1
|
Fu J, Qiu H, Tan CS. Microfluidic Liver-on-a-Chip for Preclinical Drug Discovery. Pharmaceutics 2023; 15:pharmaceutics15041300. [PMID: 37111785 PMCID: PMC10141038 DOI: 10.3390/pharmaceutics15041300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Drug discovery is an expensive, long, and complex process, usually with a high degree of uncertainty. In order to improve the efficiency of drug development, effective methods are demanded to screen lead molecules and eliminate toxic compounds in the preclinical pipeline. Drug metabolism is crucial in determining the efficacy and potential side effects, mainly in the liver. Recently, the liver-on-a-chip (LoC) platform based on microfluidic technology has attracted widespread attention. LoC systems can be applied to predict drug metabolism and hepatotoxicity or to investigate PK/PD (pharmacokinetics/pharmacodynamics) performance when combined with other artificial organ-on-chips. This review discusses the liver physiological microenvironment simulated by LoC, especially the cell compositions and roles. We summarize the current methods of constructing LoC and the pharmacological and toxicological application of LoC in preclinical research. In conclusion, we also discussed the limitations of LoC in drug discovery and proposed a direction for improvement, which may provide an agenda for further research.
Collapse
Affiliation(s)
- Jingyu Fu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Hailong Qiu
- Tianjin Key Laboratory of Functional Crystal Materials, Institute of Functional Crystal, Tianjin University of Technology, Tianjin 300384, China
| | - Cherie S Tan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
2
|
Mirdamadi ES, Khosrowpour Z, Jafari D, Gholipourmalekabadi M, Solati-Hashjin M. 3D-printed PLA/Gel hybrid in liver tissue engineering: Effects of architecture on biological functions. Biotechnol Bioeng 2023; 120:836-851. [PMID: 36479982 DOI: 10.1002/bit.28301] [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: 09/22/2022] [Revised: 11/28/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
The liver is one of the vital organs in the body, and the gold standard of treatment for liver function impairment is liver transplantation, which poses many challenges. The specific three-dimensional (3D) structure of liver, which significantly impacts the growth and function of its cells, has made biofabrication with the 3D printing of scaffolds suitable for this approach. In this study, to investigate the effect of scaffold geometry on the performance of HepG2 cells, poly-lactic acid (PLA) polymer was used as the input of the fused deposition modeling (FDM) 3D-printing machine. Samples with simple square and bioinspired hexagonal cross-sectional designs were printed. One percent and 2% of gelatin coating were applied to the 3D printed PLA to improve the wettability and surface properties of the scaffold. Scanning electron microscopy pictures were used to analyze the structural properties of PLA-Gel hybrid scaffolds, energy dispersive spectroscopy to investigate the presence of gelatin, water contact angle measurement for wettability, and weight loss for degradation. In vitro tests were performed by culturing HepG2 cells on the scaffold to evaluate the cell adhesion, viability, cytotoxicity, and specific liver functions. Then, high-precision scaffolds were printed and the presence of gelatin was detected. Also, the effect of geometry on cell function was confirmed in viability, adhesion, and functional tests. The albumin and urea production of the Hexagonal PLA scaffold was about 1.22 ± 0.02-fold higher than the square design in 3 days. This study will hopefully advance our understanding of liver tissue engineering toward a promising perspective for liver regeneration.
Collapse
Affiliation(s)
- Elnaz Sadat Mirdamadi
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Zahra Khosrowpour
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
3
|
Kibar G, Dutta S, Rege K, Usta OB. Evaluation of drug carrier hepatotoxicity using primary cell culture models. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102651. [PMID: 36623713 PMCID: PMC10492629 DOI: 10.1016/j.nano.2023.102651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/17/2022] [Accepted: 12/25/2022] [Indexed: 01/09/2023]
Abstract
This study aims to establish a primary rat hepatocyte culture model to evaluate dose-dependent hepatotoxic effects of drug carriers (lipopolymer nanoparticles; LPNs) temporal. Primary rat hepatocyte cell cultures were used to determine half-maximal Inhibition Concentrations (IC50) of the drug-carrier library. Drug-carrier library, at concentrations <50 μg/mL, is benign to primary rat hepatocytes as determined using albumin and urea secretions. Albumin, as a hepatic biomarker, exhibited a more sensitive and faster outcome, compared to urea, for the determination of the IC50 value of LPNs. Temporal measurements of hepatic biomarkers including urea and albumin, and rigorous physicochemical (hydrodynamic diameter, surface charge, etc.) characterization, should be combined to evaluate the hepatotoxicity of drug carrier libraries in screens.
Collapse
Affiliation(s)
- Güneş Kibar
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Shriners Hospitals for Children, Boston, MA 02114, USA; Department of Materials Science and Engineering, Adana Alparslan Turkes Science and Technology University, Adana 01250, Turkey
| | - Subhadeep Dutta
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Kaushal Rege
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287, USA; Chemical Engineering, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85287, USA; Biological Design Graduate Program, School for Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85287, USA.
| | - O Berk Usta
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Shriners Hospitals for Children, Boston, MA 02114, USA.
| |
Collapse
|
4
|
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
|
5
|
Diprospero TJ, Brown LG, Fachko TD, Lockett MR. HepaRG cells undergo increased levels of post-differentiation patterning in physiologic conditions when maintained as 3D cultures in paper-based scaffolds. RESEARCH SQUARE 2023:rs.3.rs-2473387. [PMID: 36711963 PMCID: PMC9882668 DOI: 10.21203/rs.3.rs-2473387/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Monolayer cultures of hepatocytes lack many aspects of the liver sinusoid, including a tissue-level organization that results from extracellular matrix interactions and gradients of soluble molecules that span from the portal triad to the central vein. We measured the activity and transcript levels of drug-metabolizing enzymes in HepaRG cells maintained in three different culture configurations: as monolayers, seeded onto paper scaffolds that were pre-loaded with a collagen matrix, and when seeded directly into the paper scaffolds as a cell-laden gel. Drug metabolism was significantly decreased in the presence of the paper scaffolds compared to monolayer configurations when cells were exposed to standard culture conditions. Despite this decreased function, transcript levels suggest the cells undergo increased polarization and adopt a biliary-like character in the paper scaffolds, including the increased expression of transporter proteins (e.g., ABCB11 and SLOC1B1) and the KRT19 cholangiocyte marker. When exposed to representative periportal or perivenous culture conditions, we observed in vivo zonal-like patterns, including increased cytochrome P450 (CYP) activity and transcript levels in the perivenous condition. This increased CYP activity is more pronounced in the laden configuration, supporting the need to include multiple aspects of the liver microenvironment to observe the post-differentiation processing of hepatocytes.
Collapse
|
6
|
Valdiviezo A, Kato Y, Baker ES, Chiu WA, Rusyn I. Evaluation of Metabolism of a Defined Pesticide Mixture through Multiple In Vitro Liver Models. TOXICS 2022; 10:566. [PMID: 36287846 PMCID: PMC9609317 DOI: 10.3390/toxics10100566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 06/16/2023]
Abstract
The evaluation of exposure to multiple contaminants in a mixture presents a number of challenges. For example, the characterization of chemical metabolism in a mixture setting remains a research area with critical knowledge gaps. Studies of chemical metabolism typically utilize suspension cultures of primary human hepatocytes; however, this model is not suitable for studies of more extended exposures and donor-to-donor variability in a metabolic capacity is unavoidable. To address this issue, we utilized several in vitro models based on human-induced pluripotent stem cell (iPSC)-derived hepatocytes (iHep) to characterize the metabolism of an equimolar (1 or 5 µM) mixture of 20 pesticides. We used iHep suspensions and 2D sandwich cultures, and a microphysiological system OrganoPlate® 2-lane 96 (MimetasTM) that also included endothelial cells and THP-1 cell-derived macrophages. When cell culture media were evaluated using gas and liquid chromatography coupled to tandem mass spectrometry methods, we found that the parent molecule concentrations diminished, consistent with metabolic activity. This effect was most pronounced in iHep suspensions with a 1 µM mixture, and was lowest in OrganoPlate® 2-lane 96 for both mixtures. Additionally, we used ion mobility spectrometry-mass spectrometry (IMS-MS) to screen for metabolite formation in these cultures. These analyses revealed the presence of five primary metabolites that allowed for a more comprehensive evaluation of chemical metabolism in vitro. These findings suggest that iHep-based suspension assays maintain higher metabolic activity compared to 2D sandwich and OrganoPlate® 2-lane 96 model. Moreover, this study illustrates that IMS-MS can characterize in vitro metabolite formation following exposure to mixtures of environmental contaminants.
Collapse
Affiliation(s)
- Alan Valdiviezo
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Yuki Kato
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Laboratory for Drug Discovery and Development, Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Erin S. Baker
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Weihsueh A. Chiu
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
7
|
Messelmani T, Morisseau L, Sakai Y, Legallais C, Le Goff A, Leclerc E, Jellali R. Liver organ-on-chip models for toxicity studies and risk assessment. LAB ON A CHIP 2022; 22:2423-2450. [PMID: 35694831 DOI: 10.1039/d2lc00307d] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The liver is a key organ that plays a pivotal role in metabolism and ensures a variety of functions in the body, including homeostasis, synthesis of essential components, nutrient storage, and detoxification. As the centre of metabolism for exogenous molecules, the liver is continuously exposed to a wide range of compounds, such as drugs, pesticides, and environmental pollutants. Most of these compounds can cause hepatotoxicity and lead to severe and irreversible liver damage. To study the effects of chemicals and drugs on the liver, most commonly, animal models or in vitro 2D cell cultures are used. However, data obtained from animal models lose their relevance when extrapolated to the human metabolic situation and pose ethical concerns, while 2D static cultures are poorly predictive of human in vivo metabolism and toxicity. As a result, there is a widespread need to develop relevant in vitro liver models for toxicology studies. In recent years, progress in tissue engineering, biomaterials, microfabrication, and cell biology has created opportunities for more relevant in vitro models for toxicology studies. Of these models, the liver organ-on-chip (OoC) has shown promising results by reproducing the in vivo behaviour of the cell/organ or a group of organs, the controlled physiological micro-environment, and in vivo cellular metabolic responses. In this review, we discuss the development of liver organ-on-chip technology and its use in toxicity studies. First, we introduce the physiology of the liver and summarize the traditional experimental models for toxicity studies. We then present liver OoC technology, including the general concept, materials used, cell sources, and different approaches. We review the prominent liver OoC and multi-OoC integrating the liver for drug and chemical toxicity studies. Finally, we conclude with the future challenges and directions for developing or improving liver OoC models.
Collapse
Affiliation(s)
- Taha Messelmani
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 Compiègne Cedex, France.
| | - Lisa Morisseau
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 Compiègne Cedex, France.
| | - Yasuyuki Sakai
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan.
- Department of Chemical Engineering, Faculty of Engineering, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | - Cécile Legallais
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 Compiègne Cedex, France.
| | - Anne Le Goff
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 Compiègne Cedex, France.
| | - Eric Leclerc
- CNRS IRL 2820, Laboratory for Integrated Micro Mechatronic Systems, Institute of Industrial Science, University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan.
| | - Rachid Jellali
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu CS 60319, 60203 Compiègne Cedex, France.
| |
Collapse
|
8
|
Exploring a Computational Method for Evaluating the Epinecidin-1 and Its Variants Binding Efficacy with Breast Cancer Receptor (HER-2). Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10424-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
9
|
Telles-Silva KA, Pacheco L, Komatsu S, Chianca F, Caires-Júnior LC, Araujo BHS, Goulart E, Zatz M. Applied Hepatic Bioengineering: Modeling the Human Liver Using Organoid and Liver-on-a-Chip Technologies. Front Bioeng Biotechnol 2022; 10:845360. [PMID: 35237587 PMCID: PMC8882846 DOI: 10.3389/fbioe.2022.845360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/19/2022] [Indexed: 12/19/2022] Open
Abstract
The liver is the most important metabolic hub of endo and xenobiotic compounds. Pre-clinical studies using rodents to evaluate the toxicity of new drugs and cosmetics may produce inconclusive results for predicting clinical outcomes in humans, moreover being banned in the European Union. Human liver modeling using primary hepatocytes presents low reproducibility due to batch-to-batch variability, while iPSC-derived hepatocytes in monolayer cultures (2D) show reduced cellular functionality. Here we review the current status of the two most robust in vitro approaches in improving hepatocyte phenotype and metabolism while mimicking the hepatic physiological microenvironment: organoids and liver-on-chip. Both technologies are reviewed in design and manufacturing techniques, following cellular composition and functionality. Furthermore, drug screening and liver diseases modeling efficiencies are summarized. Finally, organoid and liver-on-chip technologies are compared regarding advantages and limitations, aiming to guide the selection of appropriate models for translational research and the development of such technologies.
Collapse
Affiliation(s)
- Kayque Alves Telles-Silva
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Lara Pacheco
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Sabrina Komatsu
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Fernanda Chianca
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Luiz Carlos Caires-Júnior
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
| | - Bruno Henrique Silva Araujo
- Lieber Institute for Brain Development, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ernesto Goulart
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
- *Correspondence: Ernesto Goulart, ; Mayana Zatz,
| | - Mayana Zatz
- Human Genome and Stem-Cell Research Center (HUG-CEL), Institute of Biosciences, University of Sao Paulo (USP), Sao Paulo, Brazil
- *Correspondence: Ernesto Goulart, ; Mayana Zatz,
| |
Collapse
|
10
|
Panwar A, Das P, Tan LP. 3D Hepatic Organoid-Based Advancements in LIVER Tissue Engineering. Bioengineering (Basel) 2021; 8:185. [PMID: 34821751 PMCID: PMC8615121 DOI: 10.3390/bioengineering8110185] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Liver-associated diseases and tissue engineering approaches based on in vitro culture of functional Primary human hepatocytes (PHH) had been restricted by the rapid de-differentiation in 2D culture conditions which restricted their usability. It was proven that cells growing in 3D format can better mimic the in vivo microenvironment, and thus help in maintaining metabolic activity, phenotypic properties, and longevity of the in vitro cultures. Again, the culture method and type of cell population are also recognized as important parameters for functional maintenance of primary hepatocytes. Hepatic organoids formed by self-assembly of hepatic cells are microtissues, and were able to show long-term in vitro maintenance of hepato-specific characteristics. Thus, hepatic organoids were recognized as an effective tool for screening potential cures and modeling liver diseases effectively. The current review summarizes the importance of 3D hepatic organoid culture over other conventional 2D and 3D culture models and its applicability in Liver tissue engineering.
Collapse
Affiliation(s)
- Amit Panwar
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore;
- Faculty of Biotechnology, Institute of Bio-Sciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road Barabanki, Uttar Pradesh 225003, India
| | - Prativa Das
- The Henry Samueli School of Engineering, University of California, Irvine, CA 92617, USA;
| | - Lay Poh Tan
- School of Materials Science & Engineering, Nanyang Technological University, Singapore 639798, Singapore;
- Singapore Centre for 3D Printing (SC3DP), Singapore 639798, Singapore
| |
Collapse
|
11
|
Bassan A, Alves VM, Amberg A, Anger LT, Auerbach S, Beilke L, Bender A, Cronin MT, Cross KP, Hsieh JH, Greene N, Kemper R, Kim MT, Mumtaz M, Noeske T, Pavan M, Pletz J, Russo DP, Sabnis Y, Schaefer M, Szabo DT, Valentin JP, Wichard J, Williams D, Woolley D, Zwickl C, Myatt GJ. In silico approaches in organ toxicity hazard assessment: current status and future needs in predicting liver toxicity. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 20:100187. [PMID: 35340402 PMCID: PMC8955833 DOI: 10.1016/j.comtox.2021.100187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Hepatotoxicity is one of the most frequently observed adverse effects resulting from exposure to a xenobiotic. For example, in pharmaceutical research and development it is one of the major reasons for drug withdrawals, clinical failures, and discontinuation of drug candidates. The development of faster and cheaper methods to assess hepatotoxicity that are both more sustainable and more informative is critically needed. The biological mechanisms and processes underpinning hepatotoxicity are summarized and experimental approaches to support the prediction of hepatotoxicity are described, including toxicokinetic considerations. The paper describes the increasingly important role of in silico approaches and highlights challenges to the adoption of these methods including the lack of a commonly agreed upon protocol for performing such an assessment and the need for in silico solutions that take dose into consideration. A proposed framework for the integration of in silico and experimental information is provided along with a case study describing how computational methods have been used to successfully respond to a regulatory question concerning non-genotoxic impurities in chemically synthesized pharmaceuticals.
Collapse
Affiliation(s)
- Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Vinicius M. Alves
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Alexander Amberg
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | | | - Scott Auerbach
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Lisa Beilke
- Toxicology Solutions Inc., San Diego, CA, USA
| | - Andreas Bender
- AI and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW
| | - Mark T.D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | | | - Jui-Hua Hsieh
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Nigel Greene
- Data Science and AI, DSM, IMED Biotech Unit, AstraZeneca, Boston, USA
| | - Raymond Kemper
- Nuvalent, One Broadway, 14th floor, Cambridge, MA, 02142, USA
| | - Marlene T. Kim
- US Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, 20993, USA
| | - Moiz Mumtaz
- Office of the Associate Director for Science (OADS), Agency for Toxic Substances and Disease, Registry, US Department of Health and Human Services, Atlanta, GA, USA
| | - Tobias Noeske
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Julia Pletz
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Daniel P. Russo
- Department of Chemistry, Rutgers University, Camden, NJ 08102, USA
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ 08102, USA
| | - Yogesh Sabnis
- UCB Biopharma SRL, Chemin du Foriest – B-1420 Braine-l’Alleud, Belgium
| | - Markus Schaefer
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | | | | | - Joerg Wichard
- Bayer AG, Genetic Toxicology, Müllerstr. 178, 13353 Berlin, Germany
| | - Dominic Williams
- Functional & Mechanistic Safety, Clinical Pharmacology & Safety Sciences, AstraZeneca, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, UK
| | - David Woolley
- ForthTox Limited, PO Box 13550, Linlithgow, EH49 7YU, UK
| | - Craig Zwickl
- Transendix LLC, 1407 Moores Manor, Indianapolis, IN 46229, USA
| | | |
Collapse
|
12
|
Ali S, Haque N, Azhar Z, Saeinasab M, Sefat F. Regenerative Medicine of Liver: Promises, Advances and Challenges. Biomimetics (Basel) 2021; 6:biomimetics6040062. [PMID: 34698078 PMCID: PMC8544204 DOI: 10.3390/biomimetics6040062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/16/2022] Open
Abstract
Liver tissue engineering is a rapidly developing field which combines the novel use of liver cells, appropriate biochemical factors, and engineering principles, in order to replace or regenerate damaged liver tissue or the organ. The aim of this review paper is to critically investigate different possible methods to tackle issues related with liver diseases/disorders mainly using regenerative medicine. In this work the various regenerative treatment options are discussed, for improving the prognosis of chronic liver disorders. By reviewing existing literature, it is apparent that the current popular treatment option is liver transplantation, although the breakthroughs of stem cell-based therapy and bioartificial liver technology make them a promising alternative.
Collapse
Affiliation(s)
- Saiful Ali
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Nasira Haque
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Zohya Azhar
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
| | - Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran;
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford BD7 1DP, UK; (S.A.); (N.H.); (Z.A.)
- Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford BD7 1DP, UK
- Correspondence: ; Tel.: +44-(0)-1274-233679 or +44-(0)-781-381-7460
| |
Collapse
|
13
|
Agarwal T, Banerjee D, Konwarh R, Esworthy T, Kumari J, Onesto V, Das P, Lee BH, Wagener FADTG, Makvandi P, Mattoli V, Ghosh SK, Maiti TK, Zhang LG, Ozbolat IT. Recent advances in bioprinting technologies for engineering hepatic tissue. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112013. [PMID: 33812632 DOI: 10.1016/j.msec.2021.112013] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/17/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
In the sphere of liver tissue engineering (LTE), 3D bioprinting has emerged as an effective technology to mimic the complex in vivo hepatic microenvironment, enabling the development of functional 3D constructs with potential application in the healthcare and diagnostic sector. This review gears off with a note on the liver's microscopic 3D architecture and pathologies linked to liver injury. The write-up is then directed towards unmasking recent advancements and prospects of bioprinting for recapitulating 3D hepatic structure and function. The article further introduces available stem cell opportunities and different strategies for their directed differentiation towards various hepatic stem cell types, including hepatocytes, hepatic sinusoidal endothelial cells, stellate cells, and Kupffer cells. Another thrust of the article is on understanding the dynamic interplay of different hepatic cells with various microenvironmental cues, which is crucial for controlling differentiation, maturation, and maintenance of functional hepatic cell phenotype. On a concluding note, various critical issues and future research direction towards clinical translation of bioprinted hepatic constructs are discussed.
Collapse
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Dishary Banerjee
- Department of Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Rocktotpal Konwarh
- Division of Nanobiomaterials and Nanomedicine, Uniglobe Scientific Pvt. Ltd., 7/9, Kishan Garh, Vasant Kunj, New Delhi-110070, India
| | - Timothy Esworthy
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA
| | - Jyoti Kumari
- Institute for Molecules and Materials, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, the Netherlands; Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Valentina Onesto
- Institute of Nanotechnology, National Research Council (CNR-NANOTEC), Campus Ecotekne, via Monteroni, Lecce 73100, Italy
| | - Prativa Das
- NTU-Northwestern Institute of Nanomedicine (IGS-NNIN), Nanyang Technological University, 50 Nanyang Ave, Singapore 639798, Singapore
| | - Bae Hoon Lee
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Frank A D T G Wagener
- Department of Dentistry, Section of Orthodontics and Craniofacial Biology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interface, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Virgilio Mattoli
- Istituto Italiano di Tecnologia, Centre for Materials Interface, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| | - Lijie Grace Zhang
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington, DC 20052, USA.
| | - Ibrahim T Ozbolat
- Department of Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA; The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA; Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA; Materials Research Institute, Penn State University, University Park, PA 16802, USA; Department of Neurosurgery, Penn State College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
14
|
Nanoparticle-induced inflammation and fibrosis in ex vivo murine precision-cut liver slices and effects of nanoparticle exposure conditions. Arch Toxicol 2021; 95:1267-1285. [PMID: 33555372 PMCID: PMC8032640 DOI: 10.1007/s00204-021-02992-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 01/21/2021] [Indexed: 12/20/2022]
Abstract
Chronic exposure and accumulation of persistent nanomaterials by cells have led to safety concerns on potential long-term effects induced by nanoparticles, including chronic inflammation and fibrosis. With this in mind, we used murine precision-cut liver tissue slices to test potential induction of inflammation and onset of fibrosis upon 72 h exposure to different nanomaterials (0–200 µg/ml). Tissue slices were chosen as an advanced ex vivo 3D model to better resemble the complexity of the in vivo tissue environment, with a focus on the liver where most nanomaterials accumulate. Effects on the onset of fibrosis and inflammation were investigated, with particular care in optimizing nanoparticle exposure conditions to tissue. Thus, we compared the effects induced on slices exposed to nanoparticles in the presence of excess free proteins (in situ), or after corona isolation. Slices exposed to daily-refreshed nanoparticle dispersions were used to test additional effects due to ageing of the dispersions. Exposure to amino-modified polystyrene nanoparticles in serum-free conditions led to strong inflammation, with stronger effects with daily-refreshed dispersions. Instead, no inflammation was observed when slices were exposed to the same nanoparticles in medium supplemented with serum to allow corona formation. Similarly, no clear signs of inflammation nor of onset of fibrosis were detected after exposure to silica, titania or carboxylated polystyrene in all conditions tested. Overall, these results show that liver slices can be used to test nanoparticle-induced inflammation in real tissue, and that the exposure conditions and ageing of the dispersions can strongly affect tissue responses to nanoparticles.
Collapse
|
15
|
Tissue Chips and Microphysiological Systems for Disease Modeling and Drug Testing. MICROMACHINES 2021; 12:mi12020139. [PMID: 33525451 PMCID: PMC7911320 DOI: 10.3390/mi12020139] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Tissue chips (TCs) and microphysiological systems (MPSs) that incorporate human cells are novel platforms to model disease and screen drugs and provide an alternative to traditional animal studies. This review highlights the basic definitions of TCs and MPSs, examines four major organs/tissues, identifies critical parameters for organization and function (tissue organization, blood flow, and physical stresses), reviews current microfluidic approaches to recreate tissues, and discusses current shortcomings and future directions for the development and application of these technologies. The organs emphasized are those involved in the metabolism or excretion of drugs (hepatic and renal systems) and organs sensitive to drug toxicity (cardiovascular system). This article examines the microfluidic/microfabrication approaches for each organ individually and identifies specific examples of TCs. This review will provide an excellent starting point for understanding, designing, and constructing novel TCs for possible integration within MPS.
Collapse
|
16
|
Kobayashi J, Arisaka Y, Yui N, Yamato M, Okano T. Preservation of heparin-binding EGF-like growth factor activity on heparin-modified poly( N-isopropylacrylamide)-grafted surfaces. RSC Adv 2021; 11:37225-37232. [PMID: 35496401 PMCID: PMC9043771 DOI: 10.1039/d1ra07317f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/09/2021] [Indexed: 11/28/2022] Open
Abstract
A heparin-modified poly(N-isopropylacrylamide) (PIPAAm)-grafted surface bound with heparin-binding epidermal growth factor-like growth factor (HB-EGF) was able to culture hepatocytes maintaining high albumin secretion and high expression of hepatocyte-specific genes. However, the activity of HB-EGF on the surface and its binding effects on hepatocytes remain unclear. In this study, we investigated the temperature-dependent interactions of HB-EGF and EGF receptor (EGFR) with heparin-modified PIPAAm to evaluate the activity of HB-EGF on the surface. Quartz crystal microbalance (QCM) measurements revealed that the amounts of adsorbed HB-EGF on either the heparin-modified PIPAAm-grafted surface (heparin-IC1) or PIPAAm-grafted surfaces were almost the same regardless of swelling/deswelling of grafted PIPAAm chains. The heparin-IC1 surface bound to HB-EGF at 37 °C had the ability to bind to hepatocytes through specific affinity interaction with EGFR, whose activation was confirmed by western blotting. However, the physisorbed HB-EGF on the PIPAAm surface greatly diminished its activity. Taken together, the introduction of heparin into grafted PIPAAm chains on the surface plays a pivotal role in holding HB-EGF while preserving its activity. Hydration and swelling of surface-grafted PIPAAm chains at 20 °C greatly diminished the attachment of hepatocytes with HB-EGF bound to heparin-IC1, whereas hepatocytes were able to bind to HB-EGF bound to heparin-IC1 at 37 °C. Thus, the equilibrated affinity interaction between EGFRs and surface-bound HB-EGF was considered to be attenuated by steric hindrance due to hydration and/or swelling of grafted PIPAAm chains. Activity of HB-EGF bound to a heparin-modified poly(N-isopropylacrylamide) (PIPAAm)-grafted surface was preserved through specific binding to heparin, whereas physisorbed HB-EGF on a PIPAAm-grafted surface greatly diminished its activity.![]()
Collapse
Affiliation(s)
- Jun Kobayashi
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Masayuki Yamato
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Teruo Okano
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, TWIns, 8-1 Kawadacho, Shinjuku-ku, Tokyo 162-8666, Japan
- Cell Sheet Tissue Engineering Center, Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, 30 South 2000 East, Salt Lake City, UT 84112, USA
| |
Collapse
|
17
|
Moradi E, Jalili-Firoozinezhad S, Solati-Hashjin M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomater 2020; 116:67-83. [PMID: 32890749 DOI: 10.1016/j.actbio.2020.08.041] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/08/2023]
Abstract
The liver is the largest internal organ of the body with complex microarchitecture and function that plays critical roles in drug metabolism. Hepatotoxicity and drug-induced liver injury (DILI) caused by various drugs is the main reason for late-stage drug failures. Moreover, liver diseases are among the leading causes of death in the world, with the number of new cases arising each year. Although animal models have been used to understand human drug metabolism and toxicity before clinical trials, tridimensional microphysiological systems, such as liver-on-a-chip (Liver Chip) platforms, could better recapitulate features of human liver physiology and pathophysiology and thus, are often more predictive of human outcome. Liver Chip devices have shown promising results in mimicking in vivo condition by recapitulating the sinusoidal structure of the liver, maintaining high cell viability and cellular phenotypes, and emulating native liver functions. Here, we first review the cellular constituents and physiology of the liver and then critically discuss the state-of-the-art chip-based liver models and their applications in drug screening, disease modeling, and regenerative medicine. We finally address the pending issues of existing platforms and touch upon future directions for developing new, advanced on-chip models.
Collapse
Affiliation(s)
- Ehsanollah Moradi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Mehran Solati-Hashjin
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran.
| |
Collapse
|
18
|
Miyawaki I. Application of zebrafish to safety evaluation in drug discovery. J Toxicol Pathol 2020; 33:197-210. [PMID: 33239838 PMCID: PMC7677624 DOI: 10.1293/tox.2020-0021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Traditionally, safety evaluation at the early stage of drug discovery research has been done using in silico, in vitro, and in vivo systems in this order because of limitations on the amount of compounds available and the throughput ability of the assay systems. While these in vitro assays are very effective tools for detecting particular tissue-specific toxicity phenotypes, it is difficult to detect toxicity based on complex mechanisms involving multiple organs and tissues. Therefore, the development of novel high throughput in vivo evaluation systems has been expected for a long time. The zebrafish (Danio rerio) is a vertebrate with many attractive characteristics for use in drug discovery, such as a small size, transparency, gene and protein similarity with mammals (80% or more), and ease of genetic modification to establish human disease models. Actually, in recent years, the zebrafish has attracted interest as a novel experimental animal. In this article, the author summarized the features of zebrafish that make it a suitable laboratory animal, and introduced and discussed the applications of zebrafish to preclinical toxicity testing, including evaluations of teratogenicity, hepatotoxicity, and nephrotoxicity based on morphological findings, evaluation of cardiotoxicity using functional endpoints, and assessment of seizure and drug abuse liability.
Collapse
Affiliation(s)
- Izuru Miyawaki
- Preclinical Research Laboratories, Sumitomo Dainippon Pharma
Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka 554-0022, Japan
| |
Collapse
|
19
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
20
|
Agarwal T, Borrelli MR, Makvandi P, Ashrafizadeh M, Maiti TK. Paper-Based Cell Culture: Paving the Pathway for Liver Tissue Model Development on a Cellulose Paper Chip. ACS APPLIED BIO MATERIALS 2020; 3:3956-3974. [DOI: 10.1021/acsabm.0c00558] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| | - Mimi R. Borrelli
- Department of Surgery, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Pooyan Makvandi
- Institute for Polymers, Composites and Biomaterials (IPCB), National Research Council (CNR), Naples 80078, Italy
| | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 51666-16471, Iran
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, West Bengal, India
| |
Collapse
|
21
|
Umar AB, Uzairu A, Shallangwa GA, Uba S. Design of potential anti-melanoma agents against SK-MEL-5 cell line using QSAR modeling and molecular docking methods. SN APPLIED SCIENCES 2020. [DOI: 10.1007/s42452-020-2620-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
22
|
Agarwal T, Biswas P, Pal S, Maiti TK, Chakraborty S, Ghosh SK, Dhar R. Inexpensive and Versatile Paper-Based Platform for 3D Culture of Liver Cells and Related Bioassays. ACS APPLIED BIO MATERIALS 2020; 3:2522-2533. [PMID: 35025303 DOI: 10.1021/acsabm.0c00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Pratik Biswas
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Sampriti Pal
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Tapas Kumar Maiti
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Suman Chakraborty
- Department of Mechanical Engineering, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| | - Riddhiman Dhar
- Department of Biotechnology, Indian Institute of Technology, Kharagpur, West Bengal 721302, India
| |
Collapse
|
23
|
Umar AB, Uzairu A, Shallangwa GA, Uba S. QSAR modelling and molecular docking studies for anti-cancer compounds against melanoma cell line SK-MEL-2. Heliyon 2020; 6:e03640. [PMID: 32258485 PMCID: PMC7110328 DOI: 10.1016/j.heliyon.2020.e03640] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/06/2020] [Accepted: 03/18/2020] [Indexed: 11/04/2022] Open
Abstract
A dataset of seventy-two (72) cytotoxic compounds of the National Cancer Institute (NCI) was studied by QSAR and docking approaches to gain deeper insights into ligands selectivity on SK-MEL-2 cell line. The QSAR model was built using fifty (50) molecules and the best-generated model based on multiple linear regression showed, respectively good quality of fits (R 2 (0.864),R a d j u s t e d 2 (0.845), Q2 cv (0.799) andR p r e d 2 (0.706)). The model's predictive ability was determined by a test set of twenty-two (22) compounds. Compounds 30 and 41 were selected as templates for in silico design because they had high pGI50 activity and are in the model's applicability domain. The obtained information from the model was explored to design novel molecules by introducing various modifications. Moreover, the designed compounds with better-predicted activity (pGI50) values were selected and docked on the active site of the protein (PDB-CODE: 3OG7) which is responsible for melanoma cancer to elucidate their binding mode. AN2 (-12.1kcalmol-1) and AC4 (-12.4kcalmol-1) showed a better binding score for the target when compared with (vemurafenib, -11.3kcalmol-1) the known inhibitor of the target (V600E-BRAF). These findings may be very helpful in early anti-cancer drug development.
Collapse
Affiliation(s)
- Abdullahi Bello Umar
- Department of Chemistry, Faculty of Physical Sciences, Ahmad Bello University, Zaria, P.M.B.1045 Kaduna State, Nigeria
| | | | | | | |
Collapse
|
24
|
Bale SS, Manoppo A, Thompson R, Markoski A, Coppeta J, Cain B, Haroutunian N, Newlin V, Spencer A, Azizgolshani H, Lu M, Gosset J, Keegan P, Charest JL. A thermoplastic microfluidic microphysiological system to recapitulate hepatic function and multicellular interactions. Biotechnol Bioeng 2019; 116:3409-3420. [DOI: 10.1002/bit.26986] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/29/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Affiliation(s)
| | | | | | - Alex Markoski
- DraperCambridge Massachusetts
- Department of Biomedical EngineeringWorcester Polytechnic InstituteWorcester Massachusetts
| | | | | | | | | | | | | | - Mingjian Lu
- Pfizer Global Research and Development Cambridge Massachusetts
| | - James Gosset
- Pfizer Global Research and Development Cambridge Massachusetts
| | | | | |
Collapse
|
25
|
Deng J, Wei W, Chen Z, Lin B, Zhao W, Luo Y, Zhang X. Engineered Liver-on-a-Chip Platform to Mimic Liver Functions and Its Biomedical Applications: A Review. MICROMACHINES 2019; 10:E676. [PMID: 31591365 PMCID: PMC6843249 DOI: 10.3390/mi10100676] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/03/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Hepatology and drug development for liver diseases require in vitro liver models. Typical models include 2D planar primary hepatocytes, hepatocyte spheroids, hepatocyte organoids, and liver-on-a-chip. Liver-on-a-chip has emerged as the mainstream model for drug development because it recapitulates the liver microenvironment and has good assay robustness such as reproducibility. Liver-on-a-chip with human primary cells can potentially correlate clinical testing. Liver-on-a-chip can not only predict drug hepatotoxicity and drug metabolism, but also connect other artificial organs on the chip for a human-on-a-chip, which can reflect the overall effect of a drug. Engineering an effective liver-on-a-chip device requires knowledge of multiple disciplines including chemistry, fluidic mechanics, cell biology, electrics, and optics. This review first introduces the physiological microenvironments in the liver, especially the cell composition and its specialized roles, and then summarizes the strategies to build a liver-on-a-chip via microfluidic technologies and its biomedical applications. In addition, the latest advancements of liver-on-a-chip technologies are discussed, which serve as a basis for further liver-on-a-chip research.
Collapse
Affiliation(s)
- Jiu Deng
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Wenbo Wei
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Zongzheng Chen
- Integrated Chinese and Western Medicine Postdoctoral research station, Jinan University, Guangzhou 510632, China;
| | - Bingcheng Lin
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
26
|
Brovold M, Almeida JI, Pla-Palacín I, Sainz-Arnal P, Sánchez-Romero N, Rivas JJ, Almeida H, Dachary PR, Serrano-Aulló T, Soker S, Baptista PM. Naturally-Derived Biomaterials for Tissue Engineering Applications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1077:421-449. [PMID: 30357702 PMCID: PMC7526297 DOI: 10.1007/978-981-13-0947-2_23] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Naturally-derived biomaterials have been used for decades in multiple regenerative medicine applications. From the simplest cell microcarriers made of collagen or alginate, to highly complex decellularized whole-organ scaffolds, these biomaterials represent a class of substances that is usually first in choice at the time of electing a functional and useful biomaterial. Hence, in this chapter we describe the several naturally-derived biomaterials used in tissue engineering applications and their classification, based on composition. We will also describe some of the present uses of the generated tissues like drug discovery, developmental biology, bioprinting and transplantation.
Collapse
Affiliation(s)
- Matthew Brovold
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Joana I Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Iris Pla-Palacín
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Pilar Sainz-Arnal
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
- Aragon Health Sciences Institute (IACS), Zaragoza, Spain
| | | | - Jesus J Rivas
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Helen Almeida
- Health Research Institute of Aragón (IIS Aragón), Zaragoza, Spain
| | - Pablo Royo Dachary
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Liver Transplant Unit, Gastroenterology Department, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Trinidad Serrano-Aulló
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Liver Transplant Unit, Gastroenterology Department, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Shay Soker
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA.
| | - Pedro M Baptista
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain.
- Center for Biomedical Research Network Liver and Digestive Diseases (CIBERehd), Zaragoza, Spain.
- Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.
- Biomedical and Aerospace Engineering Department, Universidad Carlos III de Madrid, Madrid, Spain.
- Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
27
|
Li L, Gokduman K, Gokaltun A, Yarmush ML, Usta OB. A microfluidic 3D hepatocyte chip for hepatotoxicity testing of nanoparticles. Nanomedicine (Lond) 2019; 14:2209-2226. [PMID: 31179822 DOI: 10.2217/nnm-2019-0086] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: To develop a practical microfluidic 3D hepatocyte chip for hepatotoxicity testing of nanoparticles using proof of concept studies providing first results of the potential hepatotoxicity of superparamagnetic iron oxide nanoparticles (SPION) under microfluidic conditions. Methods: A microfluidic 3D hepatocyte chip with three material layers, which contains primary rat hepatocytes, has been fabricated and tested using different concentrations (50, 100 and 200 μg/ml) of SPION in 3-day (short-term) and 1-week (long-term) cultures. Results: Compared with standard well plates, the hepatocyte chip with flow provided comparable viability and significantly higher liver-specific functions, up to 1 week. In addition, the chip recapitulates the key physiological responses in the hepatotoxicity of SPION. Conclusion: Thus, the developed 3D hepatocyte chip is a robust and highly sensitive platform for investigating hepatotoxicity profiles of nanoparticles.
Collapse
Affiliation(s)
- Lei Li
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA.,CAS Key Laboratory of Cryogenics, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, PR China
| | - Kurtulus Gokduman
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
| | - Aslihan Gokaltun
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA.,Department of Chemical Engineering, Hacettepe University, 06800, Beytepe, Ankara, Turkey
| | - Martin L Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA.,Rutgers State University, Department of Biomedical Engineering, Piscataway, NJ 08854, USA
| | - Osman Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals, Boston, MA 02114, USA
| |
Collapse
|
28
|
Gökaltun A, Kang YBA, Yarmush ML, Usta OB, Asatekin A. Simple Surface Modification of Poly(dimethylsiloxane) via Surface Segregating Smart Polymers for Biomicrofluidics. Sci Rep 2019; 9:7377. [PMID: 31089162 PMCID: PMC6517421 DOI: 10.1038/s41598-019-43625-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/09/2019] [Indexed: 12/17/2022] Open
Abstract
Poly(dimethylsiloxane) (PDMS) is likely the most popular material for microfluidic devices in lab-on-a-chip and other biomedical applications. However, the hydrophobicity of PDMS leads to non-specific adsorption of proteins and other molecules such as therapeutic drugs, limiting its broader use. Here, we introduce a simple method for preparing PDMS materials to improve hydrophilicity and decrease non-specific protein adsorption while retaining cellular biocompatibility, transparency, and good mechanical properties without the need for any post-cure surface treatment. This approach utilizes smart copolymers comprised of poly(ethylene glycol) (PEG) and PDMS segments (PDMS-PEG) that, when blended with PDMS during device manufacture, spontaneously segregate to surfaces in contact with aqueous solutions and reduce the hydrophobicity without any added manufacturing steps. PDMS-PEG-modified PDMS samples showed contact angles as low as 23.6° ± 1° and retained this hydrophilicity for at least twenty months. Their improved wettability was confirmed using capillary flow experiments. Modified devices exhibited considerably reduced non-specific adsorption of albumin, lysozyme, and immunoglobulin G. The modified PDMS was biocompatible, displaying no adverse effects when used in a simple liver-on-a-chip model using primary rat hepatocytes. This PDMS modification method can be further applied in analytical separations, biosensing, cell studies, and drug-related studies.
Collapse
Affiliation(s)
- Aslıhan Gökaltun
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114, USA
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA, 02474, USA
- Department of Chemical Engineering, Hacettepe University, 06532, Beytepe, Ankara, Turkey
| | - Young Bok Abraham Kang
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114, USA
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ, 08854, USA
| | - O Berk Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114, USA.
| | - Ayse Asatekin
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA, 02474, USA.
| |
Collapse
|
29
|
Tan K, Keegan P, Rogers M, Lu M, Gosset JR, Charest J, Bale SS. A high-throughput microfluidic microphysiological system (PREDICT-96) to recapitulate hepatocyte function in dynamic, re-circulating flow conditions. LAB ON A CHIP 2019; 19:1556-1566. [PMID: 30855604 DOI: 10.1039/c8lc01262h] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Microphysiological systems (MPSs) are dynamic cell culture systems that provide micro-environmental and external cues to support physiologically relevant, organ-specific functions. Recent progresses in MPS fabrication technologies have enabled the development of advanced models to capture microenvironments with physiological relevance, while increasing throughput and reducing material-based artefacts. In addition to conventional cell culture systems, advanced MPSs are emerging as ideal contenders for disease modeling and incorporation into drug screening. Since liver is a central organ for drug metabolism, liver-on-chip models have been developed to recapitulate hepatic microenvironment with varying complexities, while allowing long-term culture. Recently, we have developed a novel thermoplastic, oxygen-permeable MPS for primary human hepatocyte (PHH) culture. Herein, we have adapted and extended the MPS to a) a 96 microfluidic array (PREDICT-96 array) and b) integrated a novel, ultra-low volume, re-circulating pumping system (PREDICT-96 pump) - collectively known as the PREDICT-96 platform. The PREDICT-96 platform conforms to the industrial standard 96-well footprint and enables media re-circulation. First, we demonstrate the introduction of PHHs into the PREDICT-96 array using standard handling procedures for multi-well plates and allow cells to stabilize in static conditions. Next, we introduce recirculating flow into the bottom channel (using PREDICT-96 pump) to mimic mass transport in vivo. Our results indicate an increase in metabolic and secretory functions of PHHs in the PREDICT-96 platform, and their maintenance over 10 days of flow. Furthermore, long-term culture with fluid flow allows for the periodic introduction of media components (e.g., fatty acids, cytokines) and capture cellular responses to chronic stimuli. The low-volume footprint of the pump and small media volume in the MPS allow for the interrogation of hepatic responses incorporating secretion feedback to a stimulus, which is essential for disease model development and drug interrogation. We envision future development of this liver model to incorporate key primary hepatic cells, multi-cellular co-cultures and adaptation, integration with high-throughput analytical tools.
Collapse
Affiliation(s)
- Kelly Tan
- Draper, 555 Technology Square, Cambridge, MA 02138, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Fernández‐Iglesias A, Ortega‐Ribera M, Guixé‐Muntet S, Gracia‐Sancho J. 4 in 1: Antibody-free protocol for isolating the main hepatic cells from healthy and cirrhotic single rat livers. J Cell Mol Med 2019; 23:877-886. [PMID: 30417530 PMCID: PMC6349241 DOI: 10.1111/jcmm.13988] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 10/03/2018] [Indexed: 12/11/2022] Open
Abstract
Liver cells isolated from pre-clinical models are essential tools for studying liver (patho)physiology, and also for screening new therapeutic options. We aimed at developing a new antibody-free isolation method able to obtain the four main hepatic cell types (hepatocytes, liver sinusoidal endothelial cells [LSEC], hepatic macrophages [HMΦ] and hepatic stellate cells [HSC]) from a single rat liver. Control and cirrhotic (CCl4 and TAA) rat livers (n = 6) were perfused, digested with collagenase and mechanically disaggregated obtaining a multicellular suspension. Hepatocytes were purified by low revolution centrifugations while non-parenchymal cells were subjected to differential centrifugation. Two different fractions were obtained: HSC and mixed LSEC + HMΦ. Further LSEC and HMΦ enrichment was achieved by selective adherence time to collagen-coated substrates. Isolated cells showed high viability (80%-95%) and purity (>95%) and were characterized as functional: hepatocytes synthetized albumin and urea, LSEC maintained endocytic capacity and in vivo fenestrae distribution, HMΦ increased expression of inflammatory markers in response to LPS and HSC were activated upon in vitro culture. The 4 in 1 protocol allows the simultaneous isolation of highly pure and functional hepatic cell sub-populations from control or cirrhotic single livers without antibody selection.
Collapse
Affiliation(s)
- Anabel Fernández‐Iglesias
- Liver Vascular Biology Research GroupBarcelona Hepatic Hemodynamic LaboratoryIDIBAPS Biomedical Research InstituteBarcelonaSpain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD)MadridSpain
| | - Martí Ortega‐Ribera
- Liver Vascular Biology Research GroupBarcelona Hepatic Hemodynamic LaboratoryIDIBAPS Biomedical Research InstituteBarcelonaSpain
| | - Sergi Guixé‐Muntet
- HepatologyDepartment of Biomedical ResearchInselspitalBern UniversityBernSwitzerland
| | - Jordi Gracia‐Sancho
- Liver Vascular Biology Research GroupBarcelona Hepatic Hemodynamic LaboratoryIDIBAPS Biomedical Research InstituteBarcelonaSpain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD)MadridSpain
- HepatologyDepartment of Biomedical ResearchInselspitalBern UniversityBernSwitzerland
| |
Collapse
|
31
|
Eggert S, Alexander FA, Wiest J. Enabling 3D hepatocyte spheroids for microphysiometry. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2017:1617-1620. [PMID: 29060192 DOI: 10.1109/embc.2017.8037148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Advances in the areas of tissue engineering and microfabrication techniques have enabled promising in vitro platforms, known as Organs-on-Chips, with the aim of mimicking complex in vivo conditions for more accurate toxicology studies. To analyze the physiological change induced by chemicals or toxic substances continuously, sensors can be used in order to measure the intracellular and extracellular environment of single cells, cell constructs, or tissue, and therefore the integration of monitoring techniques into 3D tissue culture platforms provides an essential step for the next generation Organ-on-Chip platforms. However, current in vitro platforms are not capable of combining the culture of 3D models with monitoring techniques. To address this, a novel spheroid encapsulation is designed for fluidic contact between 3D models in microwells and Intelligent Mobile Lab for In Vitro Diagnostics (IMOLA-IVD) BioChip sensors while preventing spheroid fusion. In this work, spheroid culturing protocols were developed for optimized spheroid growth and an evaluation of spheroid integrity on different porous layers was performed in order to provide a defined spheroid encapsulation on BioChip sensors.
Collapse
|
32
|
Ortega-Ribera M, Fernández-Iglesias A, Illa X, Moya A, Molina V, Maeso-Díaz R, Fondevila C, Peralta C, Bosch J, Villa R, Gracia-Sancho J. Resemblance of the human liver sinusoid in a fluidic device with biomedical and pharmaceutical applications. Biotechnol Bioeng 2018; 115:2585-2594. [PMID: 29940068 PMCID: PMC6220781 DOI: 10.1002/bit.26776] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/10/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022]
Abstract
Maintenance of the complex phenotype of primary hepatocytes in vitro represents a limitation for developing liver support systems and reliable tools for biomedical research and drug screening. We herein aimed at developing a biosystem able to preserve human and rodent hepatocytes phenotype in vitro based on the main characteristics of the liver sinusoid: unique cellular architecture, endothelial biodynamic stimulation, and parenchymal zonation. Primary hepatocytes and liver sinusoidal endothelial cells (LSEC) were isolated from control and cirrhotic human or control rat livers and cultured in conventional in vitro platforms or within our liver‐resembling device. Hepatocytes phenotype, function, and response to hepatotoxic drugs were analyzed. Results evidenced that mimicking the in vivo sinusoidal environment within our biosystem, primary human and rat hepatocytes cocultured with functional LSEC maintained morphology and showed high albumin and urea production, enhanced cytochrome P450 family 3 subfamily A member 4 (CYP3A4) activity, and maintained expression of hepatocyte nuclear factor 4 alpha (hnf4α) and transporters, showing delayed hepatocyte dedifferentiation. In addition, differentiated hepatocytes cultured within this liver‐resembling device responded to acute treatment with known hepatotoxic drugs significantly different from those seen in conventional culture platforms. In conclusion, this study describes a new bioengineered device that mimics the human sinusoid in vitro, representing a novel method to study liver diseases and toxicology.
Collapse
Affiliation(s)
- Martí Ortega-Ribera
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS Biomedical Research Institute, Barcelona, Spain.,Biomedical Applications Group (GAB), Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain
| | - Anabel Fernández-Iglesias
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS Biomedical Research Institute, Barcelona, Spain.,Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain
| | - Xavi Illa
- Biomedical Applications Group (GAB), Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBERBBN), Madrid, Spain
| | - Ana Moya
- Biomedical Applications Group (GAB), Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBERBBN), Madrid, Spain
| | - Víctor Molina
- Liver Surgery and Transplantation Unit, IDIBAPS, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Raquel Maeso-Díaz
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS Biomedical Research Institute, Barcelona, Spain
| | - Constantino Fondevila
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain.,Liver Surgery and Transplantation Unit, IDIBAPS, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Carmen Peralta
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain.,Protective Strategies Against Hepatic Ischemia-Reperfusion Group, IDIBAPS, Barcelona, Spain
| | - Jaume Bosch
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS Biomedical Research Institute, Barcelona, Spain.,Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain.,Hepatology, Department of Biomedical Research, Inselspital, Bern University, Bern, Switzerland
| | - Rosa Villa
- Biomedical Applications Group (GAB), Institut de Microelectrònica de Barcelona, IMB-CNM (CSIC), Esfera UAB, Bellaterra, Spain.,Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBERBBN), Madrid, Spain
| | - Jordi Gracia-Sancho
- Liver Vascular Biology Research Group, Barcelona Hepatic Hemodynamic Laboratory, IDIBAPS Biomedical Research Institute, Barcelona, Spain.,Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBEREHD), Madrid, Spain.,Hepatology, Department of Biomedical Research, Inselspital, Bern University, Bern, Switzerland
| |
Collapse
|
33
|
Metabolic Patterning on a Chip: Towards in vitro Liver Zonation of Primary Rat and Human Hepatocytes. Sci Rep 2018; 8:8951. [PMID: 29895900 PMCID: PMC5997652 DOI: 10.1038/s41598-018-27179-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/29/2018] [Indexed: 01/15/2023] Open
Abstract
An important number of healthy and diseased tissues shows spatial variations in their metabolic capacities across the tissue. The liver is a prime example of such heterogeneity where the gradual changes in various metabolic activities across the liver sinusoid is termed as “zonation” of the liver. Here, we introduce the Metabolic Patterning on a Chip (MPOC) platform capable of dynamically creating metabolic patterns across the length of a microchamber of liver tissue via actively enforced gradients of various metabolic modulators such as hormones and inducers. Using this platform, we were able to create continuous liver tissues of both rat and human origin with gradually changing metabolic activities. The gradients we have created in nitrogen, carbohydrate and xenobiotic metabolisms recapitulated an in vivo like zonation and zonal toxic response. Beyond its application in recapitulation of liver zonation in vitro as we demonstrate here, the MPOC platform can be used and expanded for a variety of purposes including better understanding of heterogeneity in many different tissues during developmental and adult stages.
Collapse
|
34
|
Wang C, Tanataweethum N, Karnik S, Bhushan A. Novel Microfluidic Colon with an Extracellular Matrix Membrane. ACS Biomater Sci Eng 2018; 4:1377-1385. [DOI: 10.1021/acsbiomaterials.7b00883] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Chengyao Wang
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616, United States
| | - Nida Tanataweethum
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616, United States
| | - Sonali Karnik
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616, United States
| | - Abhinav Bhushan
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois 60616, United States
| |
Collapse
|
35
|
Huang H, Yu Y, Hu Y, He X, Usta OB, Yarmush ML. Generation and manipulation of hydrogel microcapsules by droplet-based microfluidics for mammalian cell culture. LAB ON A CHIP 2017; 17:1913-1932. [PMID: 28509918 PMCID: PMC5548188 DOI: 10.1039/c7lc00262a] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Hydrogel microcapsules provide miniaturized and biocompatible niches for three-dimensional (3D) in vitro cell culture. They can be easily generated by droplet-based microfluidics with tunable size, morphology, and biochemical properties. Therefore, microfluidic generation and manipulation of cell-laden microcapsules can be used for 3D cell culture to mimic the in vivo environment towards applications in tissue engineering and high throughput drug screening. In this review of recent advances mainly since 2010, we will first introduce general characteristics of droplet-based microfluidic devices for cell encapsulation with an emphasis on the fluid dynamics of droplet breakup and internal mixing as they directly influence microcapsule's size and structure. We will then discuss two on-chip manipulation strategies: sorting and extraction from oil into aqueous phase, which can be integrated into droplet-based microfluidics and significantly improve the qualities of cell-laden hydrogel microcapsules. Finally, we will review various applications of hydrogel microencapsulation for 3D in vitro culture on cell growth and proliferation, stem cell differentiation, tissue development, and co-culture of different types of cells.
Collapse
Affiliation(s)
- Haishui Huang
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Yin Yu
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Yong Hu
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Xiaoming He
- Department of Biomedical Engineering, The Ohio State University,
Columbus, USA
| | - O. Berk Usta
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
| | - Martin L. Yarmush
- Center for Engineering in Medicine, Massachusetts General Hospital,
Harvard Medical School and Shriners Hospitals for Children, Boston, Massachusetts
02114, United States
- Department of Biomedical Engineering, Rutgers University,
Piscataway, New Jersey 08854, United States
| |
Collapse
|
36
|
Gokaltun A, Yarmush ML, Asatekin A, Usta OB. Recent advances in nonbiofouling PDMS surface modification strategies applicable to microfluidic technology. TECHNOLOGY 2017; 5:1-12. [PMID: 28695160 PMCID: PMC5501164 DOI: 10.1142/s2339547817300013] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
In the last decade microfabrication processes including rapid prototyping techniques have advanced rapidly and achieved a fairly mature stage. These advances have encouraged and enabled the use of microfluidic devices by a wider range of users with applications in biological separations and cell and organoid cultures. Accordingly, a significant current challenge in the field is controlling biomolecular interactions at interfaces and the development of novel biomaterials to satisfy the unique needs of the biomedical applications. Poly(dimethylsiloxane) (PDMS) is one of the most widely used materials in the fabrication of microfluidic devices. The popularity of this material is the result of its low cost, simple fabrication allowing rapid prototyping, high optical transparency, and gas permeability. However, a major drawback of PDMS is its hydrophobicity and fast hydrophobic recovery after surface hydrophilization. This results in significant nonspecific adsorption of proteins as well as small hydrophobic molecules such as therapeutic drugs limiting the utility of PDMS in biomedical microfluidic circuitry. Accordingly, here, we focus on recent advances in surface molecular treatments to prevent fouling of PDMS surfaces towards improving its utility and expanding its use cases in biomedical applications.
Collapse
Affiliation(s)
- Aslihan Gokaltun
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA 02474, USA
- Department of Chemical Engineering, Hacettepe University, 06532, Beytepe, Ankara, Turkey
| | - Martin L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ 08854, USA
| | - Ayse Asatekin
- Department of Chemical and Biological Engineering, Tufts University, 4 Colby Street, Medford, MA 02474, USA
| | - O Berk Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School, and Shriners Hospital for Children, 51 Blossom St., Boston, MA 02114, USA
| |
Collapse
|
37
|
Matli PR, Ubaid F, Shakoor RA, Parande G, Manakari V, Yusuf M, Amer Mohamed AM, Gupta M. Improved properties of Al–Si3N4 nanocomposites fabricated through a microwave sintering and hot extrusion process. RSC Adv 2017. [DOI: 10.1039/c7ra04148a] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In this study, nano-sized Si3N4 (0, 0.5, 1.0 and 1.5 vol%)/Al composites were fabricated using a powder metallurgy method involving microwave sintering technique followed by hot extrusion.
Collapse
Affiliation(s)
| | - Fareeha Ubaid
- Center for Advanced Materials
- Qatar University
- Doha
- Qatar
| | | | - Gururaj Parande
- Department of Mechanical Engineering
- National University of Singapore
- Singapore
| | - Vyasaraj Manakari
- Department of Mechanical Engineering
- National University of Singapore
- Singapore
| | | | | | - Manoj Gupta
- Department of Mechanical Engineering
- National University of Singapore
- Singapore
| |
Collapse
|
38
|
Starokozhko V, Groothuis GMM. Judging the value of ‘liver-on-a-chip’ devices for prediction of toxicity. Expert Opin Drug Metab Toxicol 2016; 13:125-128. [DOI: 10.1080/17425255.2017.1246537] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Viktoriia Starokozhko
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Geny M. M. Groothuis
- Division of Pharmacokinetics, Toxicology and Targeting, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
39
|
McCarty WJ, Usta OB, Yarmush ML. A Microfabricated Platform for Generating Physiologically-Relevant Hepatocyte Zonation. Sci Rep 2016; 6:26868. [PMID: 27240736 PMCID: PMC4886516 DOI: 10.1038/srep26868] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 05/10/2016] [Indexed: 12/29/2022] Open
Abstract
In vitro liver models have been important tools for more than 40 years for academic research and preclinical toxicity screening by the pharmaceutical industry. Hepatocytes, the highly metabolic parenchymal cells of the liver, are efficient at different metabolic chemistries depending on their relative spatial location along the sinusoid from the portal triad to the central vein. Although replicating hepatocyte metabolic zonation is vitally important for physiologically-relevant in vitro liver tissue and organ models, it is most often completely overlooked. Here, we demonstrate the creation of spatially-controlled zonation across multiple hepatocyte metabolism levels through the application of precise concentration gradients of exogenous hormone (insulin and glucagon) and chemical (3-methylcholanthrene) induction agents in a microfluidic device. Observed gradients in glycogen storage via periodic acid-Schiff staining, urea production via carbamoyl phosphatase synthetase I staining, and cell viability after exposure to allyl alcohol and acetaminophen demonstrated the in vitro creation of hepatocyte carbohydrate, nitrogen, alcohol degradation, and drug conjugation metabolic zonation. This type of advanced control system will be crucial for studies evaluating drug metabolism and toxicology using in vitro constructs.
Collapse
Affiliation(s)
- William J McCarty
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
| | - O Berk Usta
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
| | - Martin L Yarmush
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, and Shriners Hospitals for Children-Boston, Boston, MA, USA
| |
Collapse
|
40
|
Eribol P, Uguz AK, Ulgen KO. Screening applications in drug discovery based on microfluidic technology. BIOMICROFLUIDICS 2016; 10:011502. [PMID: 26865904 PMCID: PMC4733079 DOI: 10.1063/1.4940886] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/14/2016] [Indexed: 05/03/2023]
Abstract
Microfluidics has been the focus of interest for the last two decades for all the advantages such as low chemical consumption, reduced analysis time, high throughput, better control of mass and heat transfer, downsizing a bench-top laboratory to a chip, i.e., lab-on-a-chip, and many others it has offered. Microfluidic technology quickly found applications in the pharmaceutical industry, which demands working with leading edge scientific and technological breakthroughs, as drug screening and commercialization are very long and expensive processes and require many tests due to unpredictable results. This review paper is on drug candidate screening methods with microfluidic technology and focuses specifically on fabrication techniques and materials for the microchip, types of flow such as continuous or discrete and their advantages, determination of kinetic parameters and their comparison with conventional systems, assessment of toxicities and cytotoxicities, concentration generations for high throughput, and the computational methods that were employed. An important conclusion of this review is that even though microfluidic technology has been in this field for around 20 years there is still room for research and development, as this cutting edge technology requires ingenuity to design and find solutions for each individual case. Recent extensions of these microsystems are microengineered organs-on-chips and organ arrays.
Collapse
Affiliation(s)
- P Eribol
- Department of Chemical Engineering, Boğaziçi University , 34342 Bebek, Istanbul, Turkey
| | - A K Uguz
- Department of Chemical Engineering, Boğaziçi University , 34342 Bebek, Istanbul, Turkey
| | - K O Ulgen
- Department of Chemical Engineering, Boğaziçi University , 34342 Bebek, Istanbul, Turkey
| |
Collapse
|
41
|
Chen P, Güven S, Usta OB, Yarmush ML, Demirci U. Biotunable acoustic node assembly of organoids. Adv Healthc Mater 2015; 4:1937-43. [PMID: 26149464 PMCID: PMC4731612 DOI: 10.1002/adhm.201500279] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/21/2015] [Indexed: 11/06/2022]
Abstract
Bioengineering of 3D microtissues from cell spheroids is demonstrated by employing the vibration of acoustic standing waves and its hydrodynamic effect at the bottom of a liquid-carrier chamber. A large number of cell spheroids (>10(4) ) are assembled in seconds into a closely packed structure in a scaffold-free fashion under nodal pattern of the standing waves in a fluidic environment.
Collapse
Affiliation(s)
- Pu Chen
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304
| | - Sinan Güven
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304
| | - Osman Berk Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114
| | - Martin L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, MA, 02114
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, NJ 08854
| | - Utkan Demirci
- Bio-Acoustic MEMS in Medicine (BAMM) Lab, Canary Center at Stanford for Early Cancer Detection, Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA 94304
| |
Collapse
|
42
|
Prodanov L, Jindal R, Bale SS, Hegde M, McCarty WJ, Golberg I, Bhushan A, Yarmush ML, Usta OB. Long-term maintenance of a microfluidic 3D human liver sinusoid. Biotechnol Bioeng 2015; 113:241-6. [PMID: 26152452 DOI: 10.1002/bit.25700] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/24/2015] [Accepted: 07/02/2015] [Indexed: 12/21/2022]
Abstract
The development of long-term human organotypic liver-on-a-chip models for successful prediction of toxic response is one of the most important and urgent goals of the NIH/DARPA's initiative to replicate and replace chronic and acute drug testing in animals. For this purpose, we developed a microfluidic chip that consists of two microfluidic chambers separated by a porous membrane. The aim of this communication is to demonstrate the recapitulation of a liver sinusoid-on-a-chip, using human cells only for a period of 28 days. Using a step-by-step method for building a 3D microtissue on-a-chip, we demonstrate that an organotypic in vitro model that reassembles the liver sinusoid microarchitecture can be maintained successfully for a period of 28 days. In addition, higher albumin synthesis (synthetic) and urea excretion (detoxification) were observed under flow compared to static cultures. This human liver-on-a-chip should be further evaluated in drug-related studies.
Collapse
Affiliation(s)
- Ljupcho Prodanov
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - Rohit Jindal
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - Shyam Sundhar Bale
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - Manjunath Hegde
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - William J McCarty
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - Inna Golberg
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - Abhinav Bhushan
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts
| | - Martin L Yarmush
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts. .,Department of Biomedical Engineering, Rutgers University, 599 Taylor Rd., Piscataway, 08854, New Jersey.
| | - Osman Berk Usta
- Center for Engineering in Medicine at Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, 51 Blossom St., Boston, 02144, Massachusetts.
| |
Collapse
|