CC BY-NC-ND 4.0 · Semin Liver Dis 2023; 43(01): 031-049
DOI: 10.1055/a-1981-5944
Review Article

Physiomimetic In Vitro Human Models for Viral Infection in the Liver

Dennis McDuffie
1   Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
,
David Barr
2   Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
,
Madeline Helm
1   Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
,
Thomas Baumert
3   Inserm Research Institute for Viral and Liver Diseases, University of Strasbourg, Strasbourg, France
,
Ashutosh Agarwal
1   Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
4   Desai Sethi Urology Institute, Miller School of Medicine, University of Miami, Miami, Florida
6   Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
,
Emmanuel Thomas
1   Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
2   Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida
5   Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, Florida
6   Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
› Author Affiliations
Funding This work was supported by NIH-NIGMS grant R35GM124915.


Abstract

Viral hepatitis is a leading cause of liver morbidity and mortality globally. The mechanisms underlying acute infection and clearance, versus the development of chronic infection, are poorly understood. In vitro models of viral hepatitis circumvent the high costs and ethical considerations of animal models, which also translate poorly to studying the human-specific hepatitis viruses. However, significant challenges are associated with modeling long-term infection in vitro. Differentiated hepatocytes are best able to sustain chronic viral hepatitis infection, but standard two-dimensional models are limited because they fail to mimic the architecture and cellular microenvironment of the liver, and cannot maintain a differentiated hepatocyte phenotype over extended periods. Alternatively, physiomimetic models facilitate important interactions between hepatocytes and their microenvironment by incorporating liver-specific environmental factors such as three-dimensional ECM interactions and co-culture with non-parenchymal cells. These physiologically relevant interactions help maintain a functional hepatocyte phenotype that is critical for sustaining viral hepatitis infection. In this review, we provide an overview of distinct, novel, and innovative in vitro liver models and discuss their functionality and relevance in modeling viral hepatitis. These platforms may provide novel insight into mechanisms that regulate viral clearance versus progression to chronic infections that can drive subsequent liver disease.

Authors' Contribution

Paper concept, design, and drafting of the manuscript were performed by D.M., D.B., M.H., T.B., E.T., and A.A.




Publication History

Accepted Manuscript online:
19 November 2022

Article published online:
10 January 2023

© 2023. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Thieme Medical Publishers, Inc.
333 Seventh Avenue, 18th Floor, New York, NY 10001, USA

 
  • References

  • 1 Rouse BT, Sehrawat S. Immunity and immunopathology to viruses: What decides the outcome?. Nat Rev Immunol 2010; 10 (07) 514-526
  • 2 Do A, Reau NS. Chronic viral hepatitis: current management and future directions. Hepatol Commun 2020; 4 (03) 329-341
  • 3 Ringelhan M, McKeating JA, Protzer U. Viral hepatitis and liver cancer. Philos Trans R Soc Lond B Biol Sci 2017;372(1732):20160274 :20160274
  • 4 Kileng H, Bernfort L, Gutteberg T. et al. Future complications of chronic hepatitis C in a low-risk area: projections from the hepatitis c study in Northern Norway. BMC Infect Dis 2017; 17 (01) 624
  • 5 Chen Q, Ayer T, Adee MG, Wang X, Kanwal F, Chhatwal J. Assessment of incidence of and surveillance burden for hepatocellular carcinoma among patients with hepatitis C in the era of direct-acting antiviral agents. JAMA Netw Open 2020; 3 (11) e2021173
  • 6 Rawla P, Sunkara T, Muralidharan P, Raj JP. Update in global trends and aetiology of hepatocellular carcinoma. Contemp Oncol (Pozn) 2018; 22 (03) 141-150
  • 7 Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A, Roberts LR. A global view of hepatocellular carcinoma: trends, risk, prevention and management. Nat Rev Gastroenterol Hepatol 2019; 16 (10) 589-604
  • 8 Plummer M, de Martel C, Vignat J, Ferlay J, Bray F, Franceschi S. Global burden of cancers attributable to infections in 2012: a synthetic analysis. Lancet Glob Health 2016; 4 (09) e609-e616
  • 9 Low LA, Mummery C, Berridge BR, Austin CP, Tagle DA. Organs-on-chips: into the next decade. Nat Rev Drug Discov 2021; 20 (05) 345-361
  • 10 Mestas J, Hughes CCW. Of mice and not men: differences between mouse and human immunology. J Immunol 2004; 172 (05) 2731-2738
  • 11 Ruiz SI, Zumbrun EE, Nalca A. Animal models of human viral diseases. Anim Models Study Hum Dis 2017 853–901
  • 12 Dorner M, Horwitz JA, Robbins JB. et al. A genetically humanized mouse model for hepatitis C virus infection. Nature 2011; 474 (7350): 208-211
  • 13 Thomas E, Liang TJ. Experimental models of hepatitis B and C - new insights and progress. Nat Rev Gastroenterol Hepatol 2016; 13 (06) 362-374
  • 14 Louz D, Bergmans HE, Loos BP, Hoeben RC. Animal models in virus research: their utility and limitations. Crit Rev Microbiol 2013; 39 (04) 325-361
  • 15 Moradi E, Jalili-Firoozinezhad S, Solati-Hashjin M. Microfluidic organ-on-a-chip models of human liver tissue. Acta Biomater 2020; 116: 67-83
  • 16 Rowe C, Gerrard DT, Jenkins R. et al. Proteome-wide analyses of human hepatocytes during differentiation and dedifferentiation. Hepatology 2013; 58 (02) 799-809
  • 17 Powers MJ, Domansky K, Kaazempur-Mofrad MR. et al. A microfabricated array bioreactor for perfused 3D liver culture. Biotechnol Bioeng 2002; 78 (03) 257-269
  • 18 Torresi J, Tran BM, Christiansen D, Earnest-Silveira L, Schwab RHM, Vincan E. HBV-related hepatocarcinogenesis: the role of signalling pathways and innovative ex vivo research models. BMC Cancer 2019; 19 (01) 707
  • 19 Allweiss L, Dandri M. Experimental in vitro and in vivo models for the study of human hepatitis B virus infection. J Hepatol 2016; 64 (1, Suppl): S17-S31
  • 20 Chen SL, Morgan TR. The natural history of hepatitis C virus (HCV) infection. Int J Med Sci 2006; 3 (02) 47-52
  • 21 de Oliveria Andrade LJ, D'Oliveira A, Melo RC, De Souza EC, Costa Silva CA, Paraná R. Association between hepatitis C and hepatocellular carcinoma. J Glob Infect Dis 2009; 1 (01) 33-37
  • 22 Axley P, Ahmed Z, Ravi S, Singal AK. Hepatitis C virus and hepatocellular carcinoma: a narrative review. J Clin Transl Hepatol 2018; 6 (01) 79-84
  • 23 El-Serag HB. Hepatocellular carcinoma and hepatitis C in the United States. Hepatology 2002; 36 (5, Suppl 1): S74-S83
  • 24 Ioannou GN, Beste LA, Green PK. et al. Increased risk for hepatocellular carcinoma persists up to 10 years after HCV eradication in patients with baseline cirrhosis or high FIB-4 scores. Gastroenterology 2019; 157 (05) 1264-1278.e4
  • 25 Nahon P, Ganne-Carrié N. Management of patients with pre-therapeutic advanced liver fibrosis following HCV eradication. JHEP Rep 2019; 1 (06) 480-489
  • 26 Shi D, Mi G, Wang M, Webster TJ. In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2019; 198: 228-249
  • 27 Dash S, Aydin Y, Widmer KE, Nayak L. Hepatocellular carcinoma mechanisms associated with chronic HCV infection and the impact of direct-acting antiviral treatment. J Hepatocell Carcinoma 2020; 7: 45-76
  • 28 Xu L, Liu J, Lu M, Yang D, Zheng X. Liver injury during highly pathogenic human coronavirus infections. Liver Int 2020; 40 (05) 998-1004
  • 29 Ali N. Relationship between COVID-19 infection and liver injury: a review of recent data. Front Med (Lausanne) 2020; 7: 458
  • 30 Marjot T, Moon AM, Cook JA. et al. Outcomes following SARS-CoV-2 infection in patients with chronic liver disease: an international registry study. J Hepatol 2021; 74 (03) 567-577
  • 31 Wu ZH, Yang DL. A meta-analysis of the impact of COVID-19 on liver dysfunction. Eur J Med Res 2020; 25 (01) 54
  • 32 Ishibashi H, Nakamura M, Komori A, Migita K, Shimoda S. Liver architecture, cell function, and disease. Semin Immunopathol 2009; 31 (03) 399-409
  • 33 Rocha FG. Liver blood flow: physiology, measurement, and clinical relevance [Internet]. In: Jarnagin WR, Blumgart LH. eds. Blumgart's Surgery of the Liver, Pancreas and Biliary Tract. 5th ed.. Philadelphia: W.B. Saunders; 2012: 74-86
  • 34 Lautt WW. Hepatic circulation: physiology and pathophysiology. Colloquium Series on Integrated Systems Physiology: From Molecule to Function to Disease. San Rafael, CA: Morgan & Claypool Life Sciences; 2009: 1-174
  • 35 Tonon F, Giobbe GG, Zambon A. et al. In vitro metabolic zonation through oxygen gradient on a chip. Sci Rep 2019; 9 (01) 13557
  • 36 Zhou Z, Xu M-J, Gao B. Hepatocytes: a key cell type for innate immunity. Cell Mol Immunol 2016; 13 (03) 301-315
  • 37 Deng J, Wei W, Chen Z. et al. Engineered liver-on-a-chip platform to mimic liver functions and its biomedical applications: a review. Micromachines (Basel) 2019; 10 (10) 676
  • 38 Treyer A, Müsch A. Hepatocyte polarity. Compr Physiol 2013; 3 (01) 243-287
  • 39 McCuskey R. Anatomy of the liver [Internet]. In: Boyer TD, Manns MP, Sanyal AJ. eds. Zakim and Boyer's Hepatology. 6th ed.. Saint Louis: W.B. Saunders; 2012
  • 40 Bhatia SN, Balis UJ, Yarmush ML, Toner M. Effect of cell-cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J 1999; 13 (14) 1883-1900
  • 41 Wang Y, Li J, Wang X, Sang M, Ho W. Hepatic stellate cells, liver innate immunity, and hepatitis C virus. J Gastroenterol Hepatol 2013; 28 (Suppl. 01) 112-115
  • 42 Kolios G, Valatas V, Kouroumalis E. Role of Kupffer cells in the pathogenesis of liver disease. World J Gastroenterol 2006; 12 (46) 7413-7420
  • 43 Hyun J, McMahon RS, Lang AL. et al. HIV and HCV augments inflammatory responses through increased TREM-1 expression and signaling in Kupffer and Myeloid cells. PLoS Pathog 2019; 15 (07) e1007883
  • 44 Beckwitt CH, Clark AM, Wheeler S. et al. Liver ‘organ on a chip’. Exp Cell Res 2018; 363 (01) 15-25
  • 45 Zheng Z-Y, Weng S-Y, Yu Y. Signal molecule-mediated hepatic cell communication during liver regeneration. World J Gastroenterol 2009; 15 (46) 5776-5783
  • 46 Giugliano S, Kriss M, Golden-Mason L. et al. Hepatitis C virus infection induces autocrine interferon signaling by human liver endothelial cells and release of exosomes, which inhibits viral replication. Gastroenterology 2015; 148 (02) 392-402.e13
  • 47 Ni Y, Li J-M, Liu M-K. et al. Pathological process of liver sinusoidal endothelial cells in liver diseases. World J Gastroenterol 2017; 23 (43) 7666-7677
  • 48 Baiocchini A, Del Nonno F, Taibi C. et al. Liver sinusoidal endothelial cells (LSECs) modifications in patients with chronic hepatitis C. Sci Rep 2019; 9 (01) 8760
  • 49 Poisson J, Lemoinne S, Boulanger C. et al. Liver sinusoidal endothelial cells: physiology and role in liver diseases. J Hepatol 2017; 66 (01) 212-227
  • 50 Boltjes A, van Montfoort N, Biesta PJ. et al. Kupffer cells interact with hepatitis B surface antigen in vivo and in vitro, leading to proinflammatory cytokine production and natural killer cell function. J Infect Dis 2015; 211 (08) 1268-1278
  • 51 Faure-Dupuy S, Delphin M, Aillot L. et al. Hepatitis B virus-induced modulation of liver macrophage function promotes hepatocyte infection. J Hepatol 2019; 71 (06) 1086-1098
  • 52 Yuan F, Zhang W, Mu D, Gong J. Kupffer cells in immune activation and tolerance toward HBV/HCV infection. Adv Clin Exp Med 2017; 26 (04) 739-745
  • 53 Boltjes A, Movita D, Boonstra A, Woltman AM. The role of Kupffer cells in hepatitis B and hepatitis C virus infections. J Hepatol 2014; 61 (03) 660-671
  • 54 Bai Q, An J, Wu X. et al. HBV promotes the proliferation of hepatic stellate cells via the PDGF-B/PDGFR-β signaling pathway in vitro. Int J Mol Med 2012; 30 (06) 1443-1450
  • 55 Martín-Vílchez S, Sanz-Cameno P, Rodríguez-Muñoz Y. et al. The hepatitis B virus X protein induces paracrine activation of human hepatic stellate cells. Hepatology 2008; 47 (06) 1872-1883
  • 56 Thomas E, Gonzalez VD, Li Q. et al. HCV infection induces a unique hepatic innate immune response associated with robust production of type III interferons. Gastroenterology 2012; 142 (04) 978-988
  • 57 Cheng J-C, Tseng C-P, Liao M-H. et al. Activation of hepatic stellate cells by the ubiquitin C-terminal hydrolase 1 protein secreted from hepatitis C virus-infected hepatocytes. Sci Rep 2017; 7 (01) 4448
  • 58 Kang YBA, Eo J, Mert S, Yarmush ML, Usta OB. Metabolic patterning on a chip: towards in vitro liver zonation of primary rat and human hepatocytes. Sci Rep 2018; 8 (01) 8951
  • 59 Kietzmann T. Metabolic zonation of the liver: the oxygen gradient revisited. Redox Biol 2017; 11: 622-630
  • 60 Moreau M, Rivière B, Vegna S. et al. Hepatitis C viral proteins perturb metabolic liver zonation. J Hepatol 2015; 62 (02) 278-285
  • 61 Chang M-L. Metabolic alterations and hepatitis C: from bench to bedside. World J Gastroenterol 2016; 22 (04) 1461-1476
  • 62 Wahlicht T, Vièyres G, Bruns SA. et al. Controlled functional zonation of hepatocytes in vitro by engineering of Wnt signaling. ACS Synth Biol 2020; 9 (07) 1638-1649
  • 63 Stebbing J, Sánchez Nievas G, Falcone M. et al. JAK inhibition reduces SARS-CoV-2 liver infectivity and modulates inflammatory responses to reduce morbidity and mortality. Sci Adv 2021; 7 (01) eabe4724
  • 64 Chen X, Saccon E, Appelberg KS. et al. Type-I interferon signatures in SARS-CoV-2 infected Huh7 cells. bioRxiv 2021 2021.02.04.429738
  • 65 Chen D-Y, Khan N, Close BJ. et al. SARS-CoV-2 desensitizes host cells to interferon through inhibition of the JAK-STAT pathway. bioRxiv 2020:2020.10.27.358259
  • 66 Li J, Zong L, Sureau C, Barker L, Wands JR, Tong S. Unusual features of sodium taurocholate cotransporting polypeptide as a hepatitis B virus receptor. J Virol 2016; 90 (18) 8302-8313
  • 67 Wu Y, Geng XC, Wang JF, Miao YF, Lu YL, Li B. The HepaRG cell line, a superior in vitro model to L-02, HepG2 and hiHeps cell lines for assessing drug-induced liver injury. Cell Biol Toxicol 2016; 32 (01) 37-59
  • 68 Shlomai A, Schwartz RE, Ramanan V. et al. Modeling host interactions with hepatitis B virus using primary and induced pluripotent stem cell-derived hepatocellular systems. Proc Natl Acad Sci U S A 2014; 111 (33) 12193-12198
  • 69 Wang J, Qu B, Zhang F. et al. Stem cell-derived hepatocyte-like cells as model for viral hepatitis research. Stem Cells Int 2019; 2019: 9605252
  • 70 Simoneau CR, Ott M. Modeling multi-organ infection by SARS-CoV-2 using stem cell technology. Cell Stem Cell 2020; 27 (06) 859-868
  • 71 Xia Y, Carpentier A, Cheng X. et al. Human stem cell-derived hepatocytes as a model for hepatitis B virus infection, spreading and virus-host interactions. J Hepatol 2017; 66 (03) 494-503
  • 72 Schwartz RE, Bram Y, Frankel A. Pluripotent stem cell-derived hepatocyte-like cells: a tool to study infectious disease. Curr Pathobiol Rep 2016; 4 (03) 147-156
  • 73 Cai D, Mills C, Yu W. et al. Identification of disubstituted sulfonamide compounds as specific inhibitors of hepatitis B virus covalently closed circular DNA formation. Antimicrob Agents Chemother 2012; 56 (08) 4277-4288
  • 74 Seeger C, Sohn JA. Targeting hepatitis B virus with CRISPR/Cas9. Mol Ther Nucleic Acids 2014; 3: e216
  • 75 Catanese MT, Dorner M. Advances in experimental systems to study hepatitis C virus in vitro and in vivo. Virology 2015; 479-480: 221-233
  • 76 El-Shamy A, Hotta H. Impact of hepatitis C virus heterogeneity on interferon sensitivity: an overview. World J Gastroenterol 2014; 20 (24) 7555-7569
  • 77 Dustin LB, Bartolini B, Capobianchi MR, Pistello M. Hepatitis C virus: life cycle in cells, infection and host response, and analysis of molecular markers influencing the outcome of infection and response to therapy. Clin Microbiol Infect 2016; 22 (10) 826-832
  • 78 Li Y-P, Ramirez S, Gottwein JM. et al. Robust full-length hepatitis C virus genotype 2a and 2b infectious cultures using mutations identified by a systematic approach applicable to patient strains. Proc Natl Acad Sci U S A 2012; 109 (18) E1101-E1110
  • 79 Takayama K. In vitro and animal models for SARS-CoV-2 research. Trends Pharmacol Sci 2020; 41 (08) 513-517
  • 80 Wanner N, Andrieux G, Badia-I-Mompel P. et al. Molecular consequences of SARS-CoV-2 liver tropism. Nat Metab 2022; 4 (03) 310-319
  • 81 Nie J, Li Q, Wu J. et al. Establishment and validation of a pseudovirus neutralization assay for SARS-CoV-2. Emerg Microbes Infect 2020; 9 (01) 680-686
  • 82 Yang X, Zhu Y, Zhao X. et al. ASGR1 is a candidate receptor for SARS-CoV-2 that promotes infection of liver cells [Internet]. 2022 [cited May 11, 2022];2022.01.15.476426. Accessed on Dec 29, 2022, at: https://www.biorxiv.org/content/10.1101/2022.01.15.476426v1.full.pdf
  • 83 Mesel-Lemoine M, Millet J, Vidalain P-O. et al. A human coronavirus responsible for the common cold massively kills dendritic cells but not monocytes. J Virol 2012; 86 (14) 7577-7587
  • 84 Zhao X, Guo F, Liu F. et al. Interferon induction of IFITM proteins promotes infection by human coronavirus OC43. Proc Natl Acad Sci U S A 2014; 111 (18) 6756-6761
  • 85 Malik YA. Properties of coronavirus and SARS-CoV-2. Malays J Pathol 2020; 42 (01) 3-11
  • 86 Xu R, Hu P, Li Y, Tian A, Li J, Zhu C. Advances in HBV infection and replication systems in vitro. Virol J 2021; 18 (01) 105
  • 87 Lohmann V. Hepatitis C virus cell culture models: an encomium on basic research paving the road to therapy development. Med Microbiol Immunol (Berl) 2019; 208 (01) 3-24
  • 88 Petropolis DB, Faust DM, Tolle M. et al. Human liver infection in a dish: easy-to-build 3D liver models for studying microbial infection. PLoS One 2016; 11 (02) e0148667
  • 89 Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol 2008; 26 (01) 120-126
  • 90 Khetani SR, Kanchagar C, Ukairo O. et al. Use of micropatterned cocultures to detect compounds that cause drug-induced liver injury in humans. Toxicol Sci 2013; 132 (01) 107-117
  • 91 Wang WW, Khetani SR, Krzyzewski S, Duignan DB, Obach RS. Assessment of a micropatterned hepatocyte coculture system to generate major human excretory and circulating drug metabolites. Drug Metab Dispos 2010; 38 (10) 1900-1905
  • 92 Aleo MD, Ukairo O, Moore A, Irrechukwu O, Potter DM, Schneider RP. Liver safety evaluation of endothelin receptor antagonists using HepatoPac®: a single model impact assessment on hepatocellular health, function and bile acid disposition. J Appl Toxicol 2019; 39 (08) 1192-1207
  • 93 Chan TS, Yu H, Moore A, Khetani SR, Tweedie D. Meeting the challenge of predicting hepatic clearance of compounds slowly metabolized by cytochrome P450 using a novel hepatocyte model, HepatoPac. Drug Metab Dispos 2019; 47 (01) 58-66
  • 94 Trask Jr OJ, Moore A, LeCluyse EL. A micropatterned hepatocyte coculture model for assessment of liver toxicity using high-content imaging analysis. Assay Drug Dev Technol 2014; 12 (01) 16-27
  • 95 Ware BR, Berger DR, Khetani SR. Prediction of drug-induced liver injury in micropatterned co-cultures containing iPSC-derived human hepatocytes. Toxicol Sci 2015; 145 (02) 252-262
  • 96 Ploss A, Khetani SR, Jones CT. et al. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc Natl Acad Sci U S A 2010; 107 (07) 3141-3145
  • 97 Winer BY, Huang TS, Pludwinski E. et al. Long-term hepatitis B infection in a scalable hepatic co-culture system. Nat Commun 2017; 8 (01) 125
  • 98 Winer BY, Gaska JM, Lipkowitz G. et al. Analysis of host responses to hepatitis B and delta viral infections in a micro-scalable hepatic co-culture system. Hepatology 2020; 71 (01) 14-30
  • 99 Bachmann A, Moll M, Gottwald E. et al. 3D cultivation techniques for primary human hepatocytes. Microarrays (Basel) 2015; 4 (01) 64-83
  • 100 Crignis ED, Romal S, Carofiglio F. et al. Human liver organoids; a patient-derived primary model for HBV infection and related hepatocellular carcinoma. bioRxiv 2020; 568147
  • 101 Nie Y-Z, Zheng Y-W, Miyakawa K. et al. Recapitulation of hepatitis B virus-host interactions in liver organoids from human induced pluripotent stem cells. EBioMedicine 2018; 35: 114-123
  • 102 Ananthanarayanan A, Nugraha B, Triyatni M, Hart S, Sankuratri S, Yu H. Scalable spheroid model of human hepatocytes for hepatitis C infection and replication. Mol Pharm 2014; 11 (07) 2106-2114
  • 103 Tran NM, Dufresne M, Duverlie G. et al. An appropriate selection of a 3D alginate culture model for hepatic Huh-7 cell line encapsulation intended for viral studies. Tissue Eng Part A 2013; 19 (1-2): 103-113
  • 104 Molina-Jimenez F, Benedicto I, Dao Thi VL. et al. Matrigel-embedded 3D culture of Huh-7 cells as a hepatocyte-like polarized system to study hepatitis C virus cycle. Virology 2012; 425 (01) 31-39
  • 105 Cho N-J, Elazar M, Xiong A. et al. Viral infection of human progenitor and liver-derived cells encapsulated in three-dimensional PEG-based hydrogel. Biomed Mater 2009; 4 (01) 011001
  • 106 Rajalakshmy AR, Malathi J, Madhavan HN, Samuel JKA. Mebiolgel, a thermoreversible polymer as a scaffold for three dimensional culture of Huh7 cell line with improved hepatocyte differentiation marker expression and HCV replication. Indian J Med Microbiol 2015; 33 (04) 554-559
  • 107 Carpentier A, Sheldon J, Vondran FWR, Brown RJ, Pietschmann T. Efficient acute and chronic infection of stem cell-derived hepatocytes by hepatitis C virus. Gut 2020; 69 (09) 1659-1666
  • 108 Yang L, Han Y, Nilsson-Payant BE. et al. A human pluripotent stem cell-based platform to study SARS-CoV-2 tropism and model virus infection in human cells and organoids. Cell Stem Cell 2020; 27 (01) 125-136.e7
  • 109 Lui VC-H, Hui KP-Y, Babu RO. et al. Human liver organoid derived intra-hepatic bile duct cells support SARS-CoV-2 infection and replication and its comparison with SARS-CoV [Internet]. 2021 [cited May 11, 2022]; 2021.02.10.21251458. Accessed on Dec 29, 2022, at: https://www.medrxiv.org/content/10.1101/2021.02.10.21251458v1.full.pdf
  • 110 Zhao B, Ni C, Gao R. et al. Recapitulation of SARS-CoV-2 infection and cholangiocyte damage with human liver ductal organoids. Protein Cell 2020; 11 (10) 771-775
  • 111 Rossi EA, Quintanilha LF, Nonaka CKV, Souza BSF. Advances in hepatic tissue bioengineering with decellularized liver bioscaffold. [Internet] Stem Cells Int 2019; 2019: 2693189
  • 112 Zhang Z, Xu H, Mazza G. et al. Decellularized human liver scaffold-based three-dimensional culture system facilitate hepatitis B virus infection. J Biomed Mater Res A 2019; 107 (08) 1744-1753
  • 113 Darnell M, Ulvestad M, Ellis E, Weidolf L, Andersson TB. In vitro evaluation of major in vivo drug metabolic pathways using primary human hepatocytes and HepaRG cells in suspension and a dynamic three-dimensional bioreactor system. J Pharmacol Exp Ther 2012; 343 (01) 134-144
  • 114 Aoki Y, Aizaki H, Shimoike T. et al. A human liver cell line exhibits efficient translation of HCV RNAs produced by a recombinant adenovirus expressing T7 RNA polymerase. Virology 1998; 250 (01) 140-150
  • 115 Kawada M, Nagamori S, Aizaki H. et al. Massive culture of human liver cancer cells in a newly developed radial flow bioreactor system: ultrafine structure of functionally enhanced hepatocarcinoma cell lines. In Vitro Cell Dev Biol Anim 1998; 34 (02) 109-115
  • 116 Aizaki H, Nagamori S, Matsuda M. et al. Production and release of infectious hepatitis C virus from human liver cell cultures in the three-dimensional radial-flow bioreactor. Virology 2003; 314 (01) 16-25
  • 117 Pihl AF, Offersgaard AF, Mathiesen CK. et al. High density Huh7.5 cell hollow fiber bioreactor culture for high-yield production of hepatitis C virus and studies of antivirals. Sci Rep 2018; 8 (01) 17505
  • 118 Yoffe B, Darlington GJ, Soriano HE. et al. Cultures of human liver cells in simulated microgravity environment. Adv Space Res 1999; 24 (06) 829-836
  • 119 Chang TT, Hughes-Fulford M. Monolayer and spheroid culture of human liver hepatocellular carcinoma cell line cells demonstrate distinct global gene expression patterns and functional phenotypes. Tissue Eng Part A 2009; 15 (03) 559-567
  • 120 Leite SB, Wilk-Zasadna I, Zaldivar JM. et al. Three-dimensional HepaRG model as an attractive tool for toxicity testing. Toxicol Sci 2012; 130 (01) 106-116
  • 121 Sainz Jr B, TenCate V, Uprichard SL. Three-dimensional Huh7 cell culture system for the study of hepatitis C virus infection. Virol J 2009; 6: 103
  • 122 Kalyanaraman B, Supp DM, Boyce ST. Medium flow rate regulates viability and barrier function of engineered skin substitutes in perfusion culture. Tissue Eng Part A 2008; 14 (05) 583-593
  • 123 Huh D, Hamilton GA, Ingber DE. From 3D cell culture to organs-on-chips. Trends Cell Biol 2011; 21 (12) 745-754
  • 124 Ortega-Prieto AM, Skelton JK, Cherry C, Briones-Orta MA, Hateley CA, Dorner M. “Liver-on-a-chip” cultures of primary hepatocytes and Kupffer cells for hepatitis B virus infection. J Vis Exp 2019; (144) 58333
  • 125 Ortega-Prieto AM, Skelton JK, Wai SN. et al. 3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection. Nat Commun 2018; 9 (01) 682
  • 126 Kang Y, Rawat S, Duchemin N, Bouchard M, Noh M. Human liver sinusoid on a chip for hepatitis B virus replication study. Micromachines (Basel) 2017; 8: 27
  • 127 Natarajan V, Simoneau CR, Erickson AL. et al. Modelling T-cell immunity against hepatitis C virus with liver organoids in a microfluidic coculture system. Open Biol 2022; 12 (03) 210320
  • 128 Broutier L, Mastrogiovanni G, Verstegen MMA. et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat Med 2017; 23 (12) 1424-1435
  • 129 Ouchi R, Togo S, Kimura M. et al. Modeling steatohepatitis in humans with pluripotent stem cell-derived organoids. Cell Metab 2019; 30 (02) 374-384.e6
  • 130 Li X, George SM, Vernetti L, Gough AH, Taylor DLA. A glass-based, continuously zonated and vascularized human liver acinus microphysiological system (vLAMPS) designed for experimental modeling of diseases and ADME/TOX. Lab Chip 2018; 18 (17) 2614-2631
  • 131 Kostrzewski T, Cornforth T, Snow SA. et al. Three-dimensional perfused human in vitro model of non-alcoholic fatty liver disease. World J Gastroenterol 2017; 23 (02) 204-215
  • 132 Kostrzewski T, Maraver P, Ouro-Gnao L. et al. A microphysiological system for studying nonalcoholic steatohepatitis. Hepatol Commun 2019; 4 (01) 77-91
  • 133 Gori M, Simonelli MC, Giannitelli SM, Businaro L, Trombetta M, Rainer A. Investigating nonalcoholic fatty liver disease in a liver-on-a-chip microfluidic device. PLoS One 2016; 11 (07) e0159729
  • 134 Feaver RE, Cole BK, Lawson MJ. et al. Development of an in vitro human liver system for interrogating nonalcoholic steatohepatitis. JCI Insight 2016; 1 (20) e90954
  • 135 Mukherjee S, Zhelnin L, Sanfiz A. et al. Development and validation of an in vitro 3D model of NASH with severe fibrotic phenotype. Am J Transl Res 2019; 11 (03) 1531-1540
  • 136 Pingitore P, Sasidharan K, Ekstrand M, Prill S, Lindén D, Romeo S. Human multilineage 3D spheroids as a model of liver steatosis and fibrosis. Int J Mol Sci 2019; 20 (07) 1629
  • 137 Norona LM, Nguyen DG, Gerber DA, Presnell SC, LeCluyse EL. Editor's highlight: modeling compound-induced fibrogenesis in vitro using three-dimensional bioprinted human liver tissues. Toxicol Sci 2016; 154 (02) 354-367
  • 138 Norona LM, Nguyen DG, Gerber DA, Presnell SC, Mosedale M, Watkins PB. Bioprinted liver provides early insight into the role of Kupffer cells in TGF-β1 and methotrexate-induced fibrogenesis. PLoS One 2019; 14 (01) e0208958
  • 139 Vernetti LA, Senutovitch N, Boltz R. et al. A human liver microphysiology platform for investigating physiology, drug safety, and disease models. Exp Biol Med (Maywood) 2016; 241 (01) 101-114
  • 140 Bell CC, Hendriks DFG, Moro SML. et al. Characterization of primary human hepatocyte spheroids as a model system for drug-induced liver injury, liver function and disease. Sci Rep 2016; 6: 25187
  • 141 Lachowski D, Cortes E, Rice A, Pinato D, Rombouts K, Del Rio Hernandez A. Matrix stiffness modulates the activity of MMP-9 and TIMP-1 in hepatic stellate cells to perpetuate fibrosis. Sci Rep 2019; 9 (01) 7299
  • 142 Wells RG. The role of matrix stiffness in hepatic stellate cell activation and liver fibrosis. J Clin Gastroenterol 2005; 39 (4, Suppl 2): S158-S161
  • 143 Clark AM, Wheeler SE, Young CL. et al. A liver microphysiological system of tumor cell dormancy and inflammatory responsiveness is affected by scaffold properties. Lab Chip 2016; 17 (01) 156-168
  • 144 Methotrexate (Rheumatrex, Trexall) [Internet]. [cited March 10, 2021]. Accessed November 25, 2022 at: https://www.rheumatology.org/I-Am-A/Patient-Caregiver/Treatments/Methotrexate-Rheumatrex-Trexall
  • 145 Leite SB, Roosens T, El Taghdouini A. et al. Novel human hepatic organoid model enables testing of drug-induced liver fibrosis in vitro. Biomaterials 2016; 78: 1-10
  • 146 Deng J, Chen Z, Zhang X. et al. A liver-chip-based alcoholic liver disease model featuring multi-non-parenchymal cells. Biomed Microdevices 2019; 21 (03) 57
  • 147 March S, Ng S, Velmurugan S. et al. A microscale human liver platform that supports the hepatic stages of Plasmodium falciparum and vivax. Cell Host Microbe 2013; 14 (01) 104-115
  • 148 Ng S, March S, Galstian A. et al. Hypoxia promotes liver-stage malaria infection in primary human hepatocytes in vitro. Dis Model Mech 2014; 7 (02) 215-224
  • 149 Lee SY, Sung JH. Gut-liver on a chip toward an in vitro model of hepatic steatosis. Biotechnol Bioeng 2018; 115 (11) 2817-2827
  • 150 Prot JM, Maciel L, Bricks T. et al. First pass intestinal and liver metabolism of paracetamol in a microfluidic platform coupled with a mathematical modeling as a means of evaluating ADME processes in humans. Biotechnol Bioeng 2014; 111 (10) 2027-2040
  • 151 Esch MB, Ueno H, Applegate DR, Shuler ML. Modular, pumpless body-on-a-chip platform for the co-culture of GI tract epithelium and 3D primary liver tissue. Lab Chip 2016; 16 (14) 2719-2729
  • 152 Bricks T, Paullier P, Legendre A. et al. Development of a new microfluidic platform integrating co-cultures of intestinal and liver cell lines. Toxicol In Vitro 2014; 28 (05) 885-895
  • 153 Wagner I, Materne E-M, Brincker S. et al. A dynamic multi-organ-chip for long-term cultivation and substance testing proven by 3D human liver and skin tissue co-culture. Lab Chip 2013; 13 (18) 3538-3547
  • 154 Bovard D, Sandoz A, Luettich K. et al. A lung/liver-on-a-chip platform for acute and chronic toxicity studies. Lab Chip 2018; 18 (24) 3814-3829
  • 155 Vunjak-Novakovic G, Bhatia S, Chen C, Hirschi K. HeLiVa platform: integrated heart-liver-vascular systems for drug testing in human health and disease. Stem Cell Res Ther 2013; 4 (Suppl. 01) S8
  • 156 Theobald J, Ghanem A, Wallisch P. et al. Liver-kidney-on-chip to study toxicity of drug metabolites. ACS Biomater Sci Eng 2018; 4 (01) 78-89
  • 157 Maschmeyer I, Lorenz AK, Schimek K. et al. A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents. Lab Chip 2015; 15 (12) 2688-2699
  • 158 Ronaldson-Bouchard K, Teles D, Yeager K. et al. A multi-organ chip with matured tissue niches linked by vascular flow. Nat Biomed Eng 2022; 6 (04) 351-371
  • 159 Kim J-Y, Fluri DA, Kelm JM, Hierlemann A, Frey O. 96-well format-based microfluidic platform for parallel interconnection of multiple multicellular spheroids. J Lab Autom 2015; 20 (03) 274-282
  • 160 Phan DTT, Wang X, Craver BM. et al. A vascularized and perfused organ-on-a-chip platform for large-scale drug screening applications. Lab Chip 2017; 17 (03) 511-520
  • 161 Miller PG, Shuler ML. Design and demonstration of a pumpless 14 compartment microphysiological system. Biotechnol Bioeng 2016; 113 (10) 2213-2227
  • 162 Ren K, Zhou J, Wu H. Materials for microfluidic chip fabrication. Acc Chem Res 2013; 46 (11) 2396-2406
  • 163 Qin D, Xia Y, Whitesides GM. Soft lithography for micro- and nanoscale patterning. Nat Protoc 2010; 5 (03) 491-502
  • 164 Gökaltun A, Kang YBA, Yarmush ML, Usta OB, Asatekin A. Simple surface modification of poly(dimethylsiloxane) via surface segregating smart polymers for biomicrofluidics. Sci Rep 2019; 9 (01) 7377
  • 165 Matellan C, Del Río Hernández AE. Cost-effective rapid prototyping and assembly of poly(methyl methacrylate) microfluidic devices. Sci Rep 2018; 8 (01) 6971
  • 166 Lee PJ, Hung PJ, Lee LP. An artificial liver sinusoid with a microfluidic endothelial-like barrier for primary hepatocyte culture. Biotechnol Bioeng 2007; 97 (05) 1340-1346
  • 167 Deng J, Zhang X, Chen Z. et al. A cell lines derived microfluidic liver model for investigation of hepatotoxicity induced by drug-drug interaction. Biomicrofluidics 2019; 13 (02) 024101
  • 168 Bavli D, Prill S, Ezra E. et al. Real-time monitoring of metabolic function in liver-on-chip microdevices tracks the dynamics of mitochondrial dysfunction. Proc Natl Acad Sci U S A 2016; 113 (16) E2231-E2240
  • 169 Yajima Y, Lee CN, Yamada M, Utoh R, Seki M. Development of a perfusable 3D liver cell cultivation system via bundling-up assembly of cell-laden microfibers. J Biosci Bioeng 2018; 126 (01) 111-118
  • 170 Mi S, Yi X, Du Z, Xu Y, Sun W. Construction of a liver sinusoid based on the laminar flow on chip and self-assembly of endothelial cells. Biofabrication 2018; 10 (02) 025010
  • 171 Bhise NS, Manoharan V, Massa S. et al. A liver-on-a-chip platform with bioprinted hepatic spheroids. Biofabrication 2016; 8 (01) 014101
  • 172 Esch MB, Prot J-M, Wang YI. et al. Multi-cellular 3D human primary liver cell culture elevates metabolic activity under fluidic flow. Lab Chip 2015; 15 (10) 2269-2277
  • 173 Tan K, Keegan P, Rogers M. et al. A high-throughput microfluidic microphysiological system (PREDICT-96) to recapitulate hepatocyte function in dynamic, re-circulating flow conditions. Lab Chip 2019; 19 (09) 1556-1566
  • 174 Bircsak KM, DeBiasio R, Miedel M. et al. A 3D microfluidic liver model for high throughput compound toxicity screening in the OrganoPlate®. Toxicology 2021; 450: 152667
  • 175 Long TJ, Cosgrove PA, Dunn II RT. et al. Modeling therapeutic antibody-small molecule drug-drug interactions using a three-dimensional perfusable human liver coculture platform. Drug Metab Dispos 2016; 44 (12) 1940-1948
  • 176 Shoemaker JT, Zhang W, Atlas SI, Bryan RA, Inman SW, Vukasinovic J. A 3D cell culture organ-on-a-chip platform with a breathable hemoglobin analogue augments and extends primary human hepatocyte functions in vitro . Front Mol Biosci 2020; 7: 568777
  • 177 Jang K-J, Otieno MA, Ronxhi J. et al. Reproducing human and cross-species drug toxicities using a liver-chip. Sci Transl Med 2019; 11 (517) eaax5516
  • 178 Rowe C, Shaeri M, Large E. et al. Perfused human hepatocyte microtissues identify reactive metabolite-forming and mitochondria-perturbing hepatotoxins. Toxicol In Vitro 2018; 46: 29-38
  • 179 Sarkar U, Rivera-Burgos D, Large EM. et al. Metabolite profiling and pharmacokinetic evaluation of hydrocortisone in a perfused three-dimensional human liver bioreactor. Drug Metab Dispos 2015; 43 (07) 1091-1099
  • 180 Sarkar U, Ravindra KC, Large E. et al. Integrated assessment of diclofenac biotransformation, pharmacokinetics, and omics-based toxicity in a three-dimensional human liver-immunocompetent coculture system. Drug Metab Dispos 2017; 45 (07) 855-866
  • 181 Lee J-H, Ho K-L, Fan S-K. Liver microsystems in vitro for drug response. J Biomed Sci 2019; 26 (01) 88
  • 182 Novik E, Maguire TJ, Chao P, Cheng KC, Yarmush ML. A microfluidic hepatic coculture platform for cell-based drug metabolism studies. Biochem Pharmacol 2010; 79 (07) 1036-1044
  • 183 Jang M, Kleber A, Ruckelshausen T, Betzholz R, Manz A. Differentiation of the human liver progenitor cell line (HepaRG) on a microfluidic-based biochip. J Tissue Eng Regen Med 2019; 13 (03) 482-494
  • 184 Michailidis E, Pabon J, Xiang K. et al. A robust cell culture system supporting the complete life cycle of hepatitis B virus. Sci Rep 2017; 7 (01) 16616
  • 185 Dubois-Pot-Schneider H, Aninat C, Kattler K. et al. Transcriptional and epigenetic consequences of DMSO treatment on HepaRG cells. Cells 2022; 11 (15) 2298