1
|
Heydari Z, Najimi M, Mirzaei H, Shpichka A, Ruoss M, Farzaneh Z, Montazeri L, Piryaei A, Timashev P, Gramignoli R, Nussler A, Baharvand H, Vosough M. Tissue Engineering in Liver Regenerative Medicine: Insights into Novel Translational Technologies. Cells 2020; 9:E304. [PMID: 32012725 PMCID: PMC7072533 DOI: 10.3390/cells9020304] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Organ and tissue shortage are known as a crucially important public health problem as unfortunately a small percentage of patients receive transplants. In the context of emerging regenerative medicine, researchers are trying to regenerate and replace different organs and tissues such as the liver, heart, skin, and kidney. Liver tissue engineering (TE) enables us to reproduce and restore liver functions, fully or partially, which could be used in the treatment of acute or chronic liver disorders and/or generate an appropriate functional organ which can be transplanted or employed as an extracorporeal device. In this regard, a variety of techniques (e.g., fabrication technologies, cell-based technologies, microfluidic systems and, extracorporeal liver devices) could be applied in tissue engineering in liver regenerative medicine. Common TE techniques are based on allocating stem cell-derived hepatocyte-like cells or primary hepatocytes within a three-dimensional structure which leads to the improvement of their survival rate and functional phenotype. Taken together, new findings indicated that developing liver tissue engineering-based techniques could pave the way for better treatment of liver-related disorders. Herein, we summarized novel technologies used in liver regenerative medicine and their future applications in clinical settings.
Collapse
Affiliation(s)
- Zahra Heydari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran 1665659911, Iran
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental & Clinical Research, Université Catholique de Louvain, B-1200 Brussels, Belgium;
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan 121135879, Iran;
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, 119146 Moscow, Russia; (A.S.); (P.T.)
| | - Marc Ruoss
- Siegfried Weller Institute for Trauma Research, University of Tübingen, 72076 Tübingen, Germany; (M.R.); (A.N.)
| | - Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Abbas Piryaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119146 Moscow, Russia; (A.S.); (P.T.)
- Department of Polymers and Composites, N.N.Semenov Institute of Chemical Physics, 117977 Moscow, Russia
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Andreas Nussler
- Siegfried Weller Institute for Trauma Research, University of Tübingen, 72076 Tübingen, Germany; (M.R.); (A.N.)
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran 1665659911, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| |
Collapse
|
2
|
Hsieh CJ, Sun M, Osborne G, Ricker K, Tsai FC, Li K, Tomar R, Phuong J, Schmitz R, Sandy MS. Cancer Hazard Identification Integrating Human Variability: The Case of Coumarin. Int J Toxicol 2019; 38:501-552. [PMID: 31845612 DOI: 10.1177/1091581819884544] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Coumarin is a naturally occurring sweet-smelling benzopyrone that may be extracted from plants or synthesized for commercial uses. Its uses include as a flavoring agent, fragrance enhancer, and odor-masking additive. We reviewed and evaluated the scientific evidence on the carcinogenicity of coumarin, integrating information from carcinogenicity studies in animals with mechanistic and other relevant data, including data from toxicogenomic, genotoxicity, and metabolism studies, and studies of human variability of a key enzyme, CYP2A6. Increases in tumors were observed in multiple studies in rats and mice in multiple tissues. Our functional pathway analysis identified several common cancer-related biological processes/pathways affected by coumarin in rat liver following in vivo exposure and in human primary hepatocytes exposed in vitro. When coumarin 7-hydroxylation by CYP2A6 is compromised, this can lead to a shift in metabolism to the 3,4-epoxidation pathway and increased generation of electrophilic metabolites. Mechanistic data align with 3 key characteristics of carcinogens, namely formation of electrophilic metabolites, genotoxicity, and induction of oxidative stress. Considerations of metabolism, human variability in CYP2A6 activity, and coumarin hepatotoxicity in susceptible individuals provide additional support for carcinogenicity concern. Our analysis illustrates the importance of integrating information on human variability in the cancer hazard identification process.
Collapse
Affiliation(s)
- ChingYi Jennifer Hsieh
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Meng Sun
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Gwendolyn Osborne
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Karin Ricker
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Feng C Tsai
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Kate Li
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Rajpal Tomar
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA.,Retired
| | - Jimmy Phuong
- Department of Biomedical and Health Informatics, University of Washington, Seattle, WA, USA
| | - Rose Schmitz
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| | - Martha S Sandy
- Office of Environmental Health Hazard Assessment, CalEPA, Sacramento and Oakland, CA, USA
| |
Collapse
|
3
|
Dott W, Wright J, Cain K, Mistry P, Herbert KE. Integrated metabolic models for xenobiotic induced mitochondrial toxicity in skeletal muscle. Redox Biol 2018; 14:198-210. [PMID: 28942197 PMCID: PMC5610037 DOI: 10.1016/j.redox.2017.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 11/16/2022] Open
Abstract
There is a need for robust in vitro models to sensitively capture skeletal muscle adverse toxicities early in the research and development of novel xenobiotics. To this end, an in vitro rat skeletal muscle model (L6) was used to study the translation of transcriptomics data generated from an in vivo rat model. Novel sulfonyl isoxazoline herbicides were associated with skeletal muscle toxicity in an in vivo rat model. Gene expression pathway analysis on skeletal muscle tissues taken from in vivo repeat dose studies identified enriched pathways associated with mitochondrial dysfunction, oxidative stress, energy metabolism, protein regulation and cell cycle. Mitochondrial dysfunction and oxidative stress were further explored using in vitro L6 metabolic models. These models demonstrated that the sulfonyl isoxazoline compounds induced mitochondrial dysfunction, mitochondrial superoxide production and apoptosis. These in vitro findings accurately concurred with the in vivo transcriptomics data, thereby confirming the ability of the L6 skeletal muscle models to identify relevant in vivo mechanisms of xenobiotic-induced toxicity. Moreover, these results highlight the sensitivity of the L6 galactose media model to study mitochondrial perturbation associated with skeletal muscle toxicity; this model may be utilised to rank the potency of novel xenobiotics upon further validation.
Collapse
Affiliation(s)
- William Dott
- Department of Cardiovascular Sciences, University of Leicester, UK
| | | | - Kelvin Cain
- MRC Toxicology Unit, University of Leicester, Leicester, UK
| | | | - Karl E Herbert
- Department of Cardiovascular Sciences, University of Leicester, UK.
| |
Collapse
|
4
|
Agrawal G, Aung A, Varghese S. Skeletal muscle-on-a-chip: an in vitro model to evaluate tissue formation and injury. LAB ON A CHIP 2017; 17:3447-3461. [PMID: 28871305 PMCID: PMC6296378 DOI: 10.1039/c7lc00512a] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Engineered skeletal muscle tissues can be used for in vitro studies that require physiologically relevant models of native tissues. Herein, we describe the development of a three-dimensional (3D) skeletal muscle tissue that recapitulates the architectural and structural complexities of muscle within a microfluidic device. Using a 3D photo-patterning approach, we spatially confined a cell-laden gelatin network around two bio-inert hydrogel pillars, which induce uniaxial alignment of the cells and serve as anchoring sites for the encapsulated cells and muscle tissues as they form and mature. We have characterized the tissue morphology and strain profile during differentiation of the cells and skeletal muscle tissue formation by using a combination of fluorescence microscopy and computational tools. The time-dependent strain profile suggests the existence of individual cells within the gelatin matrix, which differentiated to form a multinucleated skeletal muscle tissue bundle as a function of culture time. We have also developed a method to calculate the passive tension generated by the engineered muscle tissue bundles suspended between two pillars. Finally, as a proof-of-concept we have examined the applicability of the skeletal muscle-on-chip system as a screening platform and in vitro muscle injury model. We studied the dose-dependent effect of cardiotoxin on the engineered muscle tissue architecture and its subsequent effect on the passive tension. This simple yet effective tool can be appealing for studies that necessitate the analysis of skeletal muscle structure and function, including preclinical drug discovery and development.
Collapse
Affiliation(s)
- Gaurav Agrawal
- Department of Bioengineering, University of California-San Diego, La Jolla, CA, USA.
| | | | | |
Collapse
|
5
|
Three-dimensional in vitro gut model on a villi-shaped collagen scaffold. BIOCHIP JOURNAL 2017. [DOI: 10.1007/s13206-017-1307-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
6
|
Orbach SM, Less RR, Kothari A, Rajagopalan P. In Vitro Intestinal and Liver Models for Toxicity Testing. ACS Biomater Sci Eng 2017; 3:1898-1910. [PMID: 33440548 DOI: 10.1021/acsbiomaterials.6b00699] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The human body is exposed to hundreds of chemicals every day. Many of these toxicants have unknown effects on the body that can be deleterious. Furthermore, chemicals can have a synergistic effect, resulting in toxic responses of cocktails at relatively low individual exposure levels. The gastrointestinal (GI) tract and the liver are the first organs to be exposed to ingested pharmaceuticals and environmental chemicals. As a result, these organs often experience extensive damage from xenobiotics and their metabolites. In vitro models offer a promising method for testing toxic effects. Many advanced in vitro models have been developed for GI and liver toxicity. These models strive to recapitulate the in vivo organ architecture to more accurately model chemical toxicity. In this review, we discuss many of these advances, in addition to recent efforts to integrate the GI and the liver in vitro for a more holistic toxicity model.
Collapse
Affiliation(s)
- Sophia M Orbach
- Department of Chemical Engineering, ‡School of Biomedical Engineering and Sciences, and §ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Rebekah R Less
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, and §ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Anjaney Kothari
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, and ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, School of Biomedical Engineering and Sciences, and ICTAS Center for Systems Biology of Engineered Tissue, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
7
|
Tanaka Y, Fujii W, Hori H, Kitagawa Y, Ozaki K. Relationship between coumarin-induced hepatocellular toxicity and mitochondrial function in rats. Food Chem Toxicol 2016; 90:1-9. [PMID: 26806632 DOI: 10.1016/j.fct.2016.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 01/05/2016] [Accepted: 01/13/2016] [Indexed: 12/28/2022]
Abstract
The manifestation of coumarin-induced hepatocellular toxicity may differ and depends on the frequency of administration to rats. A single coumarin dose induces hepatocellular necrosis while repeated doses induce only hepatocyte degeneration. However, the mechanism underlying these effects remains unclear. Therefore, we investigated the mechanism of coumarin-induced hepatotoxicity in rats. Coumarin was administered to male rats as a single dose or for 4 consecutive days, and samples were obtained 4 or 24 h after a single dose or 24 h after the repeated doses. A single coumarin dose significantly induced hepatocellular necrosis in rats; however, toxicity was attenuated after repeated dosing. With a single dose, hepatocellular necrosis was preceded by increased mitochondrial number and size and decreased mitochondrial function. An increased expression of granular cytochrome P450 (CYP) 2E1 protein was observed in the cytoplasm and mitochondria of coumarin-treated rats compared to the expression in the untreated controls. Nevertheless, repeated dosing showed mitochondrial function that was equivalent to that of the control while enlarged CYP2E1 protein droplets were distributed outside the mitochondria. These results suggest that mitochondrial function and CYP2E1 expression might be involved in coumarin-induced hepatocellular toxicity in rats. A reduction in mitochondrial CYP2E1 might be implicated in the acquisition of coumarin resistance after repeated doses.
Collapse
Affiliation(s)
- Yasuhiro Tanaka
- Suntory Business Expert Limited, 8-1-1 Seikadai Seika-cho, Soraku-gun, Kyoto 619-0238, Japan; Laboratory of Pathology, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge, Hirakata, Osaka 573-0101, Japan
| | - Wataru Fujii
- Suntory Business Expert Limited, 8-1-1 Seikadai Seika-cho, Soraku-gun, Kyoto 619-0238, Japan
| | - Hisako Hori
- Suntory Business Expert Limited, 8-1-1 Seikadai Seika-cho, Soraku-gun, Kyoto 619-0238, Japan
| | - Yoshinori Kitagawa
- Suntory Business Expert Limited, 8-1-1 Seikadai Seika-cho, Soraku-gun, Kyoto 619-0238, Japan
| | - Kiyokazu Ozaki
- Laboratory of Pathology, Faculty of Pharmaceutical Sciences, Setsunan University, 45-1 Nagaotoge, Hirakata, Osaka 573-0101, Japan.
| |
Collapse
|
8
|
Teng S, Barcellini-Couget S, Beaudouin R, Brochot C, Desousa G, Rahmani R, Pery A. BK/TD models for analyzing in vitro impedance data on cytotoxicity. Toxicol Lett 2015; 235:96-106. [DOI: 10.1016/j.toxlet.2015.03.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/24/2015] [Accepted: 03/27/2015] [Indexed: 02/01/2023]
|
9
|
Kim SH, Chi M, Yi B, Kim SH, Oh S, Kim Y, Park S, Sung JH. Three-dimensional intestinal villi epithelium enhances protection of human intestinal cells from bacterial infection by inducing mucin expression. Integr Biol (Camb) 2014; 6:1122-31. [PMID: 25200891 DOI: 10.1039/c4ib00157e] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Current in vitro cell culture models do not reflect human physiology, and various efforts have been made to enhance existing models. Reconstitution of three-dimensional (3D) tissue structure has been one of the strategies, since 3D tissue structure provides essential cellular environmental cues for cell functions. Previously, we developed a novel hydrogel microfabrication technique for constructing an accurate 3D replica of human intestinal villi epithelium. In this study, genetic and physiological properties of the 3D villi model were examined to gain a better insight into the barrier function of gut epithelium and its interaction with microbes. Gene expression study of Caco-2 on the 3D villi scaffold revealed that expression of MUC17, which is one of the transmembrane mucins, was highly enhanced in the 3D villi model, compared to a monolayer culture. Cells on the scaffold were almost immune to bacterial infection, while MUC17 knockdown in Caco-2 cells restored bacterial infectivity. The 3D villi model also exhibited changes in the barrier function compared to the 2D model, manifested by changes in transepithelial electrical resistance (TEER) and permeability of FITC-dextran. Knockdown of MUC17 resulted in reduction of tight junction protein expression and further increase in permeability, suggesting an important role of MUC17 in the barrier function against pathogens and xenobiotics. Our study suggests that mimicking the 3D tissue architecture of the small intestine induces physiological changes in human intestinal cells.
Collapse
Affiliation(s)
- Si Hyun Kim
- Department of Chemical Engineering, Hongik University, Seoul, Korea.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
De Bruyn T, Chatterjee S, Fattah S, Keemink J, Nicolaï J, Augustijns P, Annaert P. Sandwich-cultured hepatocytes: utility for in vitro exploration of hepatobiliary drug disposition and drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2013; 9:589-616. [PMID: 23452081 DOI: 10.1517/17425255.2013.773973] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The sandwich-cultured hepatocyte (SCH) model has become an invaluable in vitro tool for studying hepatic drug transport, metabolism, biliary excretion and toxicity. The relevant expression of many hepatocyte-specific functions together with the in vivo-like morphology favor SCHs over other preclinical models for evaluating hepatobiliary drug disposition and drug-induced hepatotoxicity. AREAS COVERED In this review, the authors highlight recommended procedures required for reproducibly culturing hepatocytes in sandwich configuration. It also provides an overview of the SCH model characteristics as a function of culture time. Lastly, the article presents a summary of the most prominent applications of the SCH model, including hepatic drug clearance prediction, drug-drug interaction potential and drug-induced hepatotoxicity. EXPERT OPINION When human (cryopreserved) hepatocytes are used to establish sandwich cultures, the model appears particularly valuable to quantitatively investigate clinically relevant mechanisms related to in vivo hepatobiliary drug disposition and hepatotoxicity. Nonetheless, the SCH model would largely benefit from better insight into the fundamental cell signaling mechanisms that are critical for long-term in vitro maintenance of the hepatocytic phenotype. Studies systematically exploring improved cell culture conditions (e.g., co-cultures or extracellular matrix modifications), as well as in vitro work identifying key transcription factors involved in hepatocyte differentiation are currently emerging.
Collapse
Affiliation(s)
- Tom De Bruyn
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, O&N2, Herestraat 49-bus-921, 3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
11
|
Queiroz ML, Torello CO, Constantino AT, Ramos AL, de Souza Queiroz J. Angelica sinensisModulates Immunohematopoietic Response and Increases Survival of Mice Infected withListeria monocytogenes. J Med Food 2010; 13:1451-9. [DOI: 10.1089/jmf.2009.0108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mary L.S. Queiroz
- Department of Pharmacology/Hemocenter, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Cristiane Okuda Torello
- Department of Pharmacology/Hemocenter, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Anderson T. Constantino
- Department of Pharmacology/Hemocenter, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Aline Lisie Ramos
- Department of Pharmacology/Hemocenter, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| | - Julia de Souza Queiroz
- Department of Pharmacology/Hemocenter, Faculty of Medical Sciences, State University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
12
|
Swift B, Pfeifer ND, Brouwer KLR. Sandwich-cultured hepatocytes: an in vitro model to evaluate hepatobiliary transporter-based drug interactions and hepatotoxicity. Drug Metab Rev 2010; 42:446-71. [PMID: 20109035 PMCID: PMC3097390 DOI: 10.3109/03602530903491881] [Citation(s) in RCA: 290] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sandwich-cultured hepatocytes (SCH) are a powerful in vitro tool that can be utilized to study hepatobiliary drug transport, species differences in drug transport, transport protein regulation, drug-drug interactions, and hepatotoxicity. This review provides an up-to-date summary of the SCH model, including a brief history of, and introduction to, the use of SCH, as well as methodology to evaluate hepatobiliary drug disposition. A summary of the literature that has utilized this model to examine the interplay between drug-metabolizing enzymes and transport proteins, drug-drug interactions at the transport level, and hepatotoxicity as a result of altered hepatic transport also is provided.
Collapse
Affiliation(s)
- Brandon Swift
- University of North Carolina at Chapel Hill, 27599-7569, USA
| | | | | |
Collapse
|
13
|
Abstract
In the last 15 years many different liver cell culture devices, consisting of functional liver cells and artificial materials, have been developed. They have been devised for numerous different applications, such as temporary organ replacement (a bridge to liver transplantation or native liver regeneration) and as in vitro screening systems in the early stages of the drug development process, like assessing hepatotoxicity, hepatic drug metabolism, and induction/inhibition studies. Relevant literature is summarized about artificial human liver cell culture systems by scrutinizing PubMed from 2003 to 2009. Existing devices are divided in 2D configurations (e.g., static monolayer, sandwich, perfused cells, and flat plate) and 3D configurations (e.g., liver slices, spheroids, and different types of bioreactors). The essential features of an ideal liver cell culture system are discussed: different types of scaffolds, oxygenation systems, extracellular matrixes (natural and artificial), cocultures with nonparenchymal cells, and the role of shear stress problems. Finally, miniaturization and high-throughput systems are discussed. All these factors contribute in their own way to the viability and functionality of liver cells in culture. Depending on the aim for which they are designed, several good systems are available for predicting hepatotoxicity and hepatic metabolism within the general population. To predict hepatotoxicity in individual cases genomic analysis might be essential as well.
Collapse
Affiliation(s)
- B Andria
- Center of Biotechnologies, Cardarelli Hospital, Naples, Italy; †Faculty of Pharmacy, 'Federico II" University, Naples, Italy
| | - A Bracco
- Center of Biotechnologies, Cardarelli Hospital , Naples , Italy
| | - G Cirino
- † Faculty of Pharmacy, 'Federico II" University , Naples , Italy
| | - R A F M Chamuleau
- ‡ Academic Medical Center, Tytgat Institute for Liver and Intestinal Research, University of Amsterdam , Amsterdam , The Netherlands
| |
Collapse
|
14
|
Elliott NT, Yuan F. A review of three-dimensional in vitro tissue models for drug discovery and transport studies. J Pharm Sci 2010; 100:59-74. [PMID: 20533556 DOI: 10.1002/jps.22257] [Citation(s) in RCA: 326] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 05/04/2010] [Indexed: 12/12/2022]
Abstract
The use of animal models in drug discovery studies presents issues with feasibility and ethical concerns. To address these limitations, in vitro tissue models have been developed to provide a means for systematic, repetitive, and quantitative investigation of drugs. By eliminating or reducing the need for animal subjects, these models can serve as platforms for more tightly controlled, high-throughput screening of drugs and for pharmacokinetic and pharmacodynamic analyses of drugs. The focus of this review is three-dimensional (3D) tissue models that can capture cell-cell and cell-matrix interactions. Compared to the 2D culture of cell monolayers, 3D models more closely mimic native tissues since the cellular microenvironment established in the 3D models often plays a significant role in disease progression and cellular responses to drugs. A growing body of research has been published in the literature, which highlights the benefits of the 3D in vitro models of various tissues. This review provides an overview of some successful 3D in vitro models that have been developed to mimic liver, breast, cardiac, muscle, bone, and corneal tissues as well as malignant tissues in solid tumors.
Collapse
Affiliation(s)
- Nelita T Elliott
- Department of Biomedical Engineering, Duke University, 136 Hudson Hall, PO Box 90281, Durham, North Carolina 27708, USA
| | | |
Collapse
|
15
|
Gómez-Lechón MJ, Castell JV, Donato MT. The use of hepatocytes to investigate drug toxicity. Methods Mol Biol 2010; 640:389-415. [PMID: 20645064 DOI: 10.1007/978-1-60761-688-7_21] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The liver is very active in metabolizing foreign compounds and the major target for toxicity caused by drugs. Hepatotoxicity may be the result of the drug itself or, more frequently, a result of the bioactivation process and the production of reactive metabolites. Prioritization of compounds based on human hepatotoxicity potential is currently a key unmet need in drug discovery, as it can become a major problem for several lead compounds in later stages of the drug discovery pipeline. Therefore, evaluation of potential hepatotoxicity represents a critical step in the development of new drugs. Cultured hepatocytes are increasingly used by the pharmaceutical industry for the screening of hepatotoxic potential of new molecules. Hepatocytes in culture retain hepatic key functions and constitute a valuable tool to identify chemically induced cellular damage. Their use has notably contributed to the understanding of mechanisms responsible for hepatotoxicity (disruption of cellular energy status, alteration of Ca(2+) homeostasis, inhibition of transport systems, metabolic activation, oxidative stress, covalent binding, etc.). Assessment of current cytotoxicity and hepatic-specific biochemical effects is limited by the inability to measure a wide spectrum of potential mechanistic changes involved in the drug-induced toxic injury. A convenient selection of endpoints allows a multiparametric evaluation of drug toxicity. In this regard, cytomic, proteomic, toxicogenomic and metabonomic approaches help to define patterns of hepatotoxicity for early identification of potential adverse effects of the drug to the liver.
Collapse
Affiliation(s)
- María José Gómez-Lechón
- Unidad de Hepatología Experimental, Centro de Investigación, Hospital La Fe, Valencia, Spain
| | | | | |
Collapse
|
16
|
A toxicogenomics-based parallelogram approach to evaluate the relevance of coumarin-induced responses in primary human hepatocytes in vitro for humans in vivo. Toxicol In Vitro 2009; 23:1163-9. [DOI: 10.1016/j.tiv.2009.06.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 06/02/2009] [Accepted: 06/09/2009] [Indexed: 11/23/2022]
|
17
|
Xia L, Ng S, Han R, Tuo X, Xiao G, Leo HL, Cheng T, Yu H. Laminar-flow immediate-overlay hepatocyte sandwich perfusion system for drug hepatotoxicity testing. Biomaterials 2009; 30:5927-36. [PMID: 19646750 DOI: 10.1016/j.biomaterials.2009.07.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 07/11/2009] [Indexed: 11/24/2022]
Abstract
Drug hepatotoxicity testing requires in vitro hepatocyte culture to maintain the long-term and stable liver specific functions. We developed a drug testing platform based on laminar-flow immediate-overlay hepatocyte sandwich perfusion culture. The immediate-overlay sandwich (collagen-coated porous polymeric membrane as top overlay) protects the cells and integrity of the top collagen matrix from the impact of flow. A bioreactor was designed that allowed proper control of shear stress and mass transfer. The culture parameters such as the optimal perfusion initiation time and flow rate were systematically and mechanistically determined. The optimized system could re-establish hepatocyte polarity to support biliary excretion and to maintain other liver specific functions, such as the biotransformation enzyme activities, for two weeks that extended the usable in vitro hepatocyte-based drug testing window. When the perfusion cultured hepatocytes from days 7 or 14 were used for drug testing, the APAP-induced hepatotoxicity measurements were more sensitive and consistent over time than the static culture control, enabling further exploitations in large-scale drug testing applications.
Collapse
Affiliation(s)
- Lei Xia
- Institute of Bioengineering and Nanotechnology, A*STAR, The Nanos, Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Serum-free collagen sandwich cultures of adult rat hepatocytes maintain liver-like properties long term: a valuable model for in vitro toxicity and drug-drug interaction studies. Chem Biol Interact 2009; 181:124-37. [PMID: 19482013 DOI: 10.1016/j.cbi.2009.05.015] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 05/21/2009] [Accepted: 05/22/2009] [Indexed: 12/29/2022]
Abstract
Cultures of primary hepatocytes from various species, including human, are used in several applications during pre-clinical drug development. Their use is however limited by cell survival and conservation of liver-specific functions in vitro. The differentiation status of hepatocytes in culture strongly depends on medium formulation and the extracellular matrix environment. We incubated primary rat hepatocytes for 10 days on collagen monolayer and in collagen sandwich cultures with or without serum. Restoration of polygonal cell shape and formation of functional bile canaliculi-like structures was stable only in serum-free sandwich cultures. Variations in general cell viability, as judged by the cellular ATP content, LDH release or apoptosis, were less pronounced between alternative cultures. The intracellular glutathione content was preserved close to in vivo levels especially in serum-free sandwich cultures. Basal activities of cytochrome P450 enzymes (P450) varied strongly between cultures. There was a minor effect on CYP1A but CYP2B activity was only detectable in the serum-free sandwich culture after 3 days and beyond. CYP2C activity was slightly elevated in both sandwich cultures, whereas CYP3A showed increased levels in both serum-free cultures. Inducibility of these P450s was fully maintained over time in serum-free collagen sandwich only. Gene expression was largely constant over time in serum-free sandwich cultures that was closest to liver. This liver-like property was supported by protein profiling results. Taken together, the serum-free collagen sandwich culture of primary rat hepatocytes maintained liver-like features over 10 days and is therefore a suitable model for long-term toxicity and drug-drug interaction studies.
Collapse
|
19
|
Wen YA, Liu D, Xiao YY, Luo D, Dong YF, Zhang LP. Enhanced glucose synthesis in three-dimensional hepatocyte collagen matrix. Toxicol In Vitro 2009; 23:744-7. [PMID: 19268699 DOI: 10.1016/j.tiv.2009.02.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2008] [Revised: 12/31/2008] [Accepted: 02/22/2009] [Indexed: 10/21/2022]
Abstract
Three-dimensional (3D) cell culture model offers a unique opportunity to study hepatocytes that require extracellular matrix to keep the cells at the differentiated state. In this report, we cultured isolated mouse hepatocytes in a 3D collagen matrix system and developed a protocol to measure glucose production at 3h, 6h, 18h and 24h after culture. The results demonstrated that hepatocytes cultured under 3D collagen matrix condition consistently produced glucose at 240-290 mg/10(6) cells for up to 24h. Contrarily, hepatocytes cultured under traditional monolayer condition produced less than 50 mg/10(6) cells glucose. We demonstrated higher expression of phosphoenolpyruvate carboxykinase (PEPCK), a key enzyme for the synthesis of glucose from pyruvate, and CCAAT/enhancer-binding protein alpha (C/EBPalpha), an important liver-specific transcription factor, under the 3D collagen matrix culture condition in comparison to the monolayer condition. Thus, the 3D collagen matrix system preserved metabolic function of hepatocytes and can be used as an in vitro model for studying hepatocyte glucose production and gluconeogenesis.
Collapse
Affiliation(s)
- Yang-an Wen
- Dept. of Clinical Laboratory Medicine, The First Affiliated Hospital, Chongqing Medical University, Yuzhong District, Chongqing 400016, PR China
| | | | | | | | | | | |
Collapse
|
20
|
Kienhuis AS, van de Poll MCG, Wortelboer H, van Herwijnen M, Gottschalk R, Dejong CHC, Boorsma A, Paules RS, Kleinjans JCS, Stierum RH, van Delft JHM. Parallelogram Approach Using Rat-Human In Vitro and Rat In Vivo Toxicogenomics Predicts Acetaminophen-induced Hepatotoxicity in Humans. Toxicol Sci 2008; 107:544-52. [DOI: 10.1093/toxsci/kfn237] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
21
|
Boess F, Durr E, Schaub N, Haiker M, Albertini S, Suter L. An in vitro study on 5-HT6 receptor antagonist induced hepatotoxicity based on biochemical assays and toxicogenomics. Toxicol In Vitro 2007; 21:1276-86. [PMID: 17513084 DOI: 10.1016/j.tiv.2007.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 02/15/2007] [Accepted: 03/23/2007] [Indexed: 10/23/2022]
Abstract
We investigated the effects of two 5-HT(6) receptor antagonists on rat primary hepatocytes using a combined biochemical and toxicogenomics approach. Both compounds share the same pharmacological target, but displayed strikingly different toxicity profiles in pre-clinical animal studies: While R7199 caused hepatic steatosis in rats, no hepatotoxicity was observed with R0074. Here, we partially reproduced the steatosis findings seen in vivo using primary rat hepatocytes. Biochemical analyses and gene expression results generally supported the findings observed in the animal model and also allowed the differentiation of both compounds with regards to hepatotoxic potential. In particular, the induction of Cyp 2B and Cyp 3A1 directly correlates to the findings in the livers of treated animals. The effects on genes of the steroideogenic pathway relate to the deregulation of cholesterol homeostasis. We also observed the inhibition of beta-oxidation, indicating impaired lipid metabolism. Hence, gene expression analysis in combination with biochemical parameters can provide additional insight into the possible mechanisms underlying adverse events.
Collapse
Affiliation(s)
- F Boess
- F. Hoffmann-La Roche AG, Toxicology Department, 4070 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|