1
|
He Y, Gao M, Zhu X, Peng W, Zhou Y, Cheng J, Bai L, Bao J. Large-Scale Formation and Long-Term Culture of Hepatocyte Organoids From Streamlined In Vivo Genome-Edited GGTA1 -/- Pigs for Bioartificial Liver Applications. Xenotransplantation 2024; 31:e12878. [PMID: 39166823 DOI: 10.1111/xen.12878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/13/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Hepatocyte transplantation and bioartificial liver (BAL) systems hold significant promise as less invasive alternatives to traditional transplantation, providing crucial temporary support for patients with acute and chronic liver failure. Although human hepatocytes are ideal, their use is limited by ethical concerns and donor availability, leading to the use of porcine hepatocytes in BAL systems due to their functional similarities. Recent advancements in gene-editing technology have improved porcine organ xenotransplantation clinical trials by addressing immune rejection issues. Gene-edited pigs, such as alpha-1,3-galactosyltransferase (GGTA1) knockout pigs, offer a secure source of primary cells for BAL systems. Our research focuses on optimizing the safety and functionality of porcine primary hepatocytes during large-scale cultivation. We achieved this by creating GGTA1 knockout pigs through one-step delivery of CRISPR/Cas9 to pig zygotes via oviduct injection of rAAV, and enhancing hepatocyte viability and function by co-culturing hepatocytes with Roof plate-specific spondin 1 overexpressing HUVECs (R-HUVECs). Using a Rocker culture system, approximately 1010 primary porcine hepatocytes and R-HUVECs rapidly formed organoids with a diameter of 92.1 ± 28.1 µm within 24 h. These organoids not only maintained excellent functionality but also supported partial hepatocyte self-renewal during long-term culture over 28 days. Gene-edited primary porcine hepatocyte organoids will significantly advance the applications of hepatocyte transplantation and BAL systems.
Collapse
Affiliation(s)
- Yuting He
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Mengyu Gao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xinglong Zhu
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wanliu Peng
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yanyan Zhou
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ji Bao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Jabri A, Khan J, Taftafa B, Alsharif M, Mhannayeh A, Chinnappan R, Alzhrani A, Kazmi S, Mir MS, Alsaud AW, Yaqinuddin A, Assiri AM, AlKattan K, Vashist YK, Broering DC, Mir TA. Bioengineered Organoids Offer New Possibilities for Liver Cancer Studies: A Review of Key Milestones and Challenges. Bioengineering (Basel) 2024; 11:346. [PMID: 38671768 PMCID: PMC11048289 DOI: 10.3390/bioengineering11040346] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Hepatic cancer is widely regarded as the leading cause of cancer-related mortality worldwide. Despite recent advances in treatment options, the prognosis of liver cancer remains poor. Therefore, there is an urgent need to develop more representative in vitro models of liver cancer for pathophysiology and drug screening studies. Fortunately, an exciting new development for generating liver models in recent years has been the advent of organoid technology. Organoid models hold huge potential as an in vitro research tool because they can recapitulate the spatial architecture of primary liver cancers and maintain the molecular and functional variations of the native tissue counterparts during long-term culture in vitro. This review provides a comprehensive overview and discussion of the establishment and application of liver organoid models in vitro. Bioengineering strategies used to construct organoid models are also discussed. In addition, the clinical potential and other relevant applications of liver organoid models in different functional states are explored. In the end, this review discusses current limitations and future prospects to encourage further development.
Collapse
Affiliation(s)
- Abdullah Jabri
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Jibran Khan
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Bader Taftafa
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Mohamed Alsharif
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Abdulaziz Mhannayeh
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Raja Chinnappan
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Alaa Alzhrani
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21423, Saudi Arabia
| | - Shadab Kazmi
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- Pathology and laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mohammad Shabab Mir
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh 147301, Punjab, India;
| | - Aljohara Waleed Alsaud
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
| | - Abdullah M. Assiri
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Yogesh K. Vashist
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Dieter C. Broering
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Tanveer Ahmad Mir
- College of Medicine, Alfaisal University, Riyadh 11211, Saudi Arabia (R.C.); (A.W.A.); (K.A.)
- Tissue/Organ Bioengineering and BioMEMS Lab, Organ Transplant Centre of Excellence (TR&I Dpt), King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
3
|
Ahmadian M, Hosseini S, Alipour A, Jahanfar M, Farrokhi N, Homaeigohar S, Shahsavarani H. In vitro modeling of hepatocellular carcinoma niche on decellularized tomato thorny leaves: a novel natural three-dimensional (3D) scaffold for liver cancer therapeutics. Front Bioeng Biotechnol 2023; 11:1189726. [PMID: 37251569 PMCID: PMC10212619 DOI: 10.3389/fbioe.2023.1189726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Liver cancer is now one of the main causes leading to death worldwide. To achieve reliable therapeutic effects, it is crucial to develop efficient approaches to test novel anticancer drugs. Considering the significant contribution of tumor microenvironment to cell's response to medications, in vitro 3D bioinspiration of cancer cell niches can be regarded as an advanced strategy to improve the accuracy and reliability of the drug-based treatment. In this regard, decellularized plant tissues can perform as suitable 3D scaffolds for mammalian cell culture to create a near-to-real condition to test drug efficacy. Here, we developed a novel 3D natural scaffold made from decellularized tomato hairy leaves (hereafter called as DTL) to mimic the microenvironment of human hepatocellular carcinoma (HCC) for pharmaceutical purposes. The surface hydrophilicity, mechanical properties, and topography measurement and molecular analyses revealed that the 3D DTL scaffold is an ideal candidate for liver cancer modeling. The cells exhibited a higher growth and proliferation rate within the DTL scaffold, as verified by quantifying the expression of related genes, DAPI staining, and SEM imaging of the cells. Moreover, prilocaine, an anticancer drug, showed a higher effectiveness against the cancer cells cultured on the 3D DTL scaffold, compared to a 2D platform. Taken together, this new cellulosic 3D scaffold can be confidently proposed for chemotherapeutic testing of drugs on hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mariye Ahmadian
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Jahanfar
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Naser Farrokhi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shahin Homaeigohar
- School of Science and Engineering, University of Dundee, Dundee, United Kingdom
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| |
Collapse
|
4
|
McDuffie D, Barr D, Helm M, Baumert T, Agarwal A, Thomas E. Physiomimetic In Vitro Human Models for Viral Infection in the Liver. Semin Liver Dis 2023; 43:31-49. [PMID: 36402129 PMCID: PMC10005888 DOI: 10.1055/a-1981-5944] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Viral hepatitis is a leading cause of liver morbidity and mortality globally. The mechanisms underlying acute infection and clearance, versus the development of chronic infection, are poorly understood. In vitro models of viral hepatitis circumvent the high costs and ethical considerations of animal models, which also translate poorly to studying the human-specific hepatitis viruses. However, significant challenges are associated with modeling long-term infection in vitro. Differentiated hepatocytes are best able to sustain chronic viral hepatitis infection, but standard two-dimensional models are limited because they fail to mimic the architecture and cellular microenvironment of the liver, and cannot maintain a differentiated hepatocyte phenotype over extended periods. Alternatively, physiomimetic models facilitate important interactions between hepatocytes and their microenvironment by incorporating liver-specific environmental factors such as three-dimensional ECM interactions and co-culture with non-parenchymal cells. These physiologically relevant interactions help maintain a functional hepatocyte phenotype that is critical for sustaining viral hepatitis infection. In this review, we provide an overview of distinct, novel, and innovative in vitro liver models and discuss their functionality and relevance in modeling viral hepatitis. These platforms may provide novel insight into mechanisms that regulate viral clearance versus progression to chronic infections that can drive subsequent liver disease.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Madeline Helm
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Thomas Baumert
- Inserm Research Institute for Viral and Liver Diseases, University of Strasbourg, Strasbourg, France
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
5
|
Seidemann L, Prinz S, Scherbel JC, Götz C, Seehofer D, Damm G. Optimization of extracellular matrix for primary human hepatocyte cultures using mixed collagen-Matrigel matrices. EXCLI JOURNAL 2023; 22:12-34. [PMID: 36660192 PMCID: PMC9837384 DOI: 10.17179/excli2022-5459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 11/17/2022] [Indexed: 01/21/2023]
Abstract
Loss of differentiation of primary human hepatocytes (PHHs) ex vivo is a known problem of in vitro liver models. Culture optimizations using collagen type I and Matrigel reduce the dedifferentiation process but are not able to prevent it. While neither of these extracellular matrices (ECMs) on their own correspond to the authentic hepatic ECM, a combination of them could more closely resemble the in vivo situation. Our study aimed to systematically analyze the influence of mixed matrices composed of collagen type I and Matrigel on the maintenance and reestablishment of hepatic functions. Therefore, PHHs were cultured on mixed collagen-Matrigel matrices in monolayer and sandwich cultures and viability, metabolic capacity, differentiation markers, cellular arrangement and the cells' ability to repolarize and form functional bile canaliculi were assessed by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), functional assays and immunofluorescence microscopy. Our results show that mixed matrices were superior to pure matrices in maintaining metabolic capacity and hepatic differentiation. In contrast, Matrigel supplementation can impair the development of a proper hepatocytic polarization. Our systematic study helps to compose an optimized ECM to maintain and reestablish hepatic differentiation on cellular and multicellular levels in human liver models.
Collapse
Affiliation(s)
- Lena Seidemann
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Sarah Prinz
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Jan-Constantin Scherbel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Christina Götz
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany,Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany,*To whom correspondence should be addressed: Georg Damm, Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Liebigstr. 20, 04103 Leipzig, Germany; Tel.: +49-341-9739656, E-mail:
| |
Collapse
|
6
|
Trossbach M, de Lucas Sanz M, Seashore-Ludlow B, Joensson HN. A Portable, Negative-Pressure Actuated, Dynamically Tunable Microfluidic Droplet Generator. MICROMACHINES 2022; 13:1823. [PMID: 36363843 PMCID: PMC9697964 DOI: 10.3390/mi13111823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/12/2022] [Accepted: 10/21/2022] [Indexed: 06/16/2023]
Abstract
Droplet microfluidics utilize a monodisperse water-in-oil emulsion, with an expanding toolbox offering a wide variety of operations on a range of droplet sizes at high throughput. However, translation of these capabilities into applications for non-expert laboratories to fully harness the inherent potential of microscale manipulations is woefully trailing behind. One major obstacle is that droplet microfluidic setups often rely on custom fabricated devices, costly liquid actuators, and are not easily set up and operated by non-specialists. This impedes wider adoption of droplet technologies in, e.g., the life sciences. Here, we demonstrate an easy-to-use minimal droplet production setup with a small footprint, built exclusively from inexpensive commercially sourced parts, powered and controlled by a laptop. We characterize the components of the system and demonstrate production of droplets ranging in volume from 3 to 21 nL in a single microfluidic device. Furthermore, we describe the dynamic tuning of droplet composition. Finally, we demonstrate the production of droplet-templated cell spheroids from primary cells, where the mobility and simplicity of the setup enables its use within a biosafety cabinet. Taken together, we believe this minimal droplet setup is ideal to drive broad adoption of droplet microfluidics technology.
Collapse
Affiliation(s)
- Martin Trossbach
- KTH Royal Institute of Technology & Science for Life Laboratory, 17165 Solna, Sweden
| | - Marta de Lucas Sanz
- KTH Royal Institute of Technology & Science for Life Laboratory, 17165 Solna, Sweden
| | | | - Haakan N. Joensson
- KTH Royal Institute of Technology & Science for Life Laboratory, 17165 Solna, Sweden
| |
Collapse
|
7
|
McDuffie D, Barr D, Agarwal A, Thomas E. Physiologically relevant microsystems to study viral infection in the human liver. Front Microbiol 2022; 13:999366. [PMID: 36246284 PMCID: PMC9555087 DOI: 10.3389/fmicb.2022.999366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Viral hepatitis is a leading cause of liver disease and mortality. Infection can occur acutely or chronically, but the mechanisms that govern the clearance of virus or lack thereof are poorly understood and merit further investigation. Though cures for viral hepatitis have been developed, they are expensive, not readily accessible in vulnerable populations and some patients may remain at an increased risk of developing hepatocellular carcinoma (HCC) even after viral clearance. To sustain infection in vitro, hepatocytes must be fully mature and remain in a differentiated state. However, primary hepatocytes rapidly dedifferentiate in conventional 2D in vitro platforms. Physiologically relevant or physiomimetic microsystems, are increasingly popular alternatives to traditional two-dimensional (2D) monocultures for in vitro studies. Physiomimetic systems reconstruct and incorporate elements of the native cellular microenvironment to improve biologic functionality in vitro. Multiple elements contribute to these models including ancillary tissue architecture, cell co-cultures, matrix proteins, chemical gradients and mechanical forces that contribute to increased viability, longevity and physiologic function for the tissue of interest. These microsystems are used in a wide variety of applications to study biological phenomena. Here, we explore the use of physiomimetic microsystems as tools for studying viral hepatitis infection in the liver and how the design of these platforms is tailored for enhanced investigation of the viral lifecycle when compared to conventional 2D cell culture models. Although liver-based physiomimetic microsystems are typically applied in the context of drug studies, the platforms developed for drug discovery purposes offer a solid foundation to support studies on viral hepatitis. Physiomimetic platforms may help prolong hepatocyte functionality in order to sustain chronic viral hepatitis infection in vitro for studying virus-host interactions for prolonged periods.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - David Barr
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmanuel Thomas
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
8
|
Pluta KD, Ciezkowska M, Wisniewska M, Wencel A, Pijanowska DG. Cell-based clinical and experimental methods for assisting the function of impaired livers – Present and future of liver support systems. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
9
|
Peng WC, Kraaier LJ, Kluiver TA. Hepatocyte organoids and cell transplantation: What the future holds. Exp Mol Med 2021; 53:1512-1528. [PMID: 34663941 PMCID: PMC8568948 DOI: 10.1038/s12276-021-00579-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/29/2022] Open
Abstract
Historically, primary hepatocytes have been difficult to expand or maintain in vitro. In this review, we will focus on recent advances in establishing hepatocyte organoids and their potential applications in regenerative medicine. First, we provide a background on the renewal of hepatocytes in the homeostatic as well as the injured liver. Next, we describe strategies for establishing primary hepatocyte organoids derived from either adult or fetal liver based on insights from signaling pathways regulating hepatocyte renewal in vivo. The characteristics of these organoids will be described herein. Notably, hepatocyte organoids can adopt either a proliferative or a metabolic state, depending on the culture conditions. Furthermore, the metabolic gene expression profile can be modulated based on the principles that govern liver zonation. Finally, we discuss the suitability of cell replacement therapy to treat different types of liver diseases and the current state of cell transplantation of in vitro-expanded hepatocytes in mouse models. In addition, we provide insights into how the regenerative microenvironment in the injured host liver may facilitate donor hepatocyte repopulation. In summary, transplantation of in vitro-expanded hepatocytes holds great potential for large-scale clinical application to treat liver diseases.
Collapse
Affiliation(s)
- Weng Chuan Peng
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands.
| | - Lianne J Kraaier
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| | - Thomas A Kluiver
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, The Netherlands
| |
Collapse
|
10
|
Sphabmixay P, Raredon MSB, Wang AJS, Lee H, Hammond PT, Fang NX, Griffith LG. High resolution stereolithography fabrication of perfusable scaffolds to enable long-term meso-scale hepatic culture for disease modeling. Biofabrication 2021; 13. [PMID: 34479229 DOI: 10.1088/1758-5090/ac23aa] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/03/2021] [Indexed: 12/18/2022]
Abstract
Microphysiological systems (MPS), comprising human cell cultured in formats that capture features of the three-dimensional (3D) microenvironments of native human organs under microperfusion, are promising tools for biomedical research. Here we report the development of a mesoscale physiological system (MePS) enabling the long-term 3D perfused culture of primary human hepatocytes at scales of over 106cells per MPS. A central feature of the MePS, which employs a commercially-available multiwell bioreactor for perfusion, is a novel scaffold comprising a dense network of nano- and micro-porous polymer channels, designed to provide appropriate convective and diffusive mass transfer of oxygen and other nutrients while maintaining physiological values of shear stress. The scaffold design is realized by a high resolution stereolithography fabrication process employing a novel resin. This new culture system sustains mesoscopic hepatic tissue-like cultures with greater hepatic functionality (assessed by albumin and urea synthesis, and CYP3A4 activity) and lower inflammation markers compared to comparable cultures on the commercial polystyrene scaffold. To illustrate applications to disease modeling, we established an insulin-resistant phenotype by exposing liver cells to hyperglycemic and hyperinsulinemic media. Future applications of the MePS include the co-culture of hepatocytes with resident immune cells and the integration with multiple organs to model complex liver-associated diseases.
Collapse
Affiliation(s)
- Pierre Sphabmixay
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America.,Whitehead Institute of Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States of America.,Vascular Biology and Therapeutics, Yale University, New Haven, CT, United States of America
| | - Alex J-S Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Howon Lee
- Department of Mechanical Engineering, Seoul National University, Seoul, Korea
| | - Paula T Hammond
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Nicholas X Fang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Linda G Griffith
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States of America.,Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| |
Collapse
|
11
|
Heydari Z, Zarkesh I, Ghanian MH, Aghdaei MH, Kotova S, Zahmatkesh E, Farzaneh Z, Piryaei A, Akbarzadeh I, Shpichka A, Gramignoli R, Timashev P, Baharvand H, Vosough M. Biofabrication of size-controlled liver microtissues incorporated with ECM-derived microparticles to prolong hepatocyte function. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00137-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Milner E, Ainsworth M, McDonough M, Stevens B, Buehrer J, Delzell R, Wilson C, Barnhill J. Emerging Three-Dimensional Hepatic Models in Relation to Traditional Two-Dimensional In Vitro Assays for Evaluating Drug Metabolism and Hepatoxicity. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
13
|
Wencel A, Ciezkowska M, Wisniewska M, Zakrzewska KE, Pijanowska DG, Pluta KD. Effects of genetically modified human skin fibroblasts, stably overexpressing hepatocyte growth factor, on hepatic functions of cocultured C3A cells. Biotechnol Bioeng 2020; 118:72-81. [PMID: 32880912 DOI: 10.1002/bit.27551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/04/2020] [Accepted: 09/01/2020] [Indexed: 01/18/2023]
Abstract
Diseases leading to terminal hepatic failure are among the most common causes of death worldwide. Transplant of the whole organ is the only effective method to cure liver failure. Unfortunately, this treatment option is not available universally due to the serious shortage of donors. Thus, alternative methods have been developed that are aimed at prolonging the life of patients, including hepatic cells transplantation and bridging therapy based on hybrid bioartificial liver devices. Parenchymal liver cells are highly differentiated and perform many complex functions, such as detoxification and protein synthesis. Unfortunately, isolated hepatocytes display a rapid decline in viability and liver-specific functions. A number of methods have been developed to maintain hepatocytes in their highly differentiated state in vitro, amongst them the most promising being 3D growth scaffolds and decellularized tissues or coculture with other cell types required for the heterotypic cell-cell interactions. Here we present a novel approach to the hepatic cells culture based on the feeder layer cells genetically modified using lentiviral vector to stably produce additional amounts of hepatocyte growth factor and show the positive influence of these coculture conditions on the preservation of the hepatic functions of the liver parenchymal cells' model-C3A cells.
Collapse
Affiliation(s)
- Agnieszka Wencel
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Ciezkowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Wisniewska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Karolina E Zakrzewska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland.,Institute of Fundamental Technological Research, Polish Academy of Sciences, Warsaw, Poland
| | - Dorota G Pijanowska
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof D Pluta
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
14
|
Chen X, Zhang YS, Zhang X, Liu C. Organ-on-a-chip platforms for accelerating the evaluation of nanomedicine. Bioact Mater 2020; 6:1012-1027. [PMID: 33102943 PMCID: PMC7566214 DOI: 10.1016/j.bioactmat.2020.09.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/22/2020] [Indexed: 02/07/2023] Open
Abstract
Nanomedicine involves the use of engineered nanoscale materials in an extensive range of diagnostic and therapeutic applications and can be applied to the treatment of many diseases. Despite the rapid progress and tremendous potential of nanomedicine in the past decades, the clinical translational process is still quite slow, owing to the difficulty in understanding, evaluating, and predicting nanomaterial behaviors within the complex environment of human beings. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to resolve these challenges. Sophisticatedly designed Organ Chip enable in vitro simulation of the in vivo microenvironments, thus providing robust platforms for evaluating nanomedicine. Herein, we review recent developments and achievements in Organ Chip models for nanomedicine evaluations, categorized into seven broad sections based on the target organ systems: respiratory, digestive, lymphatic, excretory, nervous, and vascular, as well as coverage on applications relating to cancer. We conclude by providing our perspectives on the challenges and potential future directions for applications of Organ Chip in nanomedicine. Microfluidics-based organ-on-a-chip (Organ Chip) techniques offer a promising way to understand, evaluate, and predict nanomedicine behaviors within the complex environment. Organ Chip models for nanomedicine evaluations are categorized into seven broad sections based on the targeted body systems. Limitations, challenges, and perspectives of Organ Chip for accelerating the assessment of nanomedicine are discussed, respectively.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, United States
| | - Xinping Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, Frontiers Science Center for Materiobiology and Dynamic Chemistry, Engineering Research Center for Biomaterials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, PR China
| |
Collapse
|
15
|
Oleaga C, Bridges LR, Persaud K, McAleer CW, Long CJ, Hickman JJ. A functional long-term 2D serum-free human hepatic in vitro system for drug evaluation. Biotechnol Prog 2020; 37:e3069. [PMID: 32829524 DOI: 10.1002/btpr.3069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/13/2020] [Accepted: 08/20/2020] [Indexed: 01/05/2023]
Abstract
Human in vitro hepatic models generate faster drug toxicity data with higher human predictability compared to animal models. However, for long-term studies, current models require the use of serum and 3D architecture, limiting their utility. Maintaining a functional long-term human in vitro hepatic culture that avoids complex structures and serum would improve the value of such systems for preclinical studies. This would also enable a more straightforward integration with current multi-organ devices to study human systemic toxicity to generate an alternative model to chronic animal evaluations. A human primary hepatocyte culture system was characterized for 28 days in 2D and serum-free defined conditions. Under the studied conditions, human primary hepatocytes maintained their characteristic morphology, hepatic markers and functions for 28 days. The acute and chronic administration of known drugs validated the sensitivity of the system for drug testing. This human 2D model represents a realistic system to evaluate hepatic function for long-term drug studies, without the need of animal serum, confounding variable in most models, and with less complexity and resultant cost compared to most 3D models. The defined culture conditions can easily be integrated into complex multi-organ in vitro models for studying systemic effects driven by the liver function for long-term evaluations.
Collapse
Affiliation(s)
- Carlota Oleaga
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - L Richard Bridges
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - Keisha Persaud
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | | | - Christopher J Long
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
16
|
Granz CL, Gorji A. Dental stem cells: The role of biomaterials and scaffolds in developing novel therapeutic strategies. World J Stem Cells 2020; 12:897-921. [PMID: 33033554 PMCID: PMC7524692 DOI: 10.4252/wjsc.v12.i9.897] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/05/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023] Open
Abstract
Dental stem cells (DSCs) are self-renewable cells that can be obtained easily from dental tissues, and are a desirable source of autologous stem cells. The use of DSCs for stem cell transplantation therapeutic approaches is attractive due to their simple isolation, high plasticity, immunomodulatory properties, and multipotential abilities. Using appropriate scaffolds loaded with favorable biomolecules, such as growth factors, and cytokines, can improve the proliferation, differentiation, migration, and functional capacity of DSCs and can optimize the cellular morphology to build tissue constructs for specific purposes. An enormous variety of scaffolds have been used for tissue engineering with DSCs. Of these, the scaffolds that particularly mimic tissue-specific micromilieu and loaded with biomolecules favorably regulate angiogenesis, cell-matrix interactions, degradation of extracellular matrix, organized matrix formation, and the mineralization abilities of DSCs in both in vitro and in vivo conditions. DSCs represent a promising cell source for tissue engineering, especially for tooth, bone, and neural tissue restoration. The purpose of the present review is to summarize the current developments in the major scaffolding approaches as crucial guidelines for tissue engineering using DSCs and compare their effects in tissue and organ regeneration.
Collapse
Affiliation(s)
- Cornelia Larissa Granz
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| | - Ali Gorji
- Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster 48149, Germany
| |
Collapse
|
17
|
Ruoß M, Vosough M, Königsrainer A, Nadalin S, Wagner S, Sajadian S, Huber D, Heydari Z, Ehnert S, Hengstler JG, Nussler AK. Towards improved hepatocyte cultures: Progress and limitations. Food Chem Toxicol 2020; 138:111188. [PMID: 32045649 DOI: 10.1016/j.fct.2020.111188] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/31/2020] [Accepted: 02/07/2020] [Indexed: 12/14/2022]
Abstract
Hepatotoxicity is among the most frequent reasons for drug withdrawal from the market. Therefore, there is an urgent need for reliable predictive in vitro tests, which unfailingly identify hepatotoxic drug candidates, reduce drug development time, expenses and the number of test animals. Currently, human hepatocytes represent the gold standard. However, the use of hepatocytes is challenging since the cells are not constantly available and lose their metabolic activity in culture. To solve these problems many different approaches have been developed in the past decades. The aim of this review is to present these approaches and to discuss the possibilities and limitations as well as future opportunities and directions.
Collapse
Affiliation(s)
- Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Alfred Königsrainer
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Silvia Wagner
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Sahar Sajadian
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Diana Huber
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Zahra Heydari
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sabrina Ehnert
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo), Technical University of Dortmund, Dortmund, Germany
| | - Andreas K Nussler
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University Tübingen, Tübingen, Germany.
| |
Collapse
|
18
|
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
|
19
|
Tasnim F, Singh NH, Tan EKF, Xing J, Li H, Hissette S, Manesh S, Fulwood J, Gupta K, Ng CW, Xu S, Hill J, Yu H. Tethered primary hepatocyte spheroids on polystyrene multi-well plates for high-throughput drug safety testing. Sci Rep 2020; 10:4768. [PMID: 32179810 PMCID: PMC7075904 DOI: 10.1038/s41598-020-61699-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 02/19/2020] [Indexed: 12/26/2022] Open
Abstract
Hepatocyte spheroids are useful models for mimicking liver phenotypes in vitro because of their three-dimensionality. However, the lack of a biomaterial platform which allows the facile manipulation of spheroid cultures on a large scale severely limits their application in automated high-throughput drug safety testing. In addition, there is not yet a robust way of controlling spheroid size, homogeneity and integrity during extended culture. This work addresses these bottlenecks to the automation of hepatocyte spheroid culture by tethering 3D hepatocyte spheroids directly onto surface-modified polystyrene (PS) multi-well plates. However, polystyrene surfaces are inert toward functionalization, and this makes the uniform conjugation of bioactive ligands very challenging. Surface modification of polystyrene well plates is achieved herein using a three-step sequence, resulting in a homogeneous distribution of bioactive RGD and galactose ligands required for spheroid tethering and formation. Importantly, treatment of polystyrene tethered spheroids with vehicle and paradigm hepatotoxicant (chlorpromazine) treatment using an automated liquid handling platform shows low signal deviation, intact 3D spheroidal morphology and Z’ values above 0.5, and hence confirming their amenability to high-throughput automation. Functional analyses performance (i.e. urea and albumin production, cytochrome P450 activity and induction studies) of the polystyrene tethered spheroids reveal significant improvements over hepatocytes cultured as collagen monolayers. This is the first demonstration of automated hepatotoxicant treatment on functional 3D hepatocyte spheroids tethered directly on polystyrene multi-well plates, and will serve as an important advancement in the application of 3D tethered spheroid models to high throughput drug screening.
Collapse
Affiliation(s)
- Farah Tasnim
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Nisha Hari Singh
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Elijah Keng Foo Tan
- Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore.,Yong Loo Lin School of Medicine (Department of Physiology) and Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
| | - Jiangwa Xing
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Huan Li
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Sebastien Hissette
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Sravanthy Manesh
- Experimental Therapeutics Centre (ETC), Level 3, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Justina Fulwood
- Experimental Therapeutics Centre (ETC), Level 3, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Kapish Gupta
- Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Chan Way Ng
- Yong Loo Lin School of Medicine (Department of Physiology) and Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
| | - Shuoyu Xu
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore
| | - Jeffrey Hill
- Experimental Therapeutics Centre (ETC), Level 3, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore.,Sussex Drug Discovery Centre, School of Life Sciences, University of Sussex, Brighton, BN19RH, UK
| | - Hanry Yu
- Institute of Bioengineering and Nanotechnology, #04-01, 31 Biopolis Way, The Nanos, Singapore, 138669, Singapore. .,Mechanobiology Institute, T-Labs, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore. .,Yong Loo Lin School of Medicine (Department of Physiology) and Graduate School for Integrative Sciences & Engineering (NGS), National University of Singapore, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore. .,CAMP IRG, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Enterprise Wing, Level 4, Singapore, 138602, Singapore.
| |
Collapse
|
20
|
Ruoß M, Rebholz S, Weimer M, Grom-Baumgarten C, Athanasopulu K, Kemkemer R, Käß H, Ehnert S, Nussler AK. Development of Scaffolds with Adjusted Stiffness for Mimicking Disease-Related Alterations of Liver Rigidity. J Funct Biomater 2020; 11:17. [PMID: 32183326 PMCID: PMC7151584 DOI: 10.3390/jfb11010017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022] Open
Abstract
Drug-induced liver toxicity is one of the most common reasons for the failure of drugs in clinical trials and frequent withdrawal from the market. Reasons for such failures include the low predictive power of in vivo studies, that is mainly caused by metabolic differences between humans and animals, and intraspecific variances. In addition to factors such as age and genetic background, changes in drug metabolism can also be caused by disease-related changes in the liver. Such metabolic changes have also been observed in clinical settings, for example, in association with a change in liver stiffness, a major characteristic of an altered fibrotic liver. For mimicking these changes in an in vitro model, this study aimed to develop scaffolds that represent the rigidity of healthy and fibrotic liver tissue. We observed that liver cells plated on scaffolds representing the stiffness of healthy livers showed a higher metabolic activity compared to cells plated on stiffer scaffolds. Additionally, we detected a positive effect of a scaffold pre-coated with fetal calf serum (FCS)-containing media. This pre-incubation resulted in increased cell adherence during cell seeding onto the scaffolds. In summary, we developed a scaffold-based 3D model that mimics liver stiffness-dependent changes in drug metabolism that may more easily predict drug interaction in diseased livers.
Collapse
Affiliation(s)
- Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Silas Rebholz
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Marina Weimer
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany; (K.A.); (R.K.)
| | - Carl Grom-Baumgarten
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Kiriaki Athanasopulu
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany; (K.A.); (R.K.)
| | - Ralf Kemkemer
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany; (K.A.); (R.K.)
| | - Hanno Käß
- Faculty of Basic Science, University of Applied Sciences Esslingen, 73728 Esslingen am Neckar, Germany;
| | - Sabrina Ehnert
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| | - Andreas K. Nussler
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany; (S.R.); (M.W.); (C.G.-B.); (S.E.); (A.K.N.)
| |
Collapse
|
21
|
Ruoß M, Kieber V, Rebholz S, Linnemann C, Rinderknecht H, Häussling V, Häcker M, Olde Damink LHH, Ehnert S, Nussler AK. Cell-Type-Specific Quantification of a Scaffold-Based 3D Liver Co-Culture. Methods Protoc 2019; 3:1. [PMID: 31878071 PMCID: PMC7189675 DOI: 10.3390/mps3010001] [Citation(s) in RCA: 9] [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: 06/13/2019] [Revised: 12/18/2019] [Accepted: 12/18/2019] [Indexed: 12/25/2022] Open
Abstract
In order to increase the metabolic activity of human hepatocytes and liver cancer cell lines, many approaches have been reported in recent years. The metabolic activity could be increased mainly by cultivating the cells in 3D systems or co-cultures (with other cell lines). However, if the system becomes more complex, it gets more difficult to quantify the number of cells (e.g., on a 3D matrix). Until now, it has been impossible to quantify different cell types individually in 3D co-culture systems. Therefore, we developed a PCR-based method that allows the quantification of HepG2 cells and 3T3-J2 cells separately in a 3D scaffold culture. Moreover, our results show that this method allows better comparability between 2D and 3D cultures in comparison to the often-used approaches based on metabolic activity measurements, such as the conversion of resazurin.
Collapse
Affiliation(s)
- Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Vanessa Kieber
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Silas Rebholz
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Caren Linnemann
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Helen Rinderknecht
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Victor Häussling
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Marina Häcker
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | | | - Sabrina Ehnert
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| | - Andreas K. Nussler
- Department of Traumatology, Siegfried Weller Institute, BG-Klinik Tübingen, Eberhard Karls University, 72076 Tübingen, Germany; (V.K.); (S.R.); (C.L.); (H.R.); (V.H.); (M.H.); (S.E.); (A.K.N.)
| |
Collapse
|
22
|
Ide I, Nagao E, Kajiyama S, Mizoguchi N. A novel evaluation method for determining drug-induced hepatotoxicity using 3D bio-printed human liver tissue. Toxicol Mech Methods 2019; 30:189-196. [PMID: 31736396 DOI: 10.1080/15376516.2019.1686795] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Predicting drug-induced liver injury is important in early stage drug discovery; however, an accurate prediction with existing hepatotoxicity evaluation tools is difficult. Conventional monolayer (2D) cultures have short viabilities and are therefore inappropriate for performing long-term toxicity tests. Conventionally used 200-μm spheroids also have toxicity detection limits. The goal of this study was to develop a humanized liver tissue capable of evaluating long-term toxicity with high sensitivity. Spheroids consisting of co-cultured cryopreserved primary human hepatocytes and human hepatic stellate cells were developed using a 3D bio-printer. The "3D bio-printed liver tissue", of ∼1 mm, was then used for long-term viability assessments (over 25 days) based on ATP, albumin, and urea levels. Hepatotoxicity evaluation was performed by analyzing the expression of genes involved in drug metabolism and transport over a 2-week drug exposure period. The 3D bio-printed liver tissue showed improved viability and enhanced gene expression of enzymes related to drug metabolism and transport, as compared to the controls. Additionally, the 3D bio-printed liver tissue demonstrated a high sensitivity for hepatotoxicity evaluation when combined with pathological evaluation and measurements for ATP production, and secretion of albumin and urea. In conclusion, the 3D bio-printed liver tissue was able to detect the toxicity of compounds that was, otherwise, undetected by 2D culture and conventionally used spheroids. These findings demonstrate a 3D bio-printed liver tissue with increased accuracy of hepatotoxicity prediction in the early stages of drug discovery, as compared to currently available methods.
Collapse
Affiliation(s)
- Izumi Ide
- Department of Drug Discovery Platform, Cyfuse Biomedical K.K., University of Tokyo, Tokyo, Japan
| | - Eri Nagao
- Department of Drug Discovery Platform, Cyfuse Biomedical K.K., University of Tokyo, Tokyo, Japan
| | - Sakura Kajiyama
- Department of Drug Discovery Platform, Cyfuse Biomedical K.K., University of Tokyo, Tokyo, Japan
| | - Natsumi Mizoguchi
- Department of Drug Discovery Platform, Cyfuse Biomedical K.K., University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Pászti-Gere E, Szombath G, Gütschow M, Steinmetzer T, Székács A. 3-Amidinophenylalanine-derived matriptase inhibitors can modulate hepcidin production in vitro. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:511-520. [PMID: 31659405 PMCID: PMC7280348 DOI: 10.1007/s00210-019-01743-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 09/25/2019] [Indexed: 02/07/2023]
Abstract
Matriptase-2 (MT-2) is a type II transmembrane serine protease and predominantly attached to the surface of hepatocytes. MT-2 decreases the production of hepcidin, a key regulator of iron homeostasis. In this study, the effects of four 3-amidinophenylalanine-derived combined matriptase-1/matriptase-2 (MT-1/2) inhibitors (MI-432, MI-441, MI-460, and MI-461) on hepcidin production were investigated in hepatocyte mono- and hepatocyte-Kupffer cell co-cultures. In MI-461-treated cell cultures, the extracellular hydrogen peroxide contents and the interleukin-6 and -8 (IL-6 and IL-8) levels were determined and compared to controls. Hepcidin overproduction was observed in hepatocytes upon treatment with MI-432, MI-441 and MI-461 at 50 μM. In contrast, extracellular hydrogen peroxide levels were not elevated significantly after matriptase inhibition with MI-461. Furthermore, MI-461 did not induce increases in IL-6 and IL-8 levels in these hepatic models. A model of the binding mode of inhibitor MI-461 in complex with MT-2 revealed numerous polar contacts contributing to the nanomolar potency of this compound. Based on the in vitro data on hepcidin regulation, treatment with MI-461 might be valuable in pathological states of iron metabolism without causing excessive oxidative stress.
Collapse
Affiliation(s)
- Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary.
| | - Gergely Szombath
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, Budapest, Hungary
| | | | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - András Székács
- Agro-Environmental Research Institute, National Agricultural Research and Innovation Centre, Budapest, Hungary
| |
Collapse
|
24
|
Feizi Z, Zahmatkesh E, Farzaneh Z, Piryaei A, Gramignoli R, Nussler AK, Baharvand H, Vosough M. Prenatal liver stromal cells: Favorable feeder cells for long-term culture of hepatic progenitor cells. J Cell Biochem 2019; 120:16624-16633. [PMID: 31081256 DOI: 10.1002/jcb.28921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/02/2019] [Accepted: 04/12/2019] [Indexed: 12/29/2022]
Abstract
Clinical and pharmaceutical applications of primary hepatocytes (PHs) are limited due to inadequate number of donated livers and potential challenges in successful maintenance of PHs in culture. Freshly isolated hepatocytes lose their specific features and rapidly de-differentiate in culture. Bipotent hepatoblasts, as liver precursor cells that can differentiate into both hepatocytes and cholangiocytes (Alb- and Ck19-positive cells, respectively), could be used as an alternative and reliable cell source to produce enough PHs for drug discovery or possible clinical applications. In this study, growth factor-free coculture systems of prenatal or postnatal murine liver stromal cells (pre-LSCs or post-LSCs, respectively) were used as feeder cells to support freshly isolated mice hepatoblasts. DLK1-positive hepatoblasts were isolated from mouse fetuses (E14.5) and cocultured with feeder cells under adherent conditions. The hepatoblasts' bipotent features, proliferation rate, and colony formation capacity were assessed on day 5 and 7 post-seeding. Immunofluorescence staining showed that the hepatoblasts remained double positive for Alb and Ck19 on both Pre- and Post-LSCs, after 5 and 7 days of coculture. Moreover, application of pre-LSCs as feeder cells significantly increased the number of DLK1-positive cells and their proliferation rate (ie, increased the number of Ki-67 positive cells) on day 7, compared to Post-LSCs group. Finally, to address our ultimate goal, which was an extension of hepatoblasts ex vivo maintenance, 3D spheres of isolated hepatoblasts were, cultured in conditioned medium (CM) derived from pre-LSCs until day 30. It was observed that the CM derived from Pre-LSCs could successfully prolong the maintenance of hepatic progenitor cells (HPCs) in 3D suspension culture.
Collapse
Affiliation(s)
- Zahra Feizi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ensieh Zahmatkesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Andreas K Nussler
- Siegfried Weller Institute for Trauma Research, BG Trauma Center Tübingen, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
25
|
Chang JH, Sangaraju D, Liu N, Jaochico A, Plise E. Comprehensive Evaluation of Bile Acid Homeostasis in Human Hepatocyte Co-Culture in the Presence of Troglitazone, Pioglitazone, and Acetylsalicylic Acid. Mol Pharm 2019; 16:4230-4240. [DOI: 10.1021/acs.molpharmaceut.9b00562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Jae H. Chang
- Genentech, Inc, South San Francisco, California 94080, United States
| | - Dewakar Sangaraju
- Genentech, Inc, South San Francisco, California 94080, United States
| | - Ning Liu
- Genentech, Inc, South San Francisco, California 94080, United States
| | - Allan Jaochico
- Genentech, Inc, South San Francisco, California 94080, United States
| | - Emile Plise
- Genentech, Inc, South San Francisco, California 94080, United States
| |
Collapse
|
26
|
Koeda A, Iwao T, Nakanishi A, Mizuno S, Yamashita M, Sakai Y, Nakamura K, Matsunaga T. Comparison of mRNA expression profiles of drug-metabolizing enzymes and transporters in fresh and cryopreserved cynomolgus monkey hepatocytes. Drug Metab Pharmacokinet 2019; 34:253-263. [PMID: 31174977 DOI: 10.1016/j.dmpk.2019.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 03/12/2019] [Accepted: 04/05/2019] [Indexed: 02/08/2023]
Abstract
In this study, freshly isolated and cryopreserved cynomolgus monkey hepatocytes were seeded on Cell-able® plates with feeder cells to form spheroids and were cultured for 28 days. As a control, hepatocytes were also cultured with or without feeder cells on collagen-coated plates. We verified the mRNA expression levels of drug-metabolizing enzyme-related genes and the leakage of enzymes (AST, ALT, LDH, and γ-GTP) as indicators of cell survival. As a result, the patterns of target mRNA expression in fresh and cryopreserved hepatocytes were very similar during the culture period between culture methods. mRNA expression levels were highly maintained at day 28 using the 3D spheroid and co-culture methods, demonstrating that these methods are useful for maintenance of liver function. Leakage of AST and ALT was higher at day 3 but decreased at day 14. LDH was not detected, suggesting that the cell viability was also maintained during the culture period. Furthermore, the functional differences between fresh and cryopreserved hepatocytes were not clearly detected. The co-culture method was useful for long-term culture not requiring 3D structure, and the 3D spheroid culture method was effective as well. With these techniques, cynomolgus monkey hepatocytes are expected to exhibit smaller individual differences and high reproducibility.
Collapse
Affiliation(s)
- Akiko Koeda
- Ina Research Inc., Ina, Japan; Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Takahiro Iwao
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan; Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Anna Nakanishi
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Shota Mizuno
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Misaki Yamashita
- Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | - Yoko Sakai
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| | | | - Tamihide Matsunaga
- Department of Clinical Pharmacy, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan; Educational Research Center for Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan.
| |
Collapse
|
27
|
Chen X, Jiang W, Ahmed A, Mahon CS, Müllner M, Cao B, Xia T. Engineering Protective Polymer Coatings for Liver Microtissues. Chem Res Toxicol 2018; 32:49-56. [PMID: 30499291 DOI: 10.1021/acs.chemrestox.8b00120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Three-dimensional (3D) hepatocyte microtissues (MT), also known as spheroids, have proven to be advantageous in providing more accurate information and physiologically relevant and predictive data for liver-related in vivo tests; therefore, spheroids have increasingly been used to study hepatotoxicity, drug delivery to the liver, and tissue engineering. However, variabilities in the generation of 3D MT remain a major challenge. Methods that encapsulate and protect hepatocytes offer a promising pathway in prolonging cell survival, as well as maintaining its liver cell functions. Herein, we studied the encapsulation and resultant protective effects of hydrogen bonded, biocompatible polymer coatings for hepatocyte MT in 3D cell culture. We exposed the MT to hepatotoxic nanomaterials (NMs), such as graphene oxide (GO) and cobalt oxide (Co3O4), to assess the protective effects of poly(vinylpyrrolidone) (PVPON) and tannic acid (TA) coatings. The polymer coating allowed the MT to maintain its morphology. More significantly, it increased the viability of hepatocyte-composed MT by hampering the cellular interaction between hostile NMs and hepatocytes. Based on alanine transaminase (ALT) and aspartate aminotransferase (AST) levels, the liver cell function was maintained throughout the coating process, including after NM treatment. The study provides a straightforward and safe methodology for maintaining the morphology as well as cellular function of hepatocyte MT in vitro.
Collapse
Affiliation(s)
- Xi Chen
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States.,Wyss Institute for Biologically Inspired Engineering , Harvard University , Boston , Massachusetts 02115 , United States
| | - Wen Jiang
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States.,Department of Chemistry , University of Georgia , Athens , Georgia 30602 , United States
| | - Ayman Ahmed
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States
| | - Clare S Mahon
- Key Centre for Polymers and Colloids, School of Chemistry , The University of Sydney , Sydney , New South Wales 2006 , Australia
| | - Markus Müllner
- Key Centre for Polymers and Colloids, School of Chemistry , The University of Sydney , Sydney , New South Wales 2006 , Australia.,The University of Sydney Nano Institute (Sydney Nano) , Sydney , New South Wales 2006 , Australia
| | - Bocheng Cao
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California 90095 , United States
| | - Tian Xia
- Center for Environmental Implications of Nanotechnology, California NanoSystems Institute , University of California , Los Angeles , California 90095 , United States.,Division of NanoMedicine, Department of Medicine , University of California , Los Angeles , California 90095 , United States
| |
Collapse
|
28
|
Pihl AF, Offersgaard AF, Mathiesen CK, Prentoe J, Fahnøe U, Krarup H, Bukh J, Gottwein JM. High density Huh7.5 cell hollow fiber bioreactor culture for high-yield production of hepatitis C virus and studies of antivirals. Sci Rep 2018; 8:17505. [PMID: 30504788 PMCID: PMC6269495 DOI: 10.1038/s41598-018-35010-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/26/2018] [Indexed: 12/15/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection poses a serious global public health burden. Despite the recent development of effective treatments there is a large unmet need for a prophylactic vaccine. Further, antiviral resistance might compromise treatment efficiency in the future. HCV cell culture systems are typically based on Huh7 and derived hepatoma cell lines cultured in monolayers. However, efficient high cell density culture systems for high-yield HCV production and studies of antivirals are lacking. We established a system based on Huh7.5 cells cultured in a hollow fiber bioreactor in the presence or absence of bovine serum. Using an adapted chimeric genotype 5a virus, we achieved peak HCV infectivity and RNA titers of 7.6 log10 FFU/mL and 10.4 log10 IU/mL, respectively. Bioreactor derived HCV showed high genetic stability, as well as buoyant density, sensitivity to neutralizing antibodies AR3A and AR4A, and dependency on HCV co-receptors CD81 and SR-BI comparable to that of HCV produced in monolayer cell cultures. Using the bioreactor platform, treatment with the NS5A inhibitor daclatasvir resulted in HCV escape mediated by the NS5A resistance substitution Y93H. In conclusion, we established an efficient high cell density HCV culture system with implications for studies of antivirals and vaccine development.
Collapse
Affiliation(s)
- Anne F Pihl
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anna F Offersgaard
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian K Mathiesen
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jannick Prentoe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Krarup
- Section of Molecular Diagnostics, Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Judith M Gottwein
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Hvidovre Hospital and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
29
|
Ruoß M, Häussling V, Schügner F, Olde Damink LHH, Lee SML, Ge L, Ehnert S, Nussler AK. A Standardized Collagen-Based Scaffold Improves Human Hepatocyte Shipment and Allows Metabolic Studies over 10 Days. Bioengineering (Basel) 2018; 5:86. [PMID: 30332824 PMCID: PMC6316810 DOI: 10.3390/bioengineering5040086] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 10/12/2018] [Accepted: 10/14/2018] [Indexed: 02/06/2023] Open
Abstract
Due to pronounced species differences, hepatotoxicity of new drugs often cannot be detected in animal studies. Alternatively, human hepatocytes could be used, but there are some limitations. The cells are not always available on demand or in sufficient amounts, so far there has been only limited success to allow the transport of freshly isolated hepatocytes without massive loss of function or their cultivation for a long time. Since it is well accepted that the cultivation of hepatocytes in 3D is related to an improved function, we here tested the Optimaix-3D Scaffold from Matricel for the transport and cultivation of hepatocytes. After characterization of the scaffold, we shipped cells on the scaffold and/or cultivated them over 10 days. With the evaluation of hepatocyte functions such as urea production, albumin synthesis, and CYP activity, we showed that the metabolic activity of the cells on the scaffold remained nearly constant over the culture time whereas a significant decrease in metabolic activity occurred in 2D cultures. In addition, we demonstrated that significantly fewer cells were lost during transport. In summary, the collagen-based scaffold allows the transport and cultivation of hepatocytes without loss of function over 10 days.
Collapse
Affiliation(s)
- Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany.
| | - Victor Häussling
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany.
| | | | | | - Serene M L Lee
- Hepacult GmbH, 82152 Martinsried/Planegg, Germany.
- Biobank of the Department of General, Visceral and Transplantation Surgery, Hospital of the LMU, 81377 Munich, Germany.
| | - Liming Ge
- Hepacult GmbH, 82152 Martinsried/Planegg, Germany.
| | - Sabrina Ehnert
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany.
| | - Andreas K Nussler
- Department of Traumatology, Siegfried Weller Institute, Eberhard Karls University, 72076 Tübingen, Germany.
| |
Collapse
|
30
|
Han W, Wu Q, Zhang X, Duan Z. Innovation for hepatotoxicity in vitro research models: A review. J Appl Toxicol 2018; 39:146-162. [PMID: 30182494 DOI: 10.1002/jat.3711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 12/18/2022]
Abstract
Many categories of drugs can induce hepatotoxicity, so improving the prediction of toxic drugs is important. In vitro models using human hepatocytes are more accurate than in vivo animal models. Good in vitro models require an abundance of metabolic enzyme activities and normal cellular polarity. However, none of the in vitro models can completely simulate hepatocytes in the human body. There are two ways to overcome this limitation: enhancing the metabolic function of hepatocytes and changing the cultural environment. In this review, we summarize the current state of research, including the main characteristics of in vitro models and their limitations, as well as improved technology and developmental prospects. We hope that this review provides some new ideas for hepatotoxicity research.
Collapse
Affiliation(s)
- Weijia Han
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Qiao Wu
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Xiaohui Zhang
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| | - Zhongping Duan
- Artificial Liver Center, Beijing Youan Hospital; Capital Medical University; Beijing China
- Beijing Key Laboratory of Liver Failure; Artificial Liver Treatment and Research; Beijing China
| |
Collapse
|
31
|
Mattei G, Magliaro C, Pirone A, Ahluwalia A. Bioinspired liver scaffold design criteria. Organogenesis 2018; 14:129-146. [PMID: 30156955 PMCID: PMC6300109 DOI: 10.1080/15476278.2018.1505137] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/20/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022] Open
Abstract
Maintaining hepatic functional characteristics in-vitro is considered one of the main challenges in engineering liver tissue. As hepatocytes cultured ex-vivo are deprived of their native extracellular matrix (ECM) milieu, developing scaffolds that mimic the biomechanical and physicochemical properties of the native ECM is thought to be a promising approach for successful tissue engineering and regenerative medicine applications. On the basis that the decellularized liver matrix represents the ideal design template for engineering bioinspired hepatic scaffolds, to derive quantitative descriptors of liver ECM architecture, we characterised decellularised liver matrices in terms of their biochemical, viscoelastic and structural features along with porosity, permeability and wettability. Together, these data provide a unique set of quantitative design criteria which can be used to generate guidelines for fabricating biomaterial scaffolds for liver tissue engineering. As proof-of-concept, we investigated hepatic cell response to substrate viscoelasticity. On collagen hydrogels mimicking decellularised liver mechanics, cells showed superior morphology, higher viability and albumin secretion than on stiffer and less viscous substrates. Although scaffold properties are generally inspired by those of native tissues, our results indicate significant differences between the mechano-structural characteristics of untreated and decellularised hepatic tissue. Therefore, we suggest that design rules - such as mechanical properties and swelling behaviour - for engineering biomimetic scaffolds be re-examined through further studies on substrates matching the features of decellularized liver matrices.
Collapse
Affiliation(s)
- Giorgio Mattei
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Optics11 B.V, Amsterdam, The Netherlands
- Biophotonics & Medical Imaging and Laser LaB, VU University Amsterdam, Amsterdam, The Netherlands
| | - Chiara Magliaro
- Research Centre “E. Piaggio”, University of Pisa, Pisa, Italy
| | - Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | - Arti Ahluwalia
- Department of Information Engineering, University of Pisa, Pisa, Italy
- Research Centre “E. Piaggio”, University of Pisa, Pisa, Italy
| |
Collapse
|
32
|
Fröhlich E. Comparison of conventional and advanced in vitro models in the toxicity testing of nanoparticles. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2018; 46:1091-1107. [PMID: 29956556 PMCID: PMC6214528 DOI: 10.1080/21691401.2018.1479709] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/12/2018] [Accepted: 05/15/2018] [Indexed: 01/02/2023]
Abstract
Humans are exposed to a wide variety of nanoparticles (NPs) present in the environment, in consumer, health and medical products, and in food. Conventional cytotoxicity testing compared to animal testing is less expensive, faster and avoids ethical problems at the expense of a lower predictive value. New cellular models and exposure conditions have been developed to overcome the limitations of conventional cell culture and obtain more predictive data. The use of three-dimensional culture, co-culture and inclusion of mechanical stimulation can provide physiologically more relevant culture conditions. These systems are particularly relevant for oral, respiratory and intravenous exposure to NPs and it may be assumed that physiologically relevant application of the NPs can improve the predictive value of in vitro testing. Various groups have used advanced culture and exposure systems, but few direct comparisons between data from conventional cultures and from advanced systems exist. In silico models may present another option to predict human health risk by NPs without using animal studies. In the absence of validation, the question whether these alternative models provide more predictive data than conventional testing remains elusive.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- Center for Medical Research, Medical University of Graz, Graz, Austria
| |
Collapse
|
33
|
de Boussac H, Gondeau C, Briolotti P, Duret C, Treindl F, Römer M, Fabre JM, Herrero A, Ramos J, Maurel P, Templin M, Gerbal-Chaloin S, Daujat-Chavanieu M. Epidermal Growth Factor Represses Constitutive Androstane Receptor Expression in Primary Human Hepatocytes and Favors Regulation by Pregnane X Receptor. Drug Metab Dispos 2018; 46:223-236. [PMID: 29269410 DOI: 10.1124/dmd.117.078683] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
Growth factors have key roles in liver physiology and pathology, particularly by promoting cell proliferation and growth. Recently, it has been shown that in mouse hepatocytes, epidermal growth factor receptor (EGFR) plays a crucial role in the activation of the xenosensor constitutive androstane receptor (CAR) by the antiepileptic drug phenobarbital. Due to the species selectivity of CAR signaling, here we investigated epidermal growth factor (EGF) role in CAR signaling in primary human hepatocytes. Primary human hepatocytes were incubated with CITCO, a human CAR agonist, or with phenobarbital, an indirect CAR activator, in the presence or absence of EGF. CAR-dependent gene expression modulation and PXR involvement in these responses were assessed upon siRNA-based silencing of the genes that encode CAR and PXR. EGF significantly reduced CAR expression and prevented gene induction by CITCO and, to a lower extent, by phenobarbital. In the absence of EGF, phenobarbital and CITCO modulated the expression of 144 and 111 genes, respectively, in primary human hepatocytes. Among these genes, only 15 were regulated by CITCO and one by phenobarbital in a CAR-dependent manner. Conversely, in the presence of EGF, CITCO and phenobarbital modulated gene expression only in a CAR-independent and PXR-dependent manner. Overall, our findings suggest that in primary human hepatocytes, EGF suppresses specifically CAR signaling mainly through transcriptional regulation and drives the xenobiotic response toward a pregnane X receptor (PXR)-mediated mechanism.
Collapse
Affiliation(s)
- Hugues de Boussac
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Claire Gondeau
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Philippe Briolotti
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Cédric Duret
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Fridolin Treindl
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Michael Römer
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Jean-Michel Fabre
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Astrid Herrero
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Jeanne Ramos
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Patrick Maurel
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Markus Templin
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Sabine Gerbal-Chaloin
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| | - Martine Daujat-Chavanieu
- IRMB, INSERM, University Montpellier, Montpellier, France (H.d.B., C.G., P.B., C.D., P.M., S.G.-C., M.D.-C.); CHU Montpellier, IRMB, Montpellier, France (C.G., C.D., M.D.-C.); Natural and Medical Sciences Institute, University of Tübingen, Reutlingen, Germany (F.T., M.T.); Centre of Bioinformatics Tübingen (ZBIT), University of Tübingen, Tübingen, Germany (M.R.); Department of Digestive Surgery, Hospital Saint Eloi, CHU Montpellier, Montpellier, France (J.-M.F.); Departments of General Surgery, Division of Transplantation, College of Medicine, University of Montpellier, Montpellier, France (A.H.); and Pathological Anatomy Department, Hospital Guy de Chauliac, CHU Montpellier, Montpellier, France (J.R.)
| |
Collapse
|
34
|
Liaw CY, Ji S, Guvendiren M. Engineering 3D Hydrogels for Personalized In Vitro Human Tissue Models. Adv Healthc Mater 2018; 7. [PMID: 29345429 DOI: 10.1002/adhm.201701165] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/13/2017] [Indexed: 01/17/2023]
Abstract
There is a growing interest in engineering hydrogels for 3D tissue and disease models. The major motivation is to better mimic the physiological microenvironment of the disease and human condition. 3D tissue models derived from patients' own cells can potentially revolutionize the way treatment and diagnostic alternatives are developed. This requires development of tissue mimetic hydrogels with user defined and tunable properties. In this review article, a recent summary of 3D hydrogel platforms for in vitro tissue and disease modeling is given. Hydrogel design considerations and available hydrogel systems are summarized, followed by the types of currently available hydrogel models, such as bulk hydrogels, porous scaffolds, fibrous scaffolds, hydrogel microspheres, hydrogel sandwich systems, microwells, and 3D bioprinted constructs. Although hydrogels are utilized for a wide range of tissue models, this article focuses on liver and cancer models. This article also provides a detailed section on current challenges and future perspectives of hydrogel-based tissue models.
Collapse
Affiliation(s)
- Chya-Yan Liaw
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| | - Shen Ji
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| |
Collapse
|
35
|
Elasticity-based development of functionally enhanced multicellular 3D liver encapsulated in hybrid hydrogel. Acta Biomater 2017; 64:67-79. [PMID: 28966094 DOI: 10.1016/j.actbio.2017.09.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 08/30/2017] [Accepted: 09/27/2017] [Indexed: 12/23/2022]
Abstract
Current in vitro liver models provide three-dimensional (3-D) microenvironments in combination with tissue engineering technology and can perform more accurate in vivo mimicry than two-dimensional models. However, a human cell-based, functionally mature liver model is still desired, which would provide an alternative to animal experiments and resolve low-prediction issues on species differences. Here, we prepared hybrid hydrogels of varying elasticity and compared them with a normal liver, to develop a more mature liver model that preserves liver properties in vitro. We encapsulated HepaRG cells, either alone or with supporting cells, in a biodegradable hybrid hydrogel. The elastic modulus of the 3D liver dynamically changed during culture due to the combined effects of prolonged degradation of hydrogel and extracellular matrix formation provided by the supporting cells. As a result, when the elastic modulus of the 3D liver model converges close to that of the in vivo liver (≅ 2.3 to 5.9 kPa), both phenotypic and functional maturation of the 3D liver were realized, while hepatic gene expression, albumin secretion, cytochrome p450-3A4 activity, and drug metabolism were enhanced. Finally, the 3D liver model was expanded to applications with embryonic stem cell-derived hepatocytes and primary human hepatocytes, and it supported prolonged hepatocyte survival and functionality in long-term culture. Our model represents critical progress in developing a biomimetic liver system to simulate liver tissue remodeling, and provides a versatile platform in drug development and disease modeling, ranging from physiology to pathology. STATEMENT OF SIGNIFICANCE We provide a functionally improved 3D liver model that recapitulates in vivo liver stiffness. We have experimentally addressed the issues of orchestrated effects of mechanical compliance, controlled matrix formation by stromal cells in conjunction with hepatic differentiation, and functional maturation of hepatocytes in a dynamic 3D microenvironment. Our model represents critical progress in developing a biomimetic liver system to simulate liver tissue remodeling, and provides a versatile platform in drug development and disease modeling, ranging from physiology to pathology. Additionally, recent advances in the stem-cell technologies have made the development of 3D organoid possible, and thus, our study also provides further contribution to the development of physiologically relevant stem-cell-based 3D tissues that provide an elasticity-based predefined biomimetic 3D microenvironment.
Collapse
|
36
|
Agarwal T, Narayan R, Maji S, Ghosh SK, Maiti TK. Decellularized caprine liver extracellular matrix as a 2D substrate coating and 3D hydrogel platform for vascularized liver tissue engineering. J Tissue Eng Regen Med 2017; 12:e1678-e1690. [PMID: 29052367 DOI: 10.1002/term.2594] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 09/20/2017] [Accepted: 10/09/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Tarun Agarwal
- Department of BiotechnologyIndian Institute of Technology Kharagpur Kharagpur West Bengal India
| | - Rajan Narayan
- Department of BiotechnologyIndian Institute of Technology Kharagpur Kharagpur West Bengal India
| | - Somnath Maji
- Department of BiotechnologyIndian Institute of Technology Kharagpur Kharagpur West Bengal India
| | - Sudip Kumar Ghosh
- Department of BiotechnologyIndian Institute of Technology Kharagpur Kharagpur West Bengal India
| | - Tapas Kumar Maiti
- Department of BiotechnologyIndian Institute of Technology Kharagpur Kharagpur West Bengal India
| |
Collapse
|
37
|
Azandeh S, Mohammad Gharravi A, Orazizadeh M, Khodadi A, Hashemi Tabar M. Improvement of mesenchymal stem cell differentiation into the endoderm lineage by four step sequential method in biocompatible biomaterial. ACTA ACUST UNITED AC 2016; 6:9-13. [PMID: 27340619 PMCID: PMC4916552 DOI: 10.15171/bi.2016.02] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 02/05/2016] [Accepted: 02/22/2016] [Indexed: 12/22/2022]
Abstract
![]()
Introduction: The goal of the study described here, was to investigate the potential of umbilical cord derived mesenchymal stem cell (UC-MSCs) into hepatocyte like cells in a sequential 2D and 3D differentiation protocols as alternative therapy.
Methods: Mesenchymal stem cells (MSCs) were isolated from the umbilical cord (UC) and CD markers were analyzed by flow cytometry. For hepatic differentiation of UC-MSCs, cells were induced with a sequential 4-step protocol in 3D and 2D culture system. Urea concentration and albumin secretion into the culture medium was quantified by ELISA. Gene expression levels of AFP, ALB, and CK18 were determined by RT-PCR. Data were statistically analyzed by the SPSS software. The difference between the mean was considered significant when p < 0.05.
Results: Growth factor dependent morphological changes from elongated fibroblast-like cells to round epithelial cell morphology were observed in 2D culture. Cell proliferation analysis showed round-shaped morphology with clear cytoplasm and nucleus on the alginate scaffold in 3D culture. The mean valuses of albumin production and urea secretion were significantly higher in the 3D Culture system when compared with the 2D culture (p = 0.005 vs p = 0.001), respectively. Treatment of cells with TSA in the final step of differentiation induced an increased expression of CK18 and a decreased expression of αFP in both the 3D and 2D cultures (p = 0.026), but led to a decreased albumin gene expression, and an increased expression in the 2D culture (p = 0.001).
Conclusion: Findings of the present study indicated that sequential exposure of UC-MSCs with growth factors in 3D culture improves hepatic differentiation.
Collapse
Affiliation(s)
- Saeed Azandeh
- Cellular and Molecular Research Center (CMRC), Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | | | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center (CMRC), Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Ali Khodadi
- Cancer, Petroleum and Environmental Pollutants Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmoud Hashemi Tabar
- Cellular and Molecular Research Center (CMRC), Department of Anatomical Science, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| |
Collapse
|
38
|
Dixit V, Moore A, Tsao H, Hariparsad N. Application of Micropatterned Cocultured Hepatocytes to Evaluate the Inductive Potential and Degradation Rate of Major Xenobiotic Metabolizing Enzymes. Drug Metab Dispos 2016; 44:250-61. [PMID: 26658225 DOI: 10.1124/dmd.115.067173] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 12/08/2015] [Indexed: 12/19/2022] Open
Abstract
Long-term coculture models of hepatocytes are promising tools to study drug transport, clearance, and hepatoxicity. In this report we compare the basal expression of drug disposition genes and the inductive response of prototypical inducers (rifampin, phenobarbital, phenytoin) in hepatocyte two-dimensional monocultures and the long-term coculture model (HepatoPac). All the inducers used in the study increased the expression and activity of CYP3A4, CYP2B6 and CYP2C enzymes in the HepatoPac cultures. The coculture model showed a consistent and higher induction of CYP2C enzymes compared with the monocultures. The EC50 of rifampin for CYP3A4 and CYP2C9 was up to 10-fold lower in HepatoPac than the monocultures. The EC50 of rifampin calculated from the clinical drug interaction studies correlated well with the EC50 observed in the HepatoPac cultures. Owing to the long-term stability of the HepatoPac cultures, we were able to directly measure a half-life (t1/2) for both CYP3A4 and CYP2B6 using the depletion kinetics of mRNA and functional activity. The t1/2 for CYP3A4 mRNA was 26 hours and that for the functional protein was 49 hours. The t1/2 of CYP2B6 was 38 hours (mRNA) and 68 hours (activity), which is longer than CYP3A4 and shows the differential turnover of these two proteins. This is the first study to our knowledge to report the turnover rate of CYP2B6 in human hepatocytes. The data presented here demonstrate that the HepatoPac cultures have the potential to be used in long-term culture to mimic complex clinical scenarios.
Collapse
Affiliation(s)
- Vaishali Dixit
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Amanda Moore
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Hong Tsao
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| |
Collapse
|