-D) cultures, providing much-needed cues such as perfusion, shear stress, and three-dimensional (3-D) cell-cell communication. However, such devices cover a broad range of complexity both in manufacture and in implementation. In this review, we summarize the key features of the human liver that should be reflected in a physiologically relevant liver-on-a-chip model. We also discuss different material properties of importance in producing liver-on-a-chip devices and summarize recent and current progress in the field, highlighting different types of devices at different levels of complexity.
Collapse
Affiliation(s)
| | | | - Mattias Goksör
- Department of Physics, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
38
|
Xiao J, Li X, Zhou Z, Guan S, Zhuo L, Gao B. Development of an in vitro insulin resistance dissociated model of hepatic steatosis by co-culture system. Biosci Trends 2022; 16:257-266. [PMID: 35965099 DOI: 10.5582/bst.2022.01242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The evidence shows that there is an associated relationship between hepatosteatosis and insulin resistance. While some existing genetic induction animal and patient models challenge this relationship, indicating that hepatosteatosis is dissociated from insulin resistance. However, the molecular mechanisms of this dissociation remain poorly understood due to a lack of available, reliable, and simplistic setup models. Currently, we used primary rat hepatocytes (rHPCs), co-cultured with rat hepatic stellate cells (HSC-T6) or human foreskin fibroblast cells (HFF-1) in stimulation with high insulin and glucose, to develop a model of steatosis charactered as dissociated lipid accumulation from insulin resistance. Oil-Red staining significantly showed intracellular lipid accumulated in the developed model. Gene expression of sterol regulatory element-binding protein 1c (SREBP1c) and elongase of very-long-chain fatty acids 6 (ELOVL6), key genes responsible for lipogenesis, were detected and obviously increased in this model. Inversely, the insulin resistance related genes expression included phosphoenolpyruvate carboxykinase 1 (PCK1), pyruvate dehydrogenase lipoamide kinase isozyme 4 (PDK4), and glucose-6-phosphatase (G6pase) were decreased, suggesting a dissociation relationship between steatosis and insulin resistance in the developed model. As well, the drug metabolism of this developed model was investigated and showed up-regulation of cytochrome P450 3A (CYP3A) and down-regulation of cytochrome P450 2E1 (CYP2E1) and cytochrome P450 1A2 (CYP1A2). Taken together, those results demonstrate that the in vitro model of dissociated steatosis from insulin resistance was successfully created by our co-cultured cells in high insulin and glucose medium, which will be a potential model for investigating the mechanism of insulin resistance dissociated steatosis, and discovering a novel drug for its treatment.
Collapse
Affiliation(s)
- Jiangwei Xiao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Xiang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zongbao Zhou
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Shuwen Guan
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| | - Lingjian Zhuo
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, China.,National Engineering Research Center for Healthcare Devices, Guangzhou, China.,Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou, China
| |
Collapse
|
39
|
Nahak BK, Mishra A, Preetam S, Tiwari A. Advances in Organ-on-a-Chip Materials and Devices. ACS APPLIED BIO MATERIALS 2022; 5:3576-3607. [PMID: 35839513 DOI: 10.1021/acsabm.2c00041] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The organ-on-a-chip (OoC) paves a way for biomedical applications ranging from preclinical to clinical translational precision. The current trends in the in vitro modeling is to reduce the complexity of human organ anatomy to the fundamental cellular microanatomy as an alternative of recreating the entire cell milieu that allows systematic analysis of medicinal absorption of compounds, metabolism, and mechanistic investigation. The OoC devices accurately represent human physiology in vitro; however, it is vital to choose the correct chip materials. The potential chip materials include inorganic, elastomeric, thermoplastic, natural, and hybrid materials. Despite the fact that polydimethylsiloxane is the most commonly utilized polymer for OoC and microphysiological systems, substitute materials have been continuously developed for its advanced applications. The evaluation of human physiological status can help to demonstrate using noninvasive OoC materials in real-time procedures. Therefore, this Review examines the materials used for fabricating OoC devices, the application-oriented pros and cons, possessions for device fabrication and biocompatibility, as well as their potential for downstream biochemical surface alteration and commercialization. The convergence of emerging approaches, such as advanced materials, artificial intelligence, machine learning, three-dimensional (3D) bioprinting, and genomics, have the potential to perform OoC technology at next generation. Thus, OoC technologies provide easy and precise methodologies in cost-effective clinical monitoring and treatment using standardized protocols, at even personalized levels. Because of the inherent utilization of the integrated materials, employing the OoC with biomedical approaches will be a promising methodology in the healthcare industry.
Collapse
Affiliation(s)
- Bishal Kumar Nahak
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Anshuman Mishra
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Subham Preetam
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| | - Ashutosh Tiwari
- Institute of Advanced Materials, IAAM, Gammalkilsvägen 18, Ulrika 59053, Sweden
| |
Collapse
|
40
|
Redaelli A, Long M. Bioengineering of the liver. APL Bioeng 2022; 6:020401. [DOI: 10.1063/5.0087886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano 20133, Italy
| | - Mian Long
- Institute of Mechanics, Chinese Academy of Sciences, Beijing 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
41
|
A Soft Zwitterionic Hydrogel as Potential Coating on a Polyimide Surface to Reduce Foreign Body Reaction to Intraneural Electrodes. Molecules 2022; 27:molecules27103126. [PMID: 35630604 PMCID: PMC9147366 DOI: 10.3390/molecules27103126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 12/04/2022] Open
Abstract
Invasive intraneural electrodes can control advanced neural-interfaced prostheses in human amputees. Nevertheless, in chronic implants, the progressive formation of a fibrotic capsule can gradually isolate the electrode surface from the surrounding tissue leading to loss of functionality. This is due to a nonspecific inflammatory response called foreign-body reaction (FBR). The commonly used poly(ethylene glycol) (PEG)-based low-fouling coatings of implantable devices can be easily encapsulated and are susceptible to oxidative damage in long-term in vivo applications. Recently, sulfobetaine-based zwitterionic hydrogels have emerged as an important class of robust ultra-low fouling biomaterials, holding great potential to mitigate FBR. The aim of this proof-of-principle in vitro work was to assess whether the organic zwitterionic—poly(sulfobetaine methacrylate) [poly(SBMA)]—hydrogel could be a suitable coating for Polyimide (PI)-based intraneural electrodes to reduce FBR. We first synthesized and analyzed the hydrogel through a mechanical characterization (i.e., Young’s modulus). Then, we demonstrated reduced adhesion and activation of fibrogenic and pro-inflammatory cells (i.e., human myofibroblasts and macrophages) on the hydrogel compared with PEG-coated and polystyrene surfaces using cell viability assays, confocal fluorescence microscopy and high-content analysis of oxidative stress production. Interestingly, we successfully coated PI surfaces with a thin film of the hydrogel through covalent bond and demonstrated its high hydrophilicity via water contact angle measurement. Importantly, we showed the long-term release of an anti-fibrotic drug (i.e., Everolimus) from the hydrogel. Because of the low stiffness, biocompatibility, high hydration and ultra-low fouling characteristics, our zwitterionic hydrogel could be envisioned as long-term diffusion-based delivery system for slow and controlled anti-inflammatory and anti-fibrotic drug release in vivo.
Collapse
|
42
|
Tuerxun K, He J, Ibrahim I, Yusupu Z, Yasheng A, Xu Q, Tang R, Aikebaier A, Wu Y, Tuerdi M, Nijiati M, Zou X, Xu T. Bioartificial livers: a review of their design and manufacture. Biofabrication 2022; 14. [PMID: 35545058 DOI: 10.1088/1758-5090/ac6e86] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Acute liver failure (ALF) is a rapidly progressive disease with high morbidity and mortality rates. Liver transplantation and artificial liver support systems, such as artificial livers (ALs) and bioartificial livers (BALs), are the two major therapies for ALF. Compared to ALs, BALs are composed of functional hepatocytes that provide essential liver functions, including detoxification, metabolite synthesis, and biotransformation. Furthermore, BALs can potentially provide effective support as a form of bridging therapy to liver transplantation or spontaneous recovery for patients with ALF. In this review, we systematically discussed the currently available state-of-the-art designs and manufacturing processes for BAL support systems. Specifically, we classified the cell sources and bioreactors that are applied in BALs, highlighted the advanced technologies of hepatocyte culturing and bioreactor fabrication, and discussed the current challenges and future trends in developing next generation BALs for large scale clinical applications.
Collapse
Affiliation(s)
- Kahaer Tuerxun
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Jianyu He
- Department of Mechanical Engineering, Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing, Beijing, 100084, CHINA
| | - Irxat Ibrahim
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Zainuer Yusupu
- Department of Ultrasound, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Abudoukeyimu Yasheng
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Qilin Xu
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Ronghua Tang
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Aizemaiti Aikebaier
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Yuanquan Wu
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Maimaitituerxun Tuerdi
- Department of hepatobiliary and pancreatic surgery, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Mayidili Nijiati
- Medical imaging center, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, China, Kashi, Xinjiang, 844000, CHINA
| | - Xiaoguang Zou
- Hospital Organ, First People's Hospital of Kashi, 120th, Yingbin Road, Kashi, Xinjiang, 844000, CHINA
| | - Tao Xu
- Tsinghua University, 30 Shuangqing Road, Haidian District, Beijing, 100084, CHINA
| |
Collapse
|
43
|
An Agent-Based Interpretation of Leukocyte Chemotaxis in Cancer-on-Chip Experiments. MATHEMATICS 2022. [DOI: 10.3390/math10081338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The present paper was inspired by recent developments in laboratory experiments within the framework of cancer-on-chip technology, an immune-oncology microfluidic chip aiming at studying the fundamental mechanisms of immunocompetent behavior. We focus on the laboratory setting where cancer is treated with chemotherapy drugs, and in this case, the effects of the treatment administration hypothesized by biologists are: the absence of migration and proliferation of tumor cells, which are dying; the stimulation of the production of chemical substances (annexin); the migration of leukocytes in the direction of higher concentrations of chemicals. Here, following the physiological hypotheses made by biologists on the phenomena occurring in these experiments, we introduce an agent-based model reproducing the dynamics of two cell populations (agents), i.e., tumor cells and leukocytes living in the microfluidic chip environment. Our model aims at proof of concept, demonstrating that the observations of the biological phenomena can be obtained by the model on the basis of the explicit assumptions made. In this framework, close adherence of the computational model to the biological results, as shown in the section devoted to the first calibration of the model with respect to available observations, is successfully accomplished.
Collapse
|
44
|
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
|
45
|
Cultivating human tissues and organs over lab-on-a-chip models: Recent progress and applications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:205-240. [PMID: 35094775 DOI: 10.1016/bs.pmbts.2021.07.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In vivo models are indispensable for preclinical studies for various human disease modeling and drug screening, however, face several obstacles such as animal model species differences and ethical clearance. Additionally, it is difficult to accurately predict the organ interaction, drug efficacy, and toxicity using conventional in vitro two-dimensional (2D) cell culture models. The microfluidic-based systems provide excellent opportunity to recapitulate the human organ/tissue functions under in vitro conditions. The organ/tissue-on-chip models are one of best emerging technologies that offer functional organs/tissues on a microfluidic chip. This technology has potential to noninvasively study the organ physiology, tissue development, and diseases etymology. This chapter comprises the benifits of 2D and three-dimensional (3D) in vitro cultures as well as highlights the importance of microfluidic-based lab-on-a-chip technique. The development of different organs/tissues-on-chip models and their biomedical application in various diseases such as cardiovascular diseases, neurodegenerative diseases, respiratory-based diseases, cancers, liver and kidney diseases, etc., have also been discussed.
Collapse
|
46
|
Ramos MJ, Bandiera L, Menolascina F, Fallowfield JA. In vitro models for non-alcoholic fatty liver disease: Emerging platforms and their applications. iScience 2022; 25:103549. [PMID: 34977507 PMCID: PMC8689151 DOI: 10.1016/j.isci.2021.103549] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global healthcare challenge, affecting 1 in 4 adults, and death rates are predicted to rise inexorably. The progressive form of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. However, no medical treatments are licensed for NAFLD-NASH. Identifying efficacious therapies has been hindered by the complexity of disease pathogenesis, a paucity of predictive preclinical models and inadequate validation of pharmacological targets in humans. The development of clinically relevant in vitro models of the disease will pave the way to overcome these challenges. Currently, the combined application of emerging technologies (e.g., organ-on-a-chip/microphysiological systems) and control engineering approaches promises to unravel NAFLD biology and deliver tractable treatment candidates. In this review, we will describe advances in preclinical models for NAFLD-NASH, the recent introduction of novel technologies in this space, and their importance for drug discovery endeavors in the future.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Lucia Bandiera
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK.,Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Filippo Menolascina
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK.,Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Jonathan Andrew Fallowfield
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| |
Collapse
|
47
|
Ramos MJ, Bandiera L, Menolascina F, Fallowfield JA. In vitro models for non-alcoholic fatty liver disease: Emerging platforms and their applications. iScience 2022; 25:103549. [PMID: 34977507 DOI: 10.1016/j.isci] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) represents a global healthcare challenge, affecting 1 in 4 adults, and death rates are predicted to rise inexorably. The progressive form of NAFLD, non-alcoholic steatohepatitis (NASH), can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. However, no medical treatments are licensed for NAFLD-NASH. Identifying efficacious therapies has been hindered by the complexity of disease pathogenesis, a paucity of predictive preclinical models and inadequate validation of pharmacological targets in humans. The development of clinically relevant in vitro models of the disease will pave the way to overcome these challenges. Currently, the combined application of emerging technologies (e.g., organ-on-a-chip/microphysiological systems) and control engineering approaches promises to unravel NAFLD biology and deliver tractable treatment candidates. In this review, we will describe advances in preclinical models for NAFLD-NASH, the recent introduction of novel technologies in this space, and their importance for drug discovery endeavors in the future.
Collapse
Affiliation(s)
- Maria Jimenez Ramos
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Lucia Bandiera
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK
- Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Filippo Menolascina
- Institute for Bioengineering, The University of Edinburgh, Edinburgh EH9 3BF, UK
- Synthsys - Centre for Synthetic and Systems Biology, The University of Edinburgh, Edinburgh EH9 3BF, UK
| | - Jonathan Andrew Fallowfield
- Centre for Inflammation Research, The University of Edinburgh, The Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| |
Collapse
|
48
|
Kirk LM, Waits CMK, Bashore AC, Dosso B, Meyers AK, Renaldo AC, DePalma TJ, Simms KN, Hauser N, Chuang Key CC, McCall CE, Parks JS, Sergeant S, Langefeld CD, Skardal A, Rahbar E. Exploiting three-dimensional human hepatic constructs to investigate the impact of rs174537 on fatty acid metabolism. PLoS One 2022; 17:e0262173. [PMID: 35051193 PMCID: PMC8775235 DOI: 10.1371/journal.pone.0262173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
The Modern Western Diet has been associated with the rise in metabolic and inflammatory diseases, including obesity, diabetes, and cardiovascular disease. This has been attributed, in part, to the increase in dietary omega-6 polyunsaturated fatty acid (PUFA) consumption, specifically linoleic acid (LA), arachidonic acid (ARA), and their subsequent metabolism to pro-inflammatory metabolites which may be driving human disease. Conversion of dietary LA to ARA is regulated by genetic variants near and within the fatty acid desaturase (FADS) haplotype block, most notably single nucleotide polymorphism rs174537 is strongly associated with FADS1 activity and expression. This variant and others within high linkage disequilibrium may potentially explain the diversity in both diet and inflammatory mediators that drive chronic inflammatory disease in human populations. Mechanistic exploration into this phenomenon using human hepatocytes is limited by current two-dimensional culture models that poorly replicate in vivo functionality. Therefore, we aimed to develop and characterize a three-dimensional hepatic construct for the study of human PUFA metabolism. Primary human hepatocytes cultured in 3D hydrogels were characterized for their capacity to represent basic lipid processing functions, including lipid esterification, de novo lipogenesis, and cholesterol efflux. They were then exposed to control and LA-enriched media and reproducibly displayed allele-specific metabolic activity of FADS1, based on genotype at rs174537. Hepatocytes derived from individuals homozygous with the minor allele at rs174537 (i.e., TT) displayed the slowest metabolic conversion of LA to ARA and significantly reduced FADS1 and FADS2 expression. These results support the feasibility of using 3D human hepatic cultures for the study of human PUFA and lipid metabolism and relevant gene-diet interactions, thereby enabling future nutrition targets in humans.
Collapse
Affiliation(s)
- L. Madison Kirk
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Virginia Tech – Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, Virginia, United States of America
| | - Charlotte Mae K. Waits
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Virginia Tech – Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, Virginia, United States of America
| | - Alexander C. Bashore
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Beverly Dosso
- Department of Integrative Physiology & Pharmacology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Allison K. Meyers
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Antonio C. Renaldo
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Virginia Tech – Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, Virginia, United States of America
| | - Thomas J. DePalma
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Kelli N. Simms
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Virginia Tech – Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, Virginia, United States of America
| | - Nathaniel Hauser
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Virginia Tech – Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, Virginia, United States of America
| | - Chia-Chi Chuang Key
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Charles E. McCall
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - John S. Parks
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Susan Sergeant
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Carl D. Langefeld
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Aleksander Skardal
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States of America
| | - Elaheh Rahbar
- Department of Biomedical Engineering, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
- Virginia Tech – Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, Virginia, United States of America
- Center for Precision Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| |
Collapse
|
49
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
50
|
Wang AJ, Allen A, Sofman M, Sphabmixay P, Yildiz E, Griffith LG. Engineering Modular 3D Liver Culture Microenvironments In Vitro to Parse the Interplay between Biophysical and Biochemical Microenvironment Cues on Hepatic Phenotypes. ADVANCED NANOBIOMED RESEARCH 2022; 2:2100049. [PMID: 35872804 PMCID: PMC9307216 DOI: 10.1002/anbr.202100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
In vitro models of human liver functions are used across a diverse range of applications in preclinical drug development and disease modeling, with particular increasing interest in models that capture facets of liver inflammatory status. This study investigates how the interplay between biophysical and biochemical microenvironment cues influence phenotypic responses, including inflammation signatures, of primary human hepatocytes (PHH) cultured in a commercially available perfused bioreactor. A 3D printing-based alginate microwell system was designed to form thousands of hepatic spheroids in a scalable manner as a comparator 3D culture modality to the bioreactor. Soft, synthetic extracellular matrix (ECM) hydrogel scaffolds with biophysical properties mimicking features of liver were engineered to replace polystyrene scaffolds, and the biochemical microenvironment was modulated with a defined set of growth factors and signaling modulators. The supplemented media significantly increased tissue density, albumin secretion, and CYP3A4 activity but also upregulated inflammatory markers. Basal inflammatory markers were lower for cells maintained in ECM hydrogel scaffolds or spheroid formats than polystyrene scaffolds, while hydrogel scaffolds exhibited the most sensitive response to inflammation as assessed by multiplexed cytokine and RNA-seq analyses. Together, these engineered 3D liver microenvironments provide insights for probing human liver functions and inflammatory response in vitro.
Collapse
Affiliation(s)
- Alex J Wang
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Allysa Allen
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Marianna Sofman
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Pierre Sphabmixay
- Mechanical Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Whitehead Institute for Biomedical Research, 455 Main Street, Cambridge, MA, 02142, USA
| | - Ece Yildiz
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Institute of Bioengineering, School of Life Science, École Polytechnique Fédérale de Lausanne, Route Cantonale, 1015 Lausanne, Switzerland
| | - Linda G. Griffith
- Biological Engineering Department, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Center for Gynepathology Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| |
Collapse
|