101
|
Kumari S, Chatterjee K. Biomaterials-based formulations and surfaces to combat viral infectious diseases. APL Bioeng 2021; 5:011503. [PMID: 33598595 PMCID: PMC7881627 DOI: 10.1063/5.0029486] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Rapidly growing viral infections are potent risks to public health worldwide. Accessible virus-specific antiviral vaccines and drugs are therapeutically inert to emerging viruses, such as Zika, Ebola, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, discovering ways to prevent and control viral infections is among the foremost medical challenge of our time. Recently, innovative technologies are emerging that involve the development of new biomaterial-based formulations and surfaces endowed with broad-spectrum antiviral properties. Here, we review emerging biomaterials technologies for controlling viral infections. Relevant advances in biomaterials employed with nanotechnology to inactivate viruses or to inhibit virus replication and further their translation in safe and effective antiviral formulations in clinical trials are discussed. We have included antiviral approaches based on both organic and inorganic nanoparticles (NPs), which offer many advantages over molecular medicine. An insight into the development of immunomodulatory scaffolds in designing new platforms for personalized vaccines is also considered. Substantial research on natural products and herbal medicines and their potential in novel antiviral drugs are discussed. Furthermore, to control contagious viral infections, i.e., to reduce the viral load on surfaces, current strategies focusing on biomimetic anti-adhesive surfaces through nanostructured topography and hydrophobic surface modification techniques are introduced. Biomaterial surfaces functionalized with antimicrobial polymers and nanoparticles against viral infections are also discussed. We recognize the importance of research on antiviral biomaterials and present potential strategies for future directions in applying these biomaterial-based approaches to control viral infections and SARS-CoV-2.
Collapse
Affiliation(s)
- Sushma Kumari
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
102
|
刘 婷, 葛 玉, 袁 敏, 熊 巧, 赵 建. [A review on cell-based models of human liver disease in vitro]. SHENG WU YI XUE GONG CHENG XUE ZA ZHI = JOURNAL OF BIOMEDICAL ENGINEERING = SHENGWU YIXUE GONGCHENGXUE ZAZHI 2021; 38:178-184. [PMID: 33899443 PMCID: PMC10307582 DOI: 10.7507/1001-5515.202004027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/31/2020] [Indexed: 11/03/2022]
Abstract
Unhealthy diet, habits and drug abuse cause a variety of liver diseases, including steatohepatitis, liver fibrosis, liver cirrhosis and liver cancer, which seriously affect human health. The fabrication of highly simulated cell models in vitro is important in the treatment of liver diseases and drug development. This article summarized the common strategies for the construction of liver pathology models in vitro. It introduced four typical cell models in vitro related to liver disease and provided a reference for the study of liver disease models.
Collapse
Affiliation(s)
- 婷 刘
- 上海理工大学 医疗器械与食品学院(上海 200093)School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R.China
- 中国科学院 上海微系统与信息技术研究所 传感技术联合国家重点实验室(上海 200050)State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P.R.China
| | - 玉卿 葛
- 中国科学院 上海微系统与信息技术研究所 传感技术联合国家重点实验室(上海 200050)State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P.R.China
| | - 敏 袁
- 上海理工大学 医疗器械与食品学院(上海 200093)School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P.R.China
| | - 巧 熊
- 海军军医大学附属长海医院 泌尿外科(上海 200433)Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R.China
| | - 建龙 赵
- 中国科学院 上海微系统与信息技术研究所 传感技术联合国家重点实验室(上海 200050)State Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai 200050, P.R.China
| |
Collapse
|
103
|
Pieters VM, Co IL, Wu NC, McGuigan AP. Applications of Omics Technologies for Three-Dimensional In Vitro Disease Models. Tissue Eng Part C Methods 2021; 27:183-199. [PMID: 33406987 DOI: 10.1089/ten.tec.2020.0300] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Omics technologies, such as genomics, epigenomics, transcriptomics, proteomics, metabolomics, lipidomics, multiomics, and integrated modalities, have greatly contributed to our understanding of various diseases by enabling researchers to probe the molecular wiring of cellular systems in a high-throughput and precise manner. With the development of tissue-engineered three-dimensional (3D) in vitro disease models, such as organoids and spheroids, there is potential of integrating omics technologies with 3D disease models to elucidate the complex links between genotype and phenotype. These 3D disease models have been used to model cancer, infectious disease, toxicity, neurological disorders, and others. In this review, we provide an overview of omics technologies, highlight current and emerging studies, discuss the associated experimental design considerations, barriers and challenges of omics technologies, and provide an outlook on the future applications of omics technologies with 3D models. Overall, this review aims to provide a valuable resource for tissue engineers seeking to leverage omics technologies for diving deeper into biological discovery. Impact statement With the emergence of three-dimensional (3D) in vitro disease models, tissue engineers are increasingly interested to investigate these systems to address biological questions related to disease mechanism, drug target discovery, therapy resistance, and more. Omics technologies are a powerful and high-throughput approach, but their application for 3D disease models is not maximally utilized. This review illustrates the achievements and potential of using omics technologies to leverage the full potential of 3D in vitro disease models. This will improve the quality of such models, advance our understanding of disease, and contribute to therapy development.
Collapse
Affiliation(s)
- Vera M Pieters
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ileana L Co
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Nila C Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Alison P McGuigan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada.,Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada
| |
Collapse
|
104
|
Baddal B, Marrazzo P. Refining Host-Pathogen Interactions: Organ-on-Chip Side of the Coin. Pathogens 2021; 10:203. [PMID: 33668558 PMCID: PMC7918822 DOI: 10.3390/pathogens10020203] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Bioinspired organ-level in vitro platforms that recapitulate human organ physiology and organ-specific responses have emerged as effective technologies for infectious disease research, drug discovery, and personalized medicine. A major challenge in tissue engineering for infectious diseases has been the reconstruction of the dynamic 3D microenvironment reflecting the architectural and functional complexity of the human body in order to more accurately model the initiation and progression of host-microbe interactions. By bridging the gap between in vitro experimental models and human pathophysiology and providing alternatives for animal models, organ-on-chip microfluidic devices have so far been implemented in multiple research areas, contributing to major advances in the field. Given the emergence of the recent pandemic, plug-and-play organ chips may hold the key for tackling an unmet clinical need in the development of effective therapeutic strategies. In this review, latest studies harnessing organ-on-chip platforms to unravel host-pathogen interactions are presented to highlight the prospects for the microfluidic technology in infectious diseases research.
Collapse
Affiliation(s)
- Buket Baddal
- Department of Medical Microbiology and Clinical Microbiology, Faculty of Medicine, Near East University, Nicosia 99138, Cyprus
| | - Pasquale Marrazzo
- Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
105
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
106
|
Hayes CN, Chayama K. Unmet Needs in Basic Research of Hepatitis B Virus Infection: In Vitro and In Vivo Models. HEPATITIS B VIRUS AND LIVER DISEASE 2021:29-49. [DOI: 10.1007/978-981-16-3615-8_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
107
|
Bove G, Mehnert AK, Dao Thi VL. iPSCs for modeling hepatotropic pathogen infections. IPSCS FOR STUDYING INFECTIOUS DISEASES 2021:149-213. [DOI: 10.1016/b978-0-12-823808-0.00013-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
108
|
Frtús A, Smolková B, Uzhytchak M, Lunova M, Jirsa M, Hof M, Jurkiewicz P, Lozinsky VI, Wolfová L, Petrenko Y, Kubinová Š, Dejneka A, Lunov O. Hepatic Tumor Cell Morphology Plasticity under Physical Constraints in 3D Cultures Driven by YAP-mTOR Axis. Pharmaceuticals (Basel) 2020; 13:ph13120430. [PMID: 33260691 PMCID: PMC7759829 DOI: 10.3390/ph13120430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Recent studies undoubtedly show that the mammalian target of rapamycin (mTOR) and the Hippo–Yes-associated protein 1 (YAP) pathways are important mediators of mechanical cues. The crosstalk between these pathways as well as de-regulation of their signaling has been implicated in multiple tumor types, including liver tumors. Additionally, physical cues from 3D microenvironments have been identified to alter gene expression and differentiation of different cell lineages. However, it remains incompletely understood how physical constraints originated in 3D cultures affect cell plasticity and what the key mediators are of such process. In this work, we use collagen scaffolds as a model of a soft 3D microenvironment to alter cellular size and study the mechanotransduction that regulates that process. We show that the YAP-mTOR axis is a downstream effector of 3D cellular culture-driven mechanotransduction. Indeed, we found that cell mechanics, dictated by the physical constraints of 3D collagen scaffolds, profoundly affect cellular proliferation in a YAP–mTOR-mediated manner. Functionally, the YAP–mTOR connection is key to mediate cell plasticity in hepatic tumor cell lines. These findings expand the role of YAP–mTOR-driven mechanotransduction to the control hepatic tumor cellular responses under physical constraints in 3D cultures. We suggest a tentative mechanism, which coordinates signaling rewiring with cytoplasmic restructuring during cell growth in 3D microenvironments.
Collapse
Affiliation(s)
- Adam Frtús
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
| | - Barbora Smolková
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
| | - Mariia Uzhytchak
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic;
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic;
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, 18223 Prague, Czech Republic; (M.H.); (P.J.)
| | - Piotr Jurkiewicz
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, 18223 Prague, Czech Republic; (M.H.); (P.J.)
| | - Vladimir I. Lozinsky
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Vavilov Street, 28, 119991 Moscow, Russia;
| | - Lucie Wolfová
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (L.W.); (Y.P.)
- Department of Tissue Engineering, Contipro a.s., 56102 Dolni Dobrouc, Czech Republic
| | - Yuriy Petrenko
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (L.W.); (Y.P.)
| | - Šárka Kubinová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Department of Biomaterials and Biophysical Methods, Institute of Experimental Medicine of the Czech Academy of Sciences, 14220 Prague, Czech Republic; (L.W.); (Y.P.)
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Correspondence: (A.D.); (O.L.); Tel.: +420-2660-52141 (A.D.); +420-2660-52131 (O.L.)
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; (A.F.); (B.S.); (M.U.); (M.L.); (Š.K.)
- Correspondence: (A.D.); (O.L.); Tel.: +420-2660-52141 (A.D.); +420-2660-52131 (O.L.)
| |
Collapse
|
109
|
Xu Q, Ying P, Ren J, Kong N, Wang Y, Li YG, Yao Y, Kaplan DL, Ling S. Biomimetic Design for Bio-Matrix Interfaces and Regenerative Organs. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:411-429. [PMID: 33138695 DOI: 10.1089/ten.teb.2020.0234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The urgent demand for transplanted organs has motivated the development of regenerative medicine to biomimetically reconstruct the structure and function of natural tissues or organs. The prerequisites for constructing multicellular organs include specific cell sources, suitable scaffolding material, and interconnective biofunctional interfaces. As some of the most complex systems in nature, human organs, tissues, and cellular units have unique "bio-matrix" physicochemical interfaces. Human tissues support a large number of cells with distinct biofunctional interfaces for compartmentalization related to metabolism, material exchange, and physical barriers. These naturally shaped biofunctional interfaces support critical metabolic functions that drive adaptive human behavior. In contrast, mutations and disorders during organogenesis can disrupt these interfaces as a consequence of disease and trauma. To replicate the appropriate structure and physiological function of tissues and organs, the biomaterials used in these approaches should have properties that mimic those of natural biofunctional interfaces. In this review, the focus is on the biomimetic design of functional interfaces and hierarchical structures for four regenerative organs, liver, kidney, lung, heart, and the immune system. Research on these organs provides understanding of cell-matrix interactions for hierarchically bioinspired material engineering, and guidance for the design of bioartificial organs. Finally, we provide perspectives on future challenges in biofunctional interface designs and discuss the obstacles that remain toward the generation of functional bioartificial organs.
Collapse
Affiliation(s)
- Quanfu Xu
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Pei Ying
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, China
| | - Jing Ren
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Na Kong
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yang Wang
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yi-Gang Li
- Department of Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Shengjie Ling
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
110
|
Vurat MT, Şeker Ş, Lalegül-Ülker Ö, Parmaksiz M, Elçin AE, Elçin YM. Development of a multicellular 3D-bioprinted microtissue model of human periodontal ligament-alveolar bone biointerface: Towards a pre-clinical model of periodontal diseases and personalized periodontal tissue engineering. Genes Dis 2020; 9:1008-1023. [PMID: 35685479 PMCID: PMC9170773 DOI: 10.1016/j.gendis.2020.11.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/24/2020] [Accepted: 11/22/2020] [Indexed: 12/20/2022] Open
Abstract
While periodontal (PD) disease is among principal causes of tooth loss worldwide, regulation of concomitant soft and mineralized PD tissues, and PD pathogenesis have not been completely clarified yet. Besides, relevant pre-clinical models and in vitro platforms have limitations in simulating human physiology. Here, we have harnessed three-dimensional bioprinting (3DBP) technology for developing a multi-cellular microtissue model resembling PD ligament-alveolar bone (PDL-AB) biointerface for the first time. 3DBP parameters were optimized; the physical, chemical, rheological, mechanical, and thermal properties of the constructs were assessed. Constructs containing gelatin methacryloyl (Gel-MA) and hydroxyapatite-magnetic iron oxide nanoparticles showed higher level of compressive strength when compared with that of Gel-MA constructs. Bioprinted self-supporting microtissue was cultured under flow in a microfluidic platform for >10 days without significant loss of shape fidelity. Confocal microscopy analysis indicated that encapsulated cells were homogenously distributed inside the matrix and preserved their viability for >7 days under microfluidic conditions. Immunofluorescence analysis showed the cohesion of stromal cell surface marker-1+ human PDL fibroblasts containing PDL layer with the osteocalcin+ human osteoblasts containing mineralized layer in time, demonstrating some permeability of the printed constructs to cell migration. Preliminary tetracycline interaction study indicated the uptake of model drug by the cells inside the 3D-microtissue. Also, the non-toxic levels of tetracycline were determined for the encapsulated cells. Thus, the effects of tetracyclines on PDL-AB have clinical significance for treating PD diseases. This 3D-bioprinted multi-cellular periodontal/osteoblastic microtissue model has potential as an in vitro platform for studying processes of the human PDL.
Collapse
|
111
|
Huang D, Gibeley SB, Xu C, Xiao Y, Celik O, Ginsberg HN, Leong KW. Engineering liver microtissues for disease modeling and regenerative medicine. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909553. [PMID: 33390875 PMCID: PMC7774671 DOI: 10.1002/adfm.201909553] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Indexed: 05/08/2023]
Abstract
The burden of liver diseases is increasing worldwide, accounting for two million deaths annually. In the past decade, tremendous progress has been made in the basic and translational research of liver tissue engineering. Liver microtissues are small, three-dimensional hepatocyte cultures that recapitulate liver physiology and have been used in biomedical research and regenerative medicine. This review summarizes recent advances, challenges, and future directions in liver microtissue research. Cellular engineering approaches are used to sustain primary hepatocytes or produce hepatocytes derived from pluripotent stem cells and other adult tissues. Three-dimensional microtissues are generated by scaffold-free assembly or scaffold-assisted methods such as macroencapsulation, droplet microfluidics, and bioprinting. Optimization of the hepatic microenvironment entails incorporating the appropriate cell composition for enhanced cell-cell interactions and niche-specific signals, and creating scaffolds with desired chemical, mechanical and physical properties. Perfusion-based culture systems such as bioreactors and microfluidic systems are used to achieve efficient exchange of nutrients and soluble factors. Taken together, systematic optimization of liver microtissues is a multidisciplinary effort focused on creating liver cultures and on-chip models with greater structural complexity and physiological relevance for use in liver disease research, therapeutic development, and regenerative medicine.
Collapse
Affiliation(s)
- Dantong Huang
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Sarah B. Gibeley
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Ozgenur Celik
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Henry N. Ginsberg
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
112
|
Moradi E, Jalili-Firoozinezhad S, Solati-Hashjin M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomater 2020; 116:67-83. [PMID: 32890749 DOI: 10.1016/j.actbio.2020.08.041] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/22/2020] [Accepted: 08/27/2020] [Indexed: 02/08/2023]
Abstract
The liver is the largest internal organ of the body with complex microarchitecture and function that plays critical roles in drug metabolism. Hepatotoxicity and drug-induced liver injury (DILI) caused by various drugs is the main reason for late-stage drug failures. Moreover, liver diseases are among the leading causes of death in the world, with the number of new cases arising each year. Although animal models have been used to understand human drug metabolism and toxicity before clinical trials, tridimensional microphysiological systems, such as liver-on-a-chip (Liver Chip) platforms, could better recapitulate features of human liver physiology and pathophysiology and thus, are often more predictive of human outcome. Liver Chip devices have shown promising results in mimicking in vivo condition by recapitulating the sinusoidal structure of the liver, maintaining high cell viability and cellular phenotypes, and emulating native liver functions. Here, we first review the cellular constituents and physiology of the liver and then critically discuss the state-of-the-art chip-based liver models and their applications in drug screening, disease modeling, and regenerative medicine. We finally address the pending issues of existing platforms and touch upon future directions for developing new, advanced on-chip models.
Collapse
Affiliation(s)
- Ehsanollah Moradi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran
| | - Sasan Jalili-Firoozinezhad
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Mehran Solati-Hashjin
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Iran.
| |
Collapse
|
113
|
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
|
114
|
Ogoke O, Maloy M, Parashurama N. The science and engineering of stem cell-derived organoids-examples from hepatic, biliary, and pancreatic tissues. Biol Rev Camb Philos Soc 2020; 96:179-204. [PMID: 33002311 DOI: 10.1111/brv.12650] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 08/08/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
The field of organoid engineering promises to revolutionize medicine with wide-ranging applications of scientific, engineering, and clinical interest, including precision and personalized medicine, gene editing, drug development, disease modelling, cellular therapy, and human development. Organoids are a three-dimensional (3D) miniature representation of a target organ, are initiated with stem/progenitor cells, and are extremely promising tools with which to model organ function. The biological basis for organoids is that they foster stem cell self-renewal, differentiation, and self-organization, recapitulating 3D tissue structure or function better than two-dimensional (2D) systems. In this review, we first discuss the importance of epithelial organs and the general properties of epithelial cells to provide a context and rationale for organoids of the liver, pancreas, and gall bladder. Next, we develop a general framework to understand self-organization, tissue hierarchy, and organoid cultivation. For each of these areas, we provide a historical context, and review a wide range of both biological and mathematical perspectives that enhance understanding of organoids. Next, we review existing techniques and progress in hepatobiliary and pancreatic organoid engineering. To do this, we review organoids from primary tissues, cell lines, and stem cells, and introduce engineering studies when applicable. We discuss non-invasive assessment of organoids, which can reveal the underlying biological mechanisms and enable improved assays for growth, metabolism, and function. Applications of organoids in cell therapy are also discussed. Taken together, we establish a broad scientific foundation for organoids and provide an in-depth review of hepatic, biliary and pancreatic organoids.
Collapse
Affiliation(s)
- Ogechi Ogoke
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A
| | - Mitchell Maloy
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A
| | - Natesh Parashurama
- Department of Chemical and Biological Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A.,Clinical and Translation Research Center (CTRC), University at Buffalo (State University of New York), Buffalo, NY, U.S.A.,Department of Biomedical Engineering, University at Buffalo (State University of New York), Buffalo, NY, U.S.A
| |
Collapse
|
115
|
Shpichka A, Bikmulina P, Peshkova M, Kosheleva N, Zurina I, Zahmatkesh E, Khoshdel-Rad N, Lipina M, Golubeva E, Butnaru D, Svistunov A, Vosough M, Timashev P. Engineering a Model to Study Viral Infections: Bioprinting, Microfluidics, and Organoids to Defeat Coronavirus Disease 2019 (COVID-19). Int J Bioprint 2020; 6:302. [PMID: 33089000 PMCID: PMC7557357 DOI: 10.18063/ijb.v6i4.302] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
While the number of studies related to severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) is constantly growing, it is essential to provide a framework of modeling viral infections. Therefore, this review aims to describe the background presented by earlier used models for viral studies and an approach to design an "ideal" tissue model for SARS-CoV-2 infection. Due to the previous successful achievements in antiviral research and tissue engineering, combining the emerging techniques such as bioprinting, microfluidics, and organoid formation are considered to be one of the best approaches to form in vitro tissue models. The fabrication of an integrated multi-tissue bioprinted platform tailored for SARS-CoV-2 infection can be a great breakthrough that can help defeat coronavirus disease in 2019.
Collapse
Affiliation(s)
- Anastasia Shpichka
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Polina Bikmulina
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Maria Peshkova
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Nastasia Kosheleva
- Department of Molecular and Cell Pathophysiology, FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
- Department of Embryology, Lomonosov Moscow State University, Faculty of Biology, Moscow, Russia
| | - Irina Zurina
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Molecular and Cell Pathophysiology, FSBSI Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Ensieh Zahmatkesh
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Khoshdel-Rad
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Marina Lipina
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, Moscow, Russia
| | - Elena Golubeva
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Denis Butnaru
- Rector’s Office, Sechenov University, Moscow, Russia
| | | | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Peter Timashev
- Department of Advanced Biomaterials, Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
- Department of Polymers and Composites, NN Semenov Institute of Chemical Physics, Moscow, Russia
| |
Collapse
|
116
|
Ao DS, Gao LY, Gu JH, Qiao JH, Wang H, Liu YF, Song H. Study on Adenovirus Infection in vitro with Nanoself-Assembling Peptide as Scaffolds for 3D Culture. Int J Nanomedicine 2020; 15:6327-6338. [PMID: 32922004 PMCID: PMC7457861 DOI: 10.2147/ijn.s239395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/30/2020] [Indexed: 01/04/2023] Open
Abstract
Purpose To construct a three-dimensional (3D) culture model of adenovirus in vitro using the nanoself-assembling peptide RADA16-I as a 3D cell culture scaffold combined with virology experimental technology to provide a novel research method for virus isolation and culture, pathogenesis research, antiviral drug screening and vaccine preparation. Methods The nanoself-assembling peptide RADA16-I was used as a 3D scaffold material for 293T cell culture, and adenovirus was cultured in the cells. The growth, morphological characteristics and pathological effects of 3D-cultured 293T cells after adenovirus infection were observed with an inverted microscope and MTS. The proliferation of adenovirus in 293T cells was observed by TEM and detected by qPCR. The levels of TNF-α and IL-8 secreted by adenovirus-infected 293T cells in the RADA16-I 3D culture system were detected by ELISA. Results The 293T cells grew well in the RADA16-I 3D culture system for a prolonged period of time. The adenovirus infection persisted for a long time with multiple proliferation peaks, which closely resembled those of in vivo infections. The adenovirus virions amplified in the 3D system remained infectious. There were multiple secretion peaks of TNF-α and IL-8 secretion levels in adenovirus-infected 293T cells cultured in 3D culture systems. Conclusion The nanoself-assembling peptide RADA16-I can be used as a 3D scaffold for adenovirus isolation, culture and research. The 3D culture system shows more realistic in vivo effects than two-dimensional (2D) culture.
Collapse
Affiliation(s)
- Di-Shu Ao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Lu-Yao Gao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Jing-Han Gu
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Jun-Hua Qiao
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Huan Wang
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Yan-Fei Liu
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, People's Republic of China
| | - Hong Song
- Department of Microbiology, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, People's Republic of China
| |
Collapse
|
117
|
3D In Vitro Human Organ Mimicry Devices for Drug Discovery, Development, and Assessment. ADVANCES IN POLYMER TECHNOLOGY 2020. [DOI: 10.1155/2020/6187048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The past few decades have shown significant advancement as complex in vitro humanized systems have substituted animal trials and 2D in vitro studies. 3D humanized platforms mimic the organs of interest with their stimulations (physical, electrical, chemical, and mechanical). Organ-on-chip devices, including in vitro modelling of 3D organoids, 3D microfabrication, and 3D bioprinted platforms, play an essential role in drug discovery, testing, and assessment. In this article, a thorough review is provided of the latest advancements in the area of organ-on-chip devices targeting liver, kidney, lung, gut, heart, skin, and brain mimicry devices for drug discovery, development, and/or assessment. The current strategies, fabrication methods, and the specific application of each device, as well as the advantages and disadvantages, are presented for each reported platform. This comprehensive review also provides some insights on the challenges and future perspectives for the further advancement of each organ-on-chip device.
Collapse
|
118
|
Lucifora J, Michelet M, Rivoire M, Protzer U, Durantel D, Zoulim F. Two-dimensional-cultures of primary human hepatocytes allow efficient HBV infection: Old tricks still work! J Hepatol 2020; 73:449-451. [PMID: 32423632 DOI: 10.1016/j.jhep.2020.03.042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Julie Lucifora
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, France.
| | - Maud Michelet
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, France
| | | | - Ulrike Protzer
- Institute of Virology, Technical University of Munich / Helmholtz Zentrum München, Munich, Germany; German Center for Infection Research (DZIF), Munich partner site
| | - David Durantel
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, France
| | - Fabien Zoulim
- INSERM, U1052, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL1), CNRS UMR_5286, France; Department of Hepatology, Croix-Rousse Hospital, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
119
|
Tang H, Abouleila Y, Si L, Ortega-Prieto AM, Mummery CL, Ingber DE, Mashaghi A. Human Organs-on-Chips for Virology. Trends Microbiol 2020; 28:934-946. [PMID: 32674988 PMCID: PMC7357975 DOI: 10.1016/j.tim.2020.06.005] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/03/2020] [Accepted: 06/19/2020] [Indexed: 02/03/2023]
Abstract
While conventional in vitro culture systems and animal models have been used to study the pathogenesis of viral infections and to facilitate development of vaccines and therapeutics for viral diseases, models that can accurately recapitulate human responses to infection are still lacking. Human organ-on-a-chip (Organ Chip) microfluidic culture devices that recapitulate tissue–tissue interfaces, fluid flows, mechanical cues, and organ-level physiology have been developed to narrow the gap between in vitro experimental models and human pathophysiology. Here, we describe how recent developments in Organ Chips have enabled re-creation of complex pathophysiological features of human viral infections in vitro. Microfluidic Organ Chip culture devices are emerging alternatives to conventional in vitro and animal models due to their ability to replicate many structural and functional features of human physiology and disease states. Recent innovations demonstrate that Organ Chip technology is a promising strategy for virology studies where there have been successes in reproducing various viral disease phenotypes. Organ Chips have enabled investigation of many aspects of viral infection, including virus–host interactions, viral therapy-resistance evolution, and development of new antiviral therapeutics, as well as underlying pathogenesis. As Organ Chip-based assays provide accessibility to study virus-induced diseases in real time and at high resolution, they can open new avenues to uncover viral pathogenesis in a human-relevant environment and may eventually enable development of novel therapeutics and vaccines.
Collapse
Affiliation(s)
- Huaqi Tang
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Yasmine Abouleila
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Longlong Si
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | | | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Einthovenweg 20, 2333 ZD, Leiden, The Netherlands
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA; Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Vascular Biology Program and Department of Surgery, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115, USA
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.
| |
Collapse
|
120
|
Ramadan Q, Zourob M. Organ-on-a-chip engineering: Toward bridging the gap between lab and industry. BIOMICROFLUIDICS 2020; 14:041501. [PMID: 32699563 PMCID: PMC7367691 DOI: 10.1063/5.0011583] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Organ-on-a-chip (OOC) is a very ambitious emerging technology with a high potential to revolutionize many medical and industrial sectors, particularly in preclinical-to-clinical translation in the pharmaceutical arena. In vivo, the function of the organ(s) is orchestrated by a complex cellular structure and physiochemical factors within the extracellular matrix and secreted by various types of cells. The trend in in vitro modeling is to simplify the complex anatomy of the human organ(s) to the minimal essential cellular structure "micro-anatomy" instead of recapitulating the full cellular milieu that enables studying the absorption, metabolism, as well as the mechanistic investigation of drug compounds in a "systemic manner." However, in order to reflect the human physiology in vitro and hence to be able to bridge the gap between the in vivo and in vitro data, simplification should not compromise the physiological relevance. Engineering principles have long been applied to solve medical challenges, and at this stage of organ-on-a-chip technology development, the work of biomedical engineers, focusing on device engineering, is more important than ever to accelerate the technology transfer from the academic lab bench to specialized product development institutions and to the increasingly demanding market. In this paper, instead of presenting a narrative review of the literature, we systemically present a synthesis of the best available organ-on-a-chip technology from what is found, what has been achieved, and what yet needs to be done. We emphasized mainly on the requirements of a "good in vitro model that meets the industrial need" in terms of the structure (micro-anatomy), functions (micro-physiology), and characteristics of the device that hosts the biological model. Finally, we discuss the biological model-device integration supported by an example and the major challenges that delay the OOC technology transfer to the industry and recommended possible options to realize a functional organ-on-a-chip system.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| | - Mohammed Zourob
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| |
Collapse
|
121
|
D'Costa K, Kosic M, Lam A, Moradipour A, Zhao Y, Radisic M. Biomaterials and Culture Systems for Development of Organoid and Organ-on-a-Chip Models. Ann Biomed Eng 2020; 48:2002-2027. [PMID: 32285341 PMCID: PMC7334104 DOI: 10.1007/s10439-020-02498-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
The development of novel 3D tissue culture systems has enabled the in vitro study of in vivo processes, thereby overcoming many of the limitations of previous 2D tissue culture systems. Advances in biomaterials, including the discovery of novel synthetic polymers has allowed for the generation of physiologically relevant in vitro 3D culture models. A large number of 3D culture systems, aided by novel organ-on-a-chip and bioreactor technologies have been developed to improve reproducibility and scalability of in vitro organ models. The discovery of induced pluripotent stem cells (iPSCs) and the increasing number of protocols to generate iPSC-derived cell types has allowed for the generation of novel 3D models with minimal ethical limitations. The production of iPSC-derived 3D cultures has revolutionized the field of developmental biology and in particular, the study of fetal brain development. Furthermore, physiologically relevant 3D cultures generated from PSCs or adult stem cells (ASCs) have greatly advanced in vitro disease modelling and drug discovery. This review focuses on advances in 3D culture systems over the past years to model fetal development, disease pathology and support drug discovery in vitro, with a specific focus on the enabling role of biomaterials.
Collapse
Affiliation(s)
- Katya D'Costa
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Milena Kosic
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Canada
| | - Angus Lam
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Azeen Moradipour
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
- Toronto General Research Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
122
|
Gopal S, Rodrigues AL, Dordick JS. Exploiting CRISPR Cas9 in Three-Dimensional Stem Cell Cultures to Model Disease. Front Bioeng Biotechnol 2020; 8:692. [PMID: 32671050 PMCID: PMC7326781 DOI: 10.3389/fbioe.2020.00692] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
Three-dimensional (3D) cell culture methods have been widely used on a range of cell types, including stem cells to modulate precisely the cellular biophysical and biochemical microenvironment and control various cell signaling cues. As a result, more in vivo-like microenvironments are recapitulated, particularly through the formation of multicellular spheroids and organoids, which may yield more valid mechanisms of disease. Recently, genome-engineering tools such as CRISPR Cas9 have expanded the repertoire of techniques to control gene expression, which complements external signaling cues with intracellular control elements. As a result, the combination of CRISPR Cas9 and 3D cell culture methods enhance our understanding of the molecular mechanisms underpinning several disease phenotypes and may lead to developing new therapeutics that may advance more quickly and effectively into clinical candidates. In addition, using CRISPR Cas9 tools to rescue genes brings us one step closer to its use as a gene therapy tool for various degenerative diseases. Herein, we provide an overview of bridging of CRISPR Cas9 genome editing with 3D spheroid and organoid cell culture to better understand disease progression in both patient and non-patient derived cells, and we address potential remaining gaps that must be overcome to gain widespread use.
Collapse
Affiliation(s)
- Sneha Gopal
- Department of Chemical and Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - André Lopes Rodrigues
- Department of Chemical and Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal
| | - Jonathan S. Dordick
- Department of Chemical and Biological Engineering, Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY, United States
| |
Collapse
|
123
|
Ekert JE, Deakyne J, Pribul-Allen P, Terry R, Schofield C, Jeong CG, Storey J, Mohamet L, Francis J, Naidoo A, Amador A, Klein JL, Rowan W. Recommended Guidelines for Developing, Qualifying, and Implementing Complex In Vitro Models (CIVMs) for Drug Discovery. SLAS DISCOVERY 2020; 25:1174-1190. [PMID: 32495689 DOI: 10.1177/2472555220923332] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pharmaceutical industry is continuing to face high research and development (R&D) costs and low overall success rates of clinical compounds during drug development. There is an increasing demand for development and validation of healthy or disease-relevant and physiological human cellular models that can be implemented in early-stage discovery, thereby shifting attrition of future therapeutics to a point in discovery at which the costs are significantly lower. There needs to be a paradigm shift in the early drug discovery phase (which is lengthy and costly), away from simplistic cellular models that show an inability to effectively and efficiently reproduce healthy or human disease-relevant states to steer target and compound selection for safety, pharmacology, and efficacy questions. This perspective article covers the various stages of early drug discovery from target identification (ID) and validation to the hit/lead discovery phase, lead optimization, and preclinical safety. We outline key aspects that should be considered when developing, qualifying, and implementing complex in vitro models (CIVMs) during these phases, because criteria such as cell types (e.g., cell lines, primary cells, stem cells, and tissue), platform (e.g., spheroids, scaffolds or hydrogels, organoids, microphysiological systems, and bioprinting), throughput, automation, and single and multiplexing endpoints will vary. The article emphasizes the need to adequately qualify these CIVMs such that they are suitable for various applications (e.g., context of use) of drug discovery and translational research. The article ends looking to the future, in which there is an increase in combining computational modeling, artificial intelligence and machine learning (AI/ML), and CIVMs.
Collapse
Affiliation(s)
- Jason E Ekert
- In Vitro In Vivo Translation, Research, Pharmaceutical R&D, GlaxoSmithKline, Collegeville, PA, USA
| | - Julianna Deakyne
- In Vitro In Vivo Translation, Research, Pharmaceutical R&D, GlaxoSmithKline, Collegeville, PA, USA
| | - Philippa Pribul-Allen
- In Vitro In Vivo Translation, Research, Pharmaceutical R&D, GlaxoSmithKline, Ware, UK
| | - Rebecca Terry
- In Vitro In Vivo Translation, Research, Pharmaceutical R&D, GlaxoSmithKline, Ware, UK
| | - Christopher Schofield
- Functional Genomics, Medicinal Science and Technology, Pharmaceutical R&D, GlaxoSmithKline, Stevenage, UK
| | | | - Joanne Storey
- Research Office of Animal Welfare, Ethics and Strategy, Research, Pharmaceutical R&D, GlaxoSmithKline, Stevenage, UK
| | - Lisa Mohamet
- Functional Genomics, Medicinal Science and Technology, Pharmaceutical R&D, GlaxoSmithKline, Stevenage, UK
| | - Jo Francis
- Screening Profiling and Mechanistic Biology, Medicinal Science and Technology, Pharmaceutical R&D, GlaxoSmithKline, Stevenage, UK
| | - Anita Naidoo
- In Vitro In Vivo Translation, Research, Pharmaceutical R&D, GlaxoSmithKline, Ware, UK
| | - Alejandro Amador
- Functional Genomics, Medicinal Science and Technology, Pharmaceutical R&D, GlaxoSmithKline, Collegeville, PA, USA
| | - Jean-Louis Klein
- Novel Human Genetics, Research, Pharmaceutical R&D, GlaxoSmithKline, Collegeville, PA, USA
| | - Wendy Rowan
- Novel Human Genetics, Research, Pharmaceutical R&D, GlaxoSmithKline, Stevenage, UK
| |
Collapse
|
124
|
Raimondi MT, Donnaloja F, Barzaghini B, Bocconi A, Conci C, Parodi V, Jacchetti E, Carelli S. Bioengineering tools to speed up the discovery and preclinical testing of vaccines for SARS-CoV-2 and therapeutic agents for COVID-19. Theranostics 2020; 10:7034-7052. [PMID: 32641977 PMCID: PMC7330866 DOI: 10.7150/thno.47406] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
This review provides an update for the international research community on the cell modeling tools that could accelerate the understanding of SARS-CoV-2 infection mechanisms and could thus speed up the development of vaccines and therapeutic agents against COVID-19. Many bioengineering groups are actively developing frontier tools that are capable of providing realistic three-dimensional (3D) models for biological research, including cell culture scaffolds, microfluidic chambers for the culture of tissue equivalents and organoids, and implantable windows for intravital imaging. Here, we review the most innovative study models based on these bioengineering tools in the context of virology and vaccinology. To make it easier for scientists working on SARS-CoV-2 to identify and apply specific tools, we discuss how they could accelerate the discovery and preclinical development of antiviral drugs and vaccines, compared to conventional models.
Collapse
Affiliation(s)
- Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Francesca Donnaloja
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Alberto Bocconi
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Valentina Parodi
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano, Italy
| | - Stephana Carelli
- Pediatric Clinical Research Center “Fondazione Romeo ed Enrica Invernizzi”, Department of Biomedical and Clinical Sciences L. Sacco, University of Milano, Italy
| |
Collapse
|
125
|
Beg S, Alharbi KS, Alruwaili NK, Alotaibi NH, Almalki WH, Alenezi SK, Altowayan WM, Alshammari MS, Rahman M. Nanotherapeutic systems for delivering cancer vaccines: recent advances. Nanomedicine (Lond) 2020; 15:1527-1537. [PMID: 32410483 DOI: 10.2217/nnm-2020-0046] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
With an increase in the global burden of cancer-related deaths, the quest for developing new therapeutic solutions has taken momentum. In this regard, the idea of using cancer vaccines came to existence approximately 30 years ago, where gene therapy interventions have shown significant improvement in the therapeutic outcomes against several types of cancers. Cancer vaccines usually encounter a number of challenges with limited targeting ability to the tumors. Nanocarriers have been studied as a technological innovation for tumor targeting of gene therapeutics. This article provides a critical insight into the recent progress made in nanotherapeutic strategies for genetic vaccine delivery for treatment against various types of cancers. Moreover, the article intends to provide a summary of the research work being done on this topic.
Collapse
Affiliation(s)
- Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Khalid S Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Nabil K Alruwaili
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Nasser Hadal Alotaibi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Waleed H Almalki
- Department of Pharmacology & Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Sattam K Alenezi
- Department of Pharmacology & Toxicology, Unaizah College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Waleed M Altowayan
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Mohammed S Alshammari
- Department of Pharmacy Practice, Unaizah College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Mahfoozur Rahman
- Department of Pharmaceutical Sciences, SIHAS, Faculty of Health Science, Sam Higginbottom University of Agriculture, Technology & Sciences, Allahabad, India
| |
Collapse
|
126
|
Özkan A, Stolley D, Cressman ENK, McMillin M, DeMorrow S, Yankeelov TE, Rylander MN. The Influence of Chronic Liver Diseases on Hepatic Vasculature: A Liver-on-a-chip Review. MICROMACHINES 2020; 11:E487. [PMID: 32397454 PMCID: PMC7281532 DOI: 10.3390/mi11050487] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022]
Abstract
In chronic liver diseases and hepatocellular carcinoma, the cells and extracellular matrix of the liver undergo significant alteration in response to chronic injury. Recent literature has highlighted the critical, but less studied, role of the liver vasculature in the progression of chronic liver diseases. Recent advancements in liver-on-a-chip systems has allowed in depth investigation of the role that the hepatic vasculature plays both in response to, and progression of, chronic liver disease. In this review, we first introduce the structure, gradients, mechanical properties, and cellular composition of the liver and describe how these factors influence the vasculature. We summarize state-of-the-art vascularized liver-on-a-chip platforms for investigating biological models of chronic liver disease and their influence on the liver sinusoidal endothelial cells of the hepatic vasculature. We conclude with a discussion of how future developments in the field may affect the study of chronic liver diseases, and drug development and testing.
Collapse
Affiliation(s)
- Alican Özkan
- Department of Mechanical Engineering, The University of Texas, Austin, TX 78712, USA
| | - Danielle Stolley
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
| | - Erik N K Cressman
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew McMillin
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78713, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
| | - Sharon DeMorrow
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, TX 78713, USA
- Central Texas Veterans Health Care System, Temple, TX 76504, USA
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX 78712, USA
- Departments of Diagnostic Medicine, The University of Texas, Austin, TX 78712, USA
- Department of Oncology, The University of Texas, Austin, TX 78712, USA
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas, Austin, TX 78712, USA
| | - Marissa Nichole Rylander
- Department of Mechanical Engineering, The University of Texas, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas, Austin, TX 78712, USA
- Oden Institute for Computational Engineering and Sciences, The University of Texas, Austin, TX 78712, USA
| |
Collapse
|
127
|
Luan Q, Macaraniag C, Zhou J, Papautsky I. Microfluidic systems for hydrodynamic trapping of cells and clusters. BIOMICROFLUIDICS 2020; 14:031502. [PMID: 34992704 PMCID: PMC8719525 DOI: 10.1063/5.0002866] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/07/2020] [Indexed: 05/07/2023]
Abstract
Microfluidic devices have been widely applied to trapping and isolation of cells and clusters for controllable intercellular environments and high-throughput analysis, triggering numerous advances in disease diagnosis and single-cell analysis. Passive hydrodynamic cell trapping is one of the simple and effective methods that has been gaining attention in recent years. Our aim here is to review the existing passive microfluidic trapping approaches, including microposts, microfiltration, microwells, and trapping chambers, with emphasis on design principles and performance. We summarize the remarkable advances that hydrodynamic trapping methods offer, as well as the existing challenges and prospects for development. Finally, we hope that an improved understanding of hydrodynamic trapping approaches can lead to sophisticated and useful platforms to advance medical and biological research.
Collapse
Affiliation(s)
- Qiyue Luan
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Celine Macaraniag
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | | | - Ian Papautsky
- Author to whom correspondence should be addressed:. Tel.: +1 312 413 3800
| |
Collapse
|
128
|
Baddal B. Next-generation technologies for studying host-pathogen interactions: a focus on dual transcriptomics, CRISPR/Cas9 screening and organs-on-chips. Pathog Dis 2020; 77:5593955. [PMID: 31626299 DOI: 10.1093/femspd/ftz060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Pathogens constantly interact with their hosts and the environment, and therefore have evolved unique virulence mechanisms to target and breach host defense barriers and manipulate host immune response to establish an infection. Advances in technologies that allow genome mining, gene editing such as CRISPR/Cas9, genomic, epigenomic and transcriptomic studies such as dual RNA-seq, coupled with bioinformatics, have accelerated the field of host-pathogen interactions within a broad range of infection models. Underpinning of the molecular changes that accompany invasion of eukaryotic cells with pathogenic microorganisms at the intersection of host, pathogen and their local environment has provided a better understanding of infectious disease mechanisms and antimicrobial strategies. The recent evolution of physiologically relevant three-dimensional (3-D) tissue/organ models and microfluidic organ-on-chip devices also provided a window to a more predictive framework of infectious disease processes. These approaches combined hold the potential to highly impact discovery of novel drug targets and vaccine candidates of the future. Here, we review three of the available and emerging technologies-dual RNA-seq, CRISPR/Cas9 screening and organs-on-chips, applicable to the high throughput study and deciphering of interaction networks between pathogens and their hosts that are critical for the development of novel therapeutics.
Collapse
Affiliation(s)
- Buket Baddal
- Department of Medical Microbiology and Clinical Microbiology, Faculty of Medicine, Near East University, Near East Boulevard, Nicosia 99010, Cyprus
| |
Collapse
|
129
|
Wang J, Yu Y, Guo J, Lu W, Wei Q, Zhao Y. The Construction and Application of Three-Dimensional Biomaterials. ACTA ACUST UNITED AC 2020; 4:e1900238. [PMID: 32293130 DOI: 10.1002/adbi.201900238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/26/2019] [Indexed: 12/14/2022]
Abstract
Biomaterials have been widely explored and applied in many areas, especially in the field of tissue engineering. The interface of biomaterials and cells has been deeply investigated. However, it has been demonstrated that conventional 2D biomaterials fail to maintain the 3D structures and phenotypes of cells, which is the result of their limited ability to mimic the latter's complex extracellular matrix. To overcome this challenge, cell cultivation dependent on 3D biomaterials has emerged as an alternative strategy to make the recovery of 3D structures and functions of cells possible. Thus, with the thriving development of 3D cell culture in tissue engineering, a holistic review of the construction and application of 3D biomaterials is desired. Here, recent developments in 3D biomaterials for tissue engineering are reviewed. An overview of various approaches to construct 3D biomaterials, such as electro-jetting/-spinning, micro-molding, microfluidics, and 3D bio-printing, is first presented. Their typical applications in constructing cell sheets, vascular structures, cell spheroids, and macroscopic cellular constructs are described as well. Following these two sections, the current status and challenges are analyzed, as well as the future outlook of 3D biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Jie Wang
- College of Engineering, Nanjing Agricultural University, Nanjing, 210031, China.,State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yunru Yu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jiahui Guo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Wei Lu
- College of Engineering, Nanjing Agricultural University, Nanjing, 210031, China
| | - Qiong Wei
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
130
|
Mu T, Zhao X, Zhu Y, Fan H, Tang H. The E3 Ubiquitin Ligase TRIM21 Promotes HBV DNA Polymerase Degradation. Viruses 2020; 12:v12030346. [PMID: 32245233 PMCID: PMC7150939 DOI: 10.3390/v12030346] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/26/2022] Open
Abstract
The tripartite motif (TRIM) protein family is an E3 ubiquitin ligase family. Recent reports have indicated that some TRIM proteins have antiviral functions, especially against retroviruses. However, most studies mainly focus on the relationship between TRIM21 and interferon or other antiviral effectors. The effect of TRIM21 on virus-encoded proteins remains unclear. In this study, we screened candidate interacting proteins of HBV DNA polymerase (Pol) by FLAG affinity purification and mass spectrometry assay and identified TRIM21 as its regulator. We used a coimmunoprecipitation (co-IP) assay to demonstrate that TRIM21 interacted with the TP domain of HBV DNA Pol. In addition, TRIM21 promoted the ubiquitination and degradation of HBV DNA Pol using its RING domain, which has E3 ubiquitin ligase activity. Lys260 and Lys283 of HBV DNA Pol were identified as targets for ubiquitination mediated by TRIM21. Finally, we uncovered that TRIM21 degrades HBV DNA Pol to restrict HBV DNA replication, and its SPRY domain is critical for this activity. Taken together, our results indicate that TRIM21 suppresses HBV DNA replication mainly by promoting the ubiquitination of HBV DNA Pol, which may provide a new potential target for the treatment of HBV.
Collapse
Affiliation(s)
| | | | | | | | - Hua Tang
- Correspondence: ; Tel./Fax: +86-22-2354-2503
| |
Collapse
|
131
|
Ou Q, Guo J, Zeng Y, Chen H. Insights for clinical diagnostic indicators of virus and host in chronic hepatitis B infection. J Viral Hepat 2020; 27:224-232. [PMID: 31954089 DOI: 10.1111/jvh.13260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 10/19/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
Covalently closed circular DNA (cccDNA), which is stably present in the nucleus of hepatocytes, is an important indicator for evaluating antiviral efficacy. Since cccDNA quantification requires an invasive procedure, serum biological markers that can effectively reflect the transcriptional activity of intrahepatic virus and the efficacy of treatment are required. Here, from the aspects of virus and host, we outline the focus of clinical research of HBV in recent years, including HBV RNA, empty virus, hepatitis B core-related antigen and changes in the immune response. We briefly discuss their significance in predicting disease activity and monitoring treatment response in chronic hepatitis B. On this basis, some issues worthy of attention in laboratory diagnosis are proposed.
Collapse
Affiliation(s)
- Qishui Ou
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,The Genetic Diagnostic Laboratory, Fujian Medical University, Fuzhou, China.,First Clinical College, Fujian Medical University, Fuzhou, China
| | - Jianhui Guo
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,The Genetic Diagnostic Laboratory, Fujian Medical University, Fuzhou, China.,First Clinical College, Fujian Medical University, Fuzhou, China
| | - Yongbin Zeng
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,The Genetic Diagnostic Laboratory, Fujian Medical University, Fuzhou, China.,First Clinical College, Fujian Medical University, Fuzhou, China
| | - Huijuan Chen
- Department of Laboratory Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.,The Genetic Diagnostic Laboratory, Fujian Medical University, Fuzhou, China.,First Clinical College, Fujian Medical University, Fuzhou, China
| |
Collapse
|
132
|
Means S, Ali MA, Ho H, Heffernan J. Mathematical Modeling for Hepatitis B Virus: Would Spatial Effects Play a Role and How to Model It? Front Physiol 2020; 11:146. [PMID: 32158403 PMCID: PMC7052012 DOI: 10.3389/fphys.2020.00146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/11/2020] [Indexed: 12/23/2022] Open
Affiliation(s)
- Shawn Means
- School of Natural and Computational Sciences, Massey University, Auckland, New Zealand
| | - Md A Ali
- Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Harvey Ho
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Jane Heffernan
- Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| |
Collapse
|
133
|
Peterson NC, Mahalingaiah PK, Fullerton A, Di Piazza M. Application of microphysiological systems in biopharmaceutical research and development. LAB ON A CHIP 2020; 20:697-708. [PMID: 31967156 DOI: 10.1039/c9lc00962k] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Within the last 10 years, several tissue microphysiological systems (MPS) have been developed and characterized for retention of morphologic characteristics and specific gene/protein expression profiles from their natural in vivo state. Once developed, their utility is typically further tested by comparing responses to known toxic small-molecule pharmaceuticals in efforts to develop strategies for further toxicity testing of compounds under development. More recently, application of this technology in biopharmaceutical (large molecules) development is beginning to be more appreciated. In this review, we describe some of the advances made for tissue-specific MPS and outline the advantages and challenges of applying and further developing MPS technology in preclinical biopharmaceutical research.
Collapse
Affiliation(s)
- Norman C Peterson
- Clinical Pharmacology and Safety Sciences, AstraZeneca, One Medimmune Way, Gaithersburg, MD 20878, USA.
| | | | | | - Matteo Di Piazza
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Rd, Ridgefield, CT 06877, USA
| |
Collapse
|
134
|
Argentati C, Tortorella I, Bazzucchi M, Morena F, Martino S. Harnessing the Potential of Stem Cells for Disease Modeling: Progress and Promises. J Pers Med 2020; 10:E8. [PMID: 32041088 PMCID: PMC7151621 DOI: 10.3390/jpm10010008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/18/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
Ex vivo cell/tissue-based models are an essential step in the workflow of pathophysiology studies, assay development, disease modeling, drug discovery, and development of personalized therapeutic strategies. For these purposes, both scientific and pharmaceutical research have adopted ex vivo stem cell models because of their better predictive power. As matter of a fact, the advancing in isolation and in vitro expansion protocols for culturing autologous human stem cells, and the standardization of methods for generating patient-derived induced pluripotent stem cells has made feasible to generate and investigate human cellular disease models with even greater speed and efficiency. Furthermore, the potential of stem cells on generating more complex systems, such as scaffold-cell models, organoids, or organ-on-a-chip, allowed to overcome the limitations of the two-dimensional culture systems as well as to better mimic tissues structures and functions. Finally, the advent of genome-editing/gene therapy technologies had a great impact on the generation of more proficient stem cell-disease models and on establishing an effective therapeutic treatment. In this review, we discuss important breakthroughs of stem cell-based models highlighting current directions, advantages, and limitations and point out the need to combine experimental biology with computational tools able to describe complex biological systems and deliver results or predictions in the context of personalized medicine.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Ilaria Tortorella
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy; (C.A.); (I.T.); (M.B.); (F.M.)
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy
| |
Collapse
|
135
|
Cebola I. Liver gene regulatory networks: Contributing factors to nonalcoholic fatty liver disease. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1480. [PMID: 32020788 DOI: 10.1002/wsbm.1480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/17/2022]
Abstract
Metabolic diseases such as nonalcoholic fatty liver disease (NAFLD) result from complex interactions between intrinsic and extrinsic factors, including genetics and exposure to obesogenic environments. These risk factors converge in aberrant gene expression patterns in the liver, which are underlined by altered cis-regulatory networks. In homeostasis and in disease states, liver cis-regulatory networks are established by coordinated action of liver-enriched transcription factors (TFs), which define enhancer landscapes, activating broad gene programs with spatiotemporal resolution. Recent advances in DNA sequencing have dramatically expanded our ability to map active transcripts, enhancers and TF cistromes, and to define the 3D chromatin topology that contains these elements. Deployment of these technologies has allowed investigation of the molecular processes that regulate liver development and metabolic homeostasis. Moreover, genomic studies of NAFLD patients and NAFLD models have demonstrated that the liver undergoes pervasive regulatory rewiring in NAFLD, which is reflected by aberrant gene expression profiles. We have therefore achieved an unprecedented level of detail in the understanding of liver cis-regulatory networks, particularly in physiological conditions. Future studies should aim to map active regulatory elements with added levels of resolution, addressing how the chromatin landscapes of different cell lineages contribute to and are altered in NAFLD and NAFLD-associated metabolic states. Such efforts would provide additional clues into the molecular factors that trigger this disease. This article is categorized under: Biological Mechanisms > Metabolism Biological Mechanisms > Regulatory Biology Laboratory Methods and Technologies > Genetic/Genomic Methods.
Collapse
Affiliation(s)
- Inês Cebola
- Department of Metabolism, Digestion and Reproduction, Section of Genetics and Genomics, Imperial College London, London, UK
| |
Collapse
|
136
|
de Almeida Monteiro Melo Ferraz M, Nagashima JB, Venzac B, Le Gac S, Songsasen N. A dog oviduct-on-a-chip model of serous tubal intraepithelial carcinoma. Sci Rep 2020; 10:1575. [PMID: 32005926 PMCID: PMC6994655 DOI: 10.1038/s41598-020-58507-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 01/16/2020] [Indexed: 12/29/2022] Open
Abstract
Ovarian cancer is the fifth cause of cancer-related mortality in women, with an expected 5-year survival rate of only 47%. High-grade serous carcinoma (HGSC), an epithelial cancer phenotype, is the most common malignant ovarian cancer. It is known that the precursors of HGSC originate from secretory epithelial cells within the Fallopian tube, which first develops as serous tubal intraepithelial carcinoma (STIC). Here, we used gene editing by CRISPR-Cas9 to knock out the oncogene p53 in dog oviductal epithelia cultured in a dynamic microfluidic chip to create an in vitro model that recapitulated human STIC. Similar to human STIC, the gene-edited oviduct-on-a-chip, exhibited loss of cell polarization and had reduced ciliation, increased cell atypia and proliferation, with multilayered epithelium, increased Ki67, PAX8 and Myc and decreased PTEN and RB1 mRNA expression. This study provides a biomimetic in vitro model to study STIC progression and to identify potential biomarkers for early detection of HGSC.
Collapse
Affiliation(s)
| | - Jennifer Beth Nagashima
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, 1500 Remount Road, Front Royal, Virginia, 22630, USA
| | - Bastien Venzac
- Applied Microfluidics for Bioengineering Research, MESA+ Institute for Nanotechnology and TechMed Center, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for Bioengineering Research, MESA+ Institute for Nanotechnology and TechMed Center, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Nucharin Songsasen
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, 1500 Remount Road, Front Royal, Virginia, 22630, USA
| |
Collapse
|
137
|
Wang F, Shen F, Wang Y, Li Z, Chen J, Yuan Z. Residues Asn118 and Glu119 of hepatitis B virus X protein are critical for HBx-mediated inhibition of RIG-I-MAVS signaling. Virology 2020; 539:92-103. [PMID: 31706164 DOI: 10.1016/j.virol.2019.10.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 06/24/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022]
Abstract
Hepatitis B virus (HBV) X protein (HBx) has been reported to counteract the innate immune responses through interfering with the pattern recognition receptors signaling activated by retinoic acid-inducible gene-I (RIG-I)-mitochondrial antiviral signaling protein (MAVS). Here, we showed that, compared to the HBx derived from genotype (gt) A, C and D, HBx of gtB exhibited more potent inhibitory activity on the RIG-I-MAVS-mediated interferon-β promoter activation. Functional analysis of the genotype-associated differences in amino acid sequence and the reciprocal mutation experiments in transient-transfection and infection cell models revealed that HBx with asparagine (N) and glutamic acid (E) at 118-119 positions inhibited RIG-I signaling and interacted with MAVS more efficiently than that with lysine (K) and aspartic acid (D). An impaired RIG-I-induced MAVS aggregation was observed in the presence of HBx-118N119E while MAVS-TRAF3 interaction was not affected. These results implicated that HBx gene heterogeneity may affect the innate immune responses to HBV infection.
Collapse
Affiliation(s)
- Fan Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Fang Shen
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yang Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ze Li
- Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jieliang Chen
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Zhenghong Yuan
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
138
|
Winer BY, Gaska JM, Lipkowitz G, Bram Y, Parekh A, Parsons L, Leach R, Jindal R, Cho CH, Shrirao A, Novik E, Schwartz RE, Ploss A. Analysis of Host Responses to Hepatitis B and Delta Viral Infections in a Micro-scalable Hepatic Co-culture System. Hepatology 2020; 71:14-30. [PMID: 31206195 PMCID: PMC6917996 DOI: 10.1002/hep.30815] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
Hepatitis B virus (HBV) remains a major global health problem with 257 million chronically infected individuals worldwide, of whom approximately 20 million are co-infected with hepatitis delta virus (HDV). Progress toward a better understanding of the complex interplay between these two viruses and the development of novel therapies have been hampered by the scarcity of suitable cell culture models that mimic the natural environment of the liver. Here, we established HBV and HBV/HDV co-infections and super-infections in self-assembling co-cultured primary human hepatocytes (SACC-PHHs) for up to 28 days in a 384-well format and highlight the suitability of this platform for high-throughput drug testing. We performed RNA sequencing at days 8 and 28 on SACC-PHHs, either HBV mono-infected or HBV/HDV co-infected. Our transcriptomic analysis demonstrates that hepatocytes in SACC-PHHs maintain a mature hepatic phenotype over time, regardless of infection condition. We confirm that HBV is a stealth virus, as it does not induce a strong innate immune response; rather, oxidative phosphorylation and extracellular matrix-receptor interactions are dysregulated to create an environment that promotes persistence. Notably, HDV co-infection also did not lead to statistically significant transcriptional changes across multiple donors and replicates. The lack of innate immune activation is not due to SACC-PHHs being impaired in their ability to induce interferon stimulated genes (ISGs). Rather, polyinosinic:polycytidylic acid exposure activates ISGs, and this stimulation significantly inhibits HBV infection, yet only minimally affects the ability of HDV to infect and persist. Conclusion: These data demonstrate that the SACC-PHH system is a versatile platform for studying HBV/HDV co-infections and holds promise for performing chemical library screens and improving our understanding of the host response to such infections.
Collapse
Affiliation(s)
- Benjamin Y. Winer
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Jenna M. Gaska
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Gabriel Lipkowitz
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Yaron Bram
- Division of Gastroenterology & Hepatology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Amit Parekh
- Hurel® Corporation, North Brunswick, NJ 08902, USA
| | - Lance Parsons
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Robert Leach
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Rohit Jindal
- Hurel® Corporation, North Brunswick, NJ 08902, USA
| | - Cheul H. Cho
- Hurel® Corporation, North Brunswick, NJ 08902, USA
| | - Anil Shrirao
- Hurel® Corporation, North Brunswick, NJ 08902, USA
| | - Eric Novik
- Hurel® Corporation, North Brunswick, NJ 08902, USA
| | - Robert E. Schwartz
- Division of Gastroenterology & Hepatology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
139
|
Underhill GH, Khetani SR. Emerging trends in modeling human liver disease in vitro. APL Bioeng 2019; 3:040902. [PMID: 31893256 PMCID: PMC6930139 DOI: 10.1063/1.5119090] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/29/2019] [Indexed: 12/18/2022] Open
Abstract
The liver executes 500+ functions, such as protein synthesis, xenobiotic metabolism, bile production, and metabolism of carbohydrates/fats/proteins. Such functions can be severely degraded by drug-induced liver injury, nonalcoholic fatty liver disease, hepatitis B and viral infections, and hepatocellular carcinoma. These liver diseases, which represent a significant global health burden, are the subject of novel drug discovery by the pharmaceutical industry via the use of in vitro models of the human liver, given significant species-specific differences in disease profiles and drug outcomes. Isolated primary human hepatocytes (PHHs) are a physiologically relevant cell source to construct such models; however, these cells display a rapid decline in the phenotypic function within conventional 2-dimensional monocultures. To address such a limitation, several engineered platforms have been developed such as high-throughput cellular microarrays, micropatterned cocultures, self-assembled spheroids, bioprinted tissues, and perfusion devices; many of these platforms are being used to coculture PHHs with liver nonparenchymal cells to model complex cell cross talk in liver pathophysiology. In this perspective, we focus on the utility of representative platforms for mimicking key features of liver dysfunction in the context of chronic liver diseases and liver cancer. We further discuss pending issues that will need to be addressed in this field moving forward. Collectively, these in vitro liver disease models are being increasingly applied toward the development of new therapeutics that display an optimal balance of safety and efficacy, with a focus on expediting development, reducing high costs, and preventing harm to patients.
Collapse
Affiliation(s)
- Gregory H. Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | - Salman R. Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
140
|
Parrish J, Lim K, Zhang B, Radisic M, Woodfield TBF. New Frontiers for Biofabrication and Bioreactor Design in Microphysiological System Development. Trends Biotechnol 2019; 37:1327-1343. [PMID: 31202544 PMCID: PMC6874730 DOI: 10.1016/j.tibtech.2019.04.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 01/05/2023]
Abstract
Microphysiological systems (MPSs) have been proposed as an improved tool to recreate the complex biological features of the native niche with the goal of improving in vitro-in vivo extrapolation. In just over a decade, MPS technologies have progressed from single-tissue chips to multitissue plates with integrated pumps for perfusion. Concurrently, techniques for biofabrication of complex 3D constructs for regenerative medicine and 3D in vitro models have evolved into a diverse toolbox for micrometer-scale deposition of cells and cell-laden bioinks. However, as the complexity of biological models increases, experimental throughput is often compromised. This review discusses the existing disparity between MPS complexity and throughput, then examines an MPS-terminated biofabrication line to identify the hurdles and potential approaches to overcoming this disparity.
Collapse
Affiliation(s)
- Jonathon Parrish
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, New Zealand; New Zealand Medical Technologies Centre of Research Excellence (MedTech CoRE), Auckland, New Zealand
| | - Khoon Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, New Zealand; New Zealand Medical Technologies Centre of Research Excellence (MedTech CoRE), Auckland, New Zealand
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Milica Radisic
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada; Institute for Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Toronto General Research Institute, University Health Network, Toronto, ON, Canada; The Heart and Stroke/Richard Lewar Centre of Excellence, Toronto, ON, Canada
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, New Zealand; New Zealand Medical Technologies Centre of Research Excellence (MedTech CoRE), Auckland, New Zealand.
| |
Collapse
|
141
|
Immunopathogenesis of HBV Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1179:71-107. [DOI: 10.1007/978-981-13-9151-4_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
142
|
Kostrzewski T, Maraver P, Ouro-Gnao L, Levi A, Snow S, Miedzik A, Rombouts K, Hughes D. A Microphysiological System for Studying Nonalcoholic Steatohepatitis. Hepatol Commun 2019; 4:77-91. [PMID: 31909357 PMCID: PMC6939502 DOI: 10.1002/hep4.1450] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the most severe form of nonalcoholic fatty liver disease (NAFLD), which to date has no approved drug treatments. There is an urgent need for better understanding of the genetic and molecular pathways that underlie NAFLD/NASH, and currently available preclinical models, be they in vivo or in vitro, do not fully represent key aspects of the human disease state. We have developed a human in vitro co‐culture NASH model using primary human hepatocytes, Kupffer cells and hepatic stellate cells, which are cultured together as microtissues in a perfused three‐dimensional microphysiological system (MPS). The microtissues were cultured in medium containing free fatty acids for at least 2 weeks, to induce a NASH‐like phenotype. The co‐culture microtissues within the MPS display a NASH‐like phenotype, showing key features of the disease including hepatic fat accumulation, the production of an inflammatory milieu, and the expression of profibrotic markers. Addition of lipopolysaccharide resulted in a more pro‐inflammatory milieu. In the model, obeticholic acid ameliorated the NASH phenotype. Microtissues were formed from both wild‐type and patatin‐like phospholipase domain containing 3 (PNPLA3) I148M mutant hepatic stellate cells. Stellate cells carrying the mutation enhanced the overall disease state of the model and in particular produced a more pro‐inflammatory milieu. Conclusion: The MPS model displays a phenotype akin to advanced NAFLD or NASH and has utility as a tool for exploring mechanisms underlying the disease. Furthermore, we demonstrate that in co‐culture the PNPLA3 I148M mutation alone can cause hepatic stellate cells to enhance the overall NASH disease phenotype.
Collapse
Affiliation(s)
| | - Paloma Maraver
- CN Bio Innovations Ltd. Welwyn Garden City Hertfordshire United Kingdom
| | - Larissa Ouro-Gnao
- CN Bio Innovations Ltd. Welwyn Garden City Hertfordshire United Kingdom
| | - Ana Levi
- Institute for Liver and Digestive Health, Regenerative Medicine and Fibrosis Group, Royal Free University College London United Kingdom
| | - Sophie Snow
- CN Bio Innovations Ltd. Welwyn Garden City Hertfordshire United Kingdom
| | - Alina Miedzik
- CN Bio Innovations Ltd. Welwyn Garden City Hertfordshire United Kingdom
| | - Krista Rombouts
- Institute for Liver and Digestive Health, Regenerative Medicine and Fibrosis Group, Royal Free University College London United Kingdom
| | - David Hughes
- CN Bio Innovations Ltd. Welwyn Garden City Hertfordshire United Kingdom
| |
Collapse
|
143
|
Sa-Ngiamsuntorn K, Thongsri P, Pewkliang Y, Wongkajornsilp A, Kongsomboonchoke P, Suthivanich P, Borwornpinyo S, Hongeng S. An Immortalized Hepatocyte-like Cell Line (imHC) Accommodated Complete Viral Lifecycle, Viral Persistence Form, cccDNA and Eventual Spreading of a Clinically-Isolated HBV. Viruses 2019; 11:E952. [PMID: 31623162 PMCID: PMC6832882 DOI: 10.3390/v11100952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
More than 350 million people worldwide have been persistently infected with the hepatitis B virus (HBV). Chronic HBV infection could advance toward liver cirrhosis and hepatocellular carcinoma. The intervention with prophylactic vaccine and conventional treatment could suppress HBV, but could not completely eradicate it. The major obstacle for investigating curative antiviral drugs are the incompetence of hepatocyte models that should have closely imitated natural human infection. Here, we demonstrated that an immortalized hepatocyte-like cell line (imHC) could accommodate for over 30 days the entire life cycle of HBV prepared from either established cultured cells or clinically-derived fresh isolates. Normally, imHCs had intact interferon signaling with anti-viral action. Infected imHCs responded to treatments with direct-acting antiviral drugs (DAAs) and interferons (IFNs) by diminishing HBV DNA, the covalently closed circular DNA (cccDNA) surface antigen of HBV (HBsAg, aka the Australia antigen) and the hepatitis B viral protein (HBeAg). Notably, we could observe and quantify HBV spreading from infected cells to naïve cells using an imHC co-culture model. In summary, this study constructed a convenient HBV culture model that allows the screening for novel anti-HBV agents with versatile targets, either HBV entry, replication or cccDNA formation. Combinations of agents aiming at different targets should achieve a complete HBV eradication.
Collapse
Affiliation(s)
- Khanit Sa-Ngiamsuntorn
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Piyanoot Thongsri
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Yongyut Pewkliang
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Adisak Wongkajornsilp
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | | | - Phichaya Suthivanich
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
144
|
Deng J, Wei W, Chen Z, Lin B, Zhao W, Luo Y, Zhang X. Engineered Liver-on-a-Chip Platform to Mimic Liver Functions and Its Biomedical Applications: A Review. MICROMACHINES 2019; 10:E676. [PMID: 31591365 PMCID: PMC6843249 DOI: 10.3390/mi10100676] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/03/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Hepatology and drug development for liver diseases require in vitro liver models. Typical models include 2D planar primary hepatocytes, hepatocyte spheroids, hepatocyte organoids, and liver-on-a-chip. Liver-on-a-chip has emerged as the mainstream model for drug development because it recapitulates the liver microenvironment and has good assay robustness such as reproducibility. Liver-on-a-chip with human primary cells can potentially correlate clinical testing. Liver-on-a-chip can not only predict drug hepatotoxicity and drug metabolism, but also connect other artificial organs on the chip for a human-on-a-chip, which can reflect the overall effect of a drug. Engineering an effective liver-on-a-chip device requires knowledge of multiple disciplines including chemistry, fluidic mechanics, cell biology, electrics, and optics. This review first introduces the physiological microenvironments in the liver, especially the cell composition and its specialized roles, and then summarizes the strategies to build a liver-on-a-chip via microfluidic technologies and its biomedical applications. In addition, the latest advancements of liver-on-a-chip technologies are discussed, which serve as a basis for further liver-on-a-chip research.
Collapse
Affiliation(s)
- Jiu Deng
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Wenbo Wei
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Zongzheng Chen
- Integrated Chinese and Western Medicine Postdoctoral research station, Jinan University, Guangzhou 510632, China;
| | - Bingcheng Lin
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Weijie Zhao
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Yong Luo
- State Key Laboratory of Fine Chemicals, Department of Chemical Engineering, Dalian University of Technology, Dalian 116024, China; (J.D.); (W.W.); (W.Z.); (Y.L.)
| | - Xiuli Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
145
|
Sakthivel K, O'Brien A, Kim K, Hoorfar M. Microfluidic analysis of heterotypic cellular interactions: A review of techniques and applications. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.03.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
146
|
König A, Yang J, Jo E, Park KHP, Kim H, Than TT, Song X, Qi X, Dai X, Park S, Shum D, Ryu WS, Kim JH, Yoon SK, Park JY, Ahn SH, Han KH, Gerlich WH, Windisch MP. Efficient long-term amplification of hepatitis B virus isolates after infection of slow proliferating HepG2-NTCP cells. J Hepatol 2019; 71:289-300. [PMID: 31077792 DOI: 10.1016/j.jhep.2019.04.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS As hepatitis B virus (HBV) spreads through the infected liver it is simultaneously secreted into the blood. HBV-susceptible in vitro infection models do not efficiently amplify viral progeny or support cell-to-cell spread. We sought to establish a cell culture system for the amplification of infectious HBV from clinical specimens. METHODS An HBV-susceptible sodium-taurocholate cotransporting polypeptide-overexpressing HepG2 cell clone (HepG2-NTCPsec+) producing high titers of infectious progeny was selected. Secreted HBV progeny were characterized by native gel electrophoresis and electron microscopy. Comparative RNA-seq transcriptomics was performed to quantify the expression of host proviral and restriction factors. Viral spread routes were evaluated using HBV entry- or replication inhibitors, visualization of viral cell-to-cell spread in reporter cells, and nearest neighbor infection determination. Amplification kinetics of HBV genotypes B-D were analyzed. RESULTS Infected HepG2-NTCPsec+ secreted high levels of large HBV surface protein-enveloped infectious HBV progeny with typical appearance under electron microscopy. RNA-seq transcriptomics revealed that HBV does not induce significant gene expression changes in HepG2-NTCPsec+, however, transcription factors favoring HBV amplification were more strongly expressed than in less permissive HepG2-NTCPsec-. Upon inoculation with HBV-containing patient sera, rates of infected cells increased from 10% initially to 70% by viral spread to adjacent cells, and viral progeny and antigens were efficiently secreted. HepG2-NTCPsec+ supported up to 1,300-fold net amplification of HBV genomes depending on the source of virus. Viral spread and amplification were abolished by entry and replication inhibitors; viral rebound was observed after inhibitor discontinuation. CONCLUSIONS The novel HepG2-NTCPsec+ cells efficiently support the complete HBV life cycle, long-term viral spread and amplification of HBV derived from patients or cell culture, resembling relevant features of HBV-infected patients. LAY SUMMARY Currently available laboratory systems are unable to reproduce the dynamics of hepatitis B virus (HBV) spread through the infected liver and release into the blood. We developed a slowly dividing liver-derived cell line which multiplies infectious viral particles upon inoculation with patient- or cell culture-derived HBV. This new infection model can improve therapy by measuring, in advance, the sensitivity of a patient's HBV strain to specific antiviral drugs.
Collapse
Affiliation(s)
- Alexander König
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Jaewon Yang
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Eunji Jo
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Kyu Ho Paul Park
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Hyun Kim
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea; Division of Bio-Medical Science and Technology, University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Thoa Thi Than
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Xiyong Song
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Xiaoxuan Qi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Xinghong Dai
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Soonju Park
- Screening Discovery Platform, Institut Pasteur Korea, Seongnam-si, South Korea
| | - David Shum
- Screening Discovery Platform, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Wang-Shick Ryu
- Department of Biochemistry, Yonsei University, Seoul, South Korea
| | - Jung-Hee Kim
- Catholic University Liver Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Seung Kew Yoon
- Catholic University Liver Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Marc Peter Windisch
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea; Division of Bio-Medical Science and Technology, University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon, South Korea.
| |
Collapse
|
147
|
Li N, Zhang W, Li Y, Lin JM. Analysis of cellular biomolecules and behaviors using microfluidic chip and fluorescence method. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.05.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
148
|
Wang J, Wang C, Xu N, Liu ZF, Pang DW, Zhang ZL. A virus-induced kidney disease model based on organ-on-a-chip: Pathogenesis exploration of virus-related renal dysfunctions. Biomaterials 2019; 219:119367. [PMID: 31344514 DOI: 10.1016/j.biomaterials.2019.119367] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 07/13/2019] [Accepted: 07/14/2019] [Indexed: 02/03/2023]
Abstract
Renal dysfunctions usually happen in viral infections and many viruses specially infect distal renal tubules, however the pathogenesis remains unknown. Here, in order to explore the pathogenesis of virus-related renal dysfunctions, a Pseudorabies Virus (PrV) induced kidney disease model was built on a distal tubule-on-a-chip (DTC), for the first time. The barrier structure and Na reabsorption of distal renal tubules were successfully reconstituted in DTCs. After PrV infection, results showed electrolyte regulation dysfunction in Na reabsorption for the disordered Na transporters, the broken reabsorption barrier, and the transformed microvilli. And it would lead to virus induced serum electrolyte abnormalities. This work brought us a new cognition about the advantages of organ-on-a-chip (OOC) in virus research, for it had given us a better insight into the pathogenesis of virus induced dysfunctions, based on its unique ability in function reproduction.
Collapse
Affiliation(s)
- Ji Wang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, and State Key Laboratory of Virology, Wuhan University, Wuhan 430072, PR China
| | - Cheng Wang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, and State Key Laboratory of Virology, Wuhan University, Wuhan 430072, PR China
| | - Na Xu
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, and State Key Laboratory of Virology, Wuhan University, Wuhan 430072, PR China
| | - Zheng-Fei Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, PR China.
| | - Dai-Wen Pang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, and State Key Laboratory of Virology, Wuhan University, Wuhan 430072, PR China
| | - Zhi-Ling Zhang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, and State Key Laboratory of Virology, Wuhan University, Wuhan 430072, PR China.
| |
Collapse
|
149
|
Kinetics of Early Innate Immune Activation during HIV-1 Infection of Humanized Mice. J Virol 2019; 93:JVI.02123-18. [PMID: 30867315 PMCID: PMC6532090 DOI: 10.1128/jvi.02123-18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/01/2019] [Indexed: 02/07/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection is associated with aberrant immune activation; however, most model systems for HIV-1 have been used during established infection. Here, we utilize ultrasensitive HIV-1 quantification to delineate early events during the eclipse, burst, and chronic phases of HIV-1 infection in humanized mice. We show that very early in infection, HIV-1 suppresses peripheral type I interferon (IFN) and interferon-stimulated gene (ISG) responses, including the HIV-1 restriction factor IFI44. At the peak of innate immune activation, prior to CD4 T cell loss, HIV-1 infection differentially affects peripheral and lymphoid Toll-like receptor (TLR) expression profiles in T cells and macrophages. This results in a trend toward an altered activation of nuclear factor κB (NF-κB), TANK-binding kinase 1 (TBK1), and interferon regulatory factor 3 (IRF3). The subsequent type I and III IFN responses result in preferential induction of peripheral ISG responses. Following this initial innate immune activation, peripheral expression of the HIV-1 restriction factor SAM domain- and HD domain-containing protein 1 (SAMHD1) returns to levels below those observed in uninfected mice, suggesting that HIV-1 interferes with their basal expression. However, peripheral cells still retain their responsiveness to exogenous type I IFN, whereas splenic cells show a reduction in select ISGs in response to IFN. This demonstrates the highly dynamic nature of very early HIV-1 infection and suggests that blocks to the induction of HIV-1 restriction factors contribute to the establishment of viral persistence.IMPORTANCE Human immunodeficiency virus type 1 (HIV-1) infection is restricted to humans and some nonhuman primates (e.g., chimpanzee and gorilla). Alternative model systems based on simian immunodeficiency virus (SIV) infection of macaques are available but do not recapitulate all aspects of HIV-1 infection and disease. Humanized mice, which contain a human immune system, can be used to study HIV-1, but only limited information on early events and immune responses is available to date. Here, we describe very early immune responses to HIV-1 and demonstrate a suppression of cell-intrinsic innate immunity. Furthermore, we show that HIV-1 infection interacts differently with innate immune responses in blood and lymphoid organs.
Collapse
|
150
|
Evaluation of Drug Biliary Excretion Using Sandwich-Cultured Human Hepatocytes. Eur J Drug Metab Pharmacokinet 2019; 44:13-30. [PMID: 30167999 DOI: 10.1007/s13318-018-0502-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Evaluation of hepatobiliary transport of drugs is an important challenge, notably during the development of new molecular identities. In this context, sandwich-cultured human hepatocytes (SCHH) have been proposed as an interesting and integrated tool for predicting in vitro biliary excretion of drugs. The present review was therefore designed to summarize key findings about SCHH, including their establishment, their main functional features and their use for the determination of canalicular transport and the prediction of in vivo biliary clearance and hepatobiliary excretion-related drug-drug interactions. Reviewed data highlight the fact that SCHH represent an original and probably unique holistic in vitro approach to predict biliary clearance in humans, through taking into account sinusoidal drug uptake, passive drug diffusion, drug metabolism and sinusoidal and canalicular drug efflux. Limits and proposed refinements for SCHH-based analysis of drug biliary excretion, as well as putative human alternative in vitro models to SCHH are also discussed.
Collapse
|