Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2446 word(s) 2446 2021-07-14 08:03:18 |
2 format correct Meta information modification 2446 2021-07-27 04:22:48 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Vosough, M. Modeling Hepatotropic Viral Infections. Encyclopedia. Available online: https://encyclopedia.pub/entry/12462 (accessed on 29 March 2024).
Vosough M. Modeling Hepatotropic Viral Infections. Encyclopedia. Available at: https://encyclopedia.pub/entry/12462. Accessed March 29, 2024.
Vosough, Massoud. "Modeling Hepatotropic Viral Infections" Encyclopedia, https://encyclopedia.pub/entry/12462 (accessed March 29, 2024).
Vosough, M. (2021, July 26). Modeling Hepatotropic Viral Infections. In Encyclopedia. https://encyclopedia.pub/entry/12462
Vosough, Massoud. "Modeling Hepatotropic Viral Infections." Encyclopedia. Web. 26 July, 2021.
Modeling Hepatotropic Viral Infections
Edit

The hepatotropic viruses A, B, C, D, and E, are the most common causes of viral infections that can lead to liver failure. Hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatitis D virus (HDV) are normally transmitted through organ transplants, transfusions, sex, and injections. The HBV genome is a double-strand DNA, and this virus is classified into the genus of Orthohepadnavirus and the family of Hepadnaviridae. 

animal models hepatotropic virus cell culture viral hepatitis modeling hepatotropic viruses

1. Overview

The lack of an appropriate platform for a better understanding of the molecular basis of hepatitis viruses and the absence of reliable models to identify novel therapeutic agents for a targeted treatment are the two major obstacles for launching efficient clinical protocols in different types of viral hepatitis. Viruses are obligate intracellular parasites, and the development of model systems for efficient viral replication is necessary for basic and applied studies. Viral hepatitis is a major health issue and a leading cause of morbidity and mortality. Despite the extensive efforts that have been made on fundamental and translational research, traditional models are not effective in representing this viral infection in a laboratory. In this review, we discuss in vitro cell-based models and in vivo animal models, with their strengths and weaknesses. In addition, the most important findings that have been retrieved from each model are described.

2. Hepatitis Viruses

The lack of an appropriate platform for a better understanding of the molecular basis and the absence of reliable models to identify novel therapeutic agents for a targeted treatment are the two major obstacles for launching efficient clinical protocols in different types of viral hepatitis [1][2]. The hepatotropic viruses A, B, C, D, and E, are the most common causes of viral infections that can lead to liver failure. Hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatitis D virus (HDV) are normally transmitted through organ transplants, transfusions, sex, and injections [3]. The HBV genome is a double-strand DNA, and this virus is classified into the genus of Orthohepadnavirus and the family of Hepadnaviridae. HCV is a small virus (55–65 nm in size) with an RNA genome and is categorized into the genus of Hepacivirus and the family of Flaviviridae [4]. HDV is a small and uncommon human pathogen containing a single-stranded circular RNA genome. This pathogen is classified into the genus of Deltaviridae. HDV has hepatitis B surface antigen (HBsAg), which surrounds the genomic RNA-nucleoprotein complex, thereafter, requires HBV to complete its life cycle and replicate [5][6] HBV, HCV, and HDV induce chronic liver inflammation that can finally result in cirrhosis and hepatocellular carcinoma [5][7]. HAV (Hepatitis A virus) and HEV (Hepatitis E virus) are generally transmitted through water or food [8]. The studies conducted in many countries have demonstrated that HAV is the most common type causing acute viral hepatitis. HAV is a single strand of plus sense RNA and belongs to the genus of Hepatovirus in the Picornaviridae family, and it is transmitted through the fecal-oral route [9]. HEV is classified into the genus of Orthohepenvirus A and the family of Hepeviridae. HEV is a nonenveloped positive-strand RNA virus that generally causes acute infections. Clinical observations indicate that HEV can lead to chronic infections in immunocompromised patients, including transplant recipients [8][10]. On the other hand, it is difficult to develop therapeutics for these viral hepatitis, due to the lack of a reliable model. Therefore, establishing a defined and suitable model as a platform for studying hepatotropic viruses is necessary [11]. Studying hepatotropic viruses in vivo is limited because most of them are species-specific. For instance, HBV and HCV infect only humans, tree shrews, and some nonhuman primates [2][12]. Thus, the most promising approach to study human hepatotropic viruses is using genetically modified animals. In parallel, cell-based systems have received remarkable attention in hepatotropic viral infection modeling. Monolayer and three-dimensional (3D) culture systems have been used for understanding the molecular basis of hepatotropic viruses and evaluating novel antiviral agents; however, they did not help the scientific community in understanding some aspects of viral pathogenesis, including the differences between the genotypes of hepatotropic viruses. In this review, we discuss different in vitro and in vivo model systems for the study of hepatotropic viruses, and their advantages and disadvantages.

3. A monolayer Culture (Primary Cells, Cell Lines, and Coculture System)

Primary human hepatocytes (PHH), as the most authentic cell culture model for hepatotropic viruses, were used in many studies [13][14][15]. For in vitro HBV studies, PHH has been used as a gold standard platform in cell culture for many years, and many studies used PHH as a model for HBV and HCV infection [16][17]. A later study indicated that PHH also supported an HEV infection with a Kernow-C1/p6 strain [18]. Other studies demonstrated that PHH was susceptible to HDV [19][20][21]. Taylor et al. provided the first evidence that purinergic receptor functionality was essential for the process of PHH infection by HDV and HBV [22]. Since the source of chronic HEV infection is unknown, some studies explored extrahepatic sources for HEV replication. In 2019, a study [23] introduced human polarized enterocytes (primary intestinal cells) as a model for HEV replication. Moreover, El-Mokhtar and his colleagues demonstrated that primary human endometrial stromal cells were susceptible and permissive to HEV infection, and this type of primary cells could be an endogenous source of HEV infection during pregnancy and mediate the HEV vertical transmission [24]. However, their utility is hampered by some shortcomings, including the viability of donated cells, availability, and limitation for a long-term ex vivo culture [25]. These challenges have led to establishing various culture models [13].
To preserve the liver-specific function and extend the lifetime of PHH, some studies employed a coculture strategy that consisted of PHH and supportive stromal cells (murine fibroblast cells) with the ratio of PHH and supportive cells estimated as 1:4. In this micropatterned coculture system, the phenotype and functionality of PHH were maintained for more than several weeks, and the results showed a robust infection of PHH with HCV [26][27]. Inconsistency with the previous studies, March and collaborators established a micropatterned coculture system in which islands of PHH in a 2D culture were surrounded by fibroblast cells. The results demonstrated that this culture system was suitable for the study of HBV and HCV colonization and replication [28]. Furthermore, Zhou and colleagues applied a coculture system that consisted of fetal PHH and liver nonparenchymal cells for prolonged susceptibility to HBV infection [29]. Winer and collaborators, by self-assembling of PHH and mouse stromal cells in a co-culture system, provided a scalable platform for long-term colonization and replication of HBV [30]. Later, this group demonstrated that self-assembly of PHH with nonparenchymal mouse embryonic fibroblast 3T3-J2 cells in a co-culture system is a versatile platform for studying HBV/ HDV coinfections and holds great promise for performing chemical library screens and improving our understanding of the host response to such infections [31]. Furthermore, in 2020, a study [32] reported that peripheral blood mononuclear cells and bone marrow-derived macrophages from healthy donors were susceptible to HEV in vitro. Since renal diseases are associated with HEV infection, in 2020, a study [33] reported a possible mechanism for HEV-mediated renal disease. The authors isolated CD10+/CD13+ primary proximal tubular epithelial cells, infected them in vitro with HEV inoculum, and then the expression of inflammatory and kidney injury markers was assessed in cocultivation with/without immune cells isolated from the same donors. The results demonstrated that coculture of immune cells with HEV-infected epithelial cells exacerbated the inflammatory response and induced kidney injury [33]. However, applying the coculture system for PHH could not overcome the drawbacks of the broad usage of PHH.
Some studies reported establishing two hepatoma cell lines named HepAD38 and HepDE19, which expressed HBV pgRNA under the control of the tetracycline-repressible promoter instead of the native viral core promoter [34][35]. A Chinese group introduced a new hepatoma HLCZ01 cell line which supported the whole life cycle of both HBV and HCV. The results indicated that this cell line provided a powerful tool for supporting the accurate life cycle of the virus with a normal genetic background [36]. Since the HBV infection also occurs at extrahepatic sites, the identification of the relevant host factor in nonhepatic cells is essential. Yang and collaborators reconstituted the HBV infection in the human embryonic kidney (HEK) 293T cells by exogenous expression of the nuclear hormone receptors HNF4α, RXRα, and PPARα, and the HBV receptor, sodium taurocholate cotransporting polypeptide (NTCP). Their results suggested that these factors could play a pivotal role in the HBV infection of nonhepatic cells [37]. A German group established a stable new cell line named HepNB2.7, which was susceptible to HDV and supported the full viral life cycle [38].
Several polarized human liver cell lines were produced, such as HepaRG and HepG2 [39]. The hepatoma HepaRG cell line, a bipotent liver progenitor cell line, upon induction by dimethyl sulfoxide (DMSO), was permissive to HBV [40]. To optimize the in vitro differentiation of HepaRG, Yuan and his colleagues used four small molecules (FPH1, FPH2, FH1, and XMU-MP-1) and increased both the hepatic differentiation and proliferation capacity of HepaRG cells in vitro and in vivo, which is essential for the HBV infection [41]. However, using this cell type has some drawbacks, including a low viral yield and replication, a lack of covalently closed circular DNA (cccDNA) amplification, and difficulty understanding the HBV life cycle [42]. Sophie Roge’e and her colleagues introduced the HepaRG cell line and PICM19, derived from the primary culture of pig embryonic stem cells as in vitro models for HEV replication. They reported that these in vitro culture systems support HEV replication and release of encapsulated RNA [43]. In accordance with this study, Pellerin and her colleagues showed that HepaRG is a relevant and efficient in vitro model of HEV replication that could be used to study HEV and identify effective antiviral drugs against chronic HEV infection [44]. Besides, another study introduced a new procedure using a cocktail of 5 chemicals (Forskolin, SB431542, IWP2, DAPT, and LDN193189), allowing fast differentiation and efficient HDV-infection of HepaRG cells [45]. Many studies applied the HepaRG cell line as a unique model to study the interplay between HBV/HDV and hepatocyte-specific innate immunity, as well as to explore new therapeutic developments [46][47].
HepG2 [(Hepatoma G2); derived from a hepatoblastoma] and Huh7 [(human hepatocellular carcinoma cell line 7); derived from a hepatocellular carcinoma] are two human hepatoma cell lines which are widely used in antiviral studies, especially those regarding HBV. These cell lines support the virus replication when transfected with HBV [48][49]. Some studies indicated that HepG2 and Huh-7 cell lines as NTCP-expressing lines could be efficiently infected with HBV and HDV [47][50]. Another study showed altered gene expression in HepG2 cells induced by HBV and HCV, which provides new insight into the mechanism of HBV and HCV infection and improves the understanding of the differences in the molecular pathogenesis of HBV and HCV [51]. Not long ago, it has been found that HepG2-NTCP cells are hardly infected with HBV-positive sera and that a clonal section is needed to recognize clones producing high titers of infectious progeny [52]. Recently, Kempp et al. established a stable cell line (HepNB2.7) by transducing HepG2 cells with genes encoding the NTCP-receptor and the HBV envelope proteins that support the full viral life cycle of HDV and HBV [38]. Furthermore, to support HEV replication, in a study [53] HepG2 and Huh-7 lines were used. A study [54] reported a simple yet robust cell culture HEV infection method. The model was based on the HEV genotype 3 Kernow-C1 p6 strain and the two human hepatoma cell lines (HepG2 and HepG2/C3A) combined with various media conditions.
To investigate various viruses’ interactions, Jian and collaborators developed a scalable and visualizable HAV/HCV coinfection model in Huh-7 cells. Their finding revealed that the simultaneous presence of HAV-HCV did not affect the viral RNA synthesis of both viruses. They suggested that indirect interactions may lead to the suppression and clearance of HCV in HAV/HCV coinfected patients [55]. Sun and colleagues reported the creation of stable TetOFF hepatoma cell lines (HepG2 and Huh7) to control HBV production. Their approach presented some advantages, including applying both hepatoma cell lines and using a two-step procedure rather than cotransfection [56]. König and collaborators tried to develop a perfect cell culture platform for HBV amplification from clinical specimens. To achieve this, they applied slow proliferating HepG2- NTCP for the HBV infection. The obtained results demonstrated that this cell line successfully supported the whole HBV life cycle, as well as long-term amplification of HBV [52]. However, these cell lines could do mediate the early-stage virus infection, such as the entry, uncoating, and the formation of cccDNA [25]. Besides, the causes of limited HCV permissiveness in cell lines are not completely understood, but the most important aspects have been identified. Restricted expression of cell surface receptors, such as CD81 and scavenger receptor class B type I (SRBI), is recognized to be associated with the restriction of the HCV entrance [25][57][58].
Therefore, to better mimic the viral life cycle and host-virus interactions, more representative and functional cell types are urgently needed. Pluripotent stem cells (PSCs) are a renewable source of cells and can be obtained via different protocols [59]. Since the cells derived from PSCs would be functional and similar to primary cells, they may be suggested as an ideal replacement for currently used cell-based models [60]. In 2012 and 2015, 2D cultures of HLCs derived from human PSCs were shown to support the entry and replication of HCVcc [61][62]. In 2017, these findings were corroborated by Yan et al. [63]. Various host factors are critical to HBV infection. Hepatic-like cells (HLCs) mostly resemble PHH, due to the high expression of crucial factors for HBV infection and replication. As a first confirmation, Shlomi and colleagues indicated that PSCs-derived HLCs were permissive to the HBV infection [27]. Consistent with the previous study, two other studies demonstrated that stem cell-derived HLCs could completely support the HBV infection for about one month [25][63][64]. Moreover, several studies reported that PSCs-derived HLCs could successfully support various forms of HCV infection [65][66]. Some studies have successfully demonstrated that the complete replication cycle of HEV is supported by iPSC-derived HLCs [67][68][69]. In cell-based models of liver disease, researchers understand the importance of hepatocyte polarity. Because these viruses enter hepatocytes through the basolateral membrane, the hepatocyte polarity is crucial for the productive entry of hepatitis viruses [70]. In one study [70], researchers differentiated hPSCs into columnar polarized HLCs using transwell filters. These HLCs secreted urea, albumin, and lipoproteins basolaterally, while bile acids were produced apically. The authors showed that polarized HLC supported HEV infection and replication, and mimicked fundamental steps of the natural infectious cycle in vivo.
Nevertheless, although great efforts are directed toward improving differentiation protocols to achieve better maturation, little progress has been achieved so far. Therefore, there is a need to provide a suitable niche that is more similar to the in vivo architecture, such as spheroids, organoids, bioprinted microtissues, and microfluidic chip devices [60].

References

  1. Burwitz, B.J.; Zhou, Z.; Li, W. Animal models for the study of human hepatitis B and D virus infection: New insights and progress. Antivir. Res. 2020, 182, 104898.
  2. Ortega-Prieto, A.M.; Cherry, C.; Gunn, H.; Dorner, M. In Vivo Model Systems for Hepatitis B Virus Research. Acs Infect. Dis. 2019, 5, 688–702.
  3. Inoue, T.; Tanaka, Y. Hepatitis B virus and its sexually transmitted infection—An update. Microb. Cell 2016, 3, 419–436.
  4. Kushner, T.; Sperling, R.S.; Dieterich, D. Family Counseling for Hepatitis B and Hepatitis C. Clin. Liver Dis. A Multimed. Rev. J. 2019, 13, 93–97.
  5. Farci, P.; Niro, G.A.; Zamboni, F.; Diaz, G. Hepatitis D and hepatocellular carcinoma. Viruses 2021, 13, 538.
  6. Usman, Z.; Velkov, S.; Protzer, U.; Roggendorf, M.; Frishman, D.; Karimzadeh, H. HDVdb: A comprehensive hepatitis d virus database. Viruses 2020, 12, 538.
  7. Ringehan, M.; Mckeating, J.A.; Protzer, U. Viral hepatitis and liver cancer. Philos. Trans. R. Soc. L. B Biol. Sci. 2017, 372, 20160274.
  8. Wang, J.; Qu, B.; Zhang, F.; Zhang, C.; Deng, W.; Thi, V.L.D.; Xia, Y. Stem cell-derived hepatocyte-like cells as model for viral hepatitis research. Stem Cells Int. 2019, 2019.
  9. Yu, J.M.; Li, L.L.; Zhang, C.Y.; Lu, S.; Ao, Y.Y.; Gao, H.C.; Xie, Z.; Xie, G.C.; Sun, X.M.; Pang, L.L.; et al. A novel hepatovirus identified in wild woodchuck Marmota himalayana. Sci. Rep. 2016, 6, 22361.
  10. Debing, Y.; Moradpour, D.; Neyts, J.; Gouttenoire, J. Update on hepatitis e virology: Implications for clinical practice. J. Hepatol. 2016, 65, 200–212.
  11. Lamontagne, J.; Mell, J.C.; Bouchard, M.J. Transcriptome-Wide Analysis of Hepatitis B Virus-Mediated Changes to Normal Hepatocyte Gene Expression. PLoS Pathog. 2016, 12, e1005438.
  12. Catanese, M.T.; Dorner, M. Advances in experimental systems to study hepatitis C virus in vitro and in vivo. Virology 2015, 479–480, 221–233.
  13. Ortega-Prieto, A.M.; Skelton, J.K.; Wai, S.N.; Large, E.; Lussignol, M.; Vizcay-Barrena, G.; Hughes, D.; Fleck, R.A.; Thursz, M.; Catanese, M.T.; et al. 3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection. Nat. Commun. 2018, 9, 1–15.
  14. Xiang, C.; Du, Y.; Meng, G.; Yi, L.S.; Sun, S.; Song, N.; Zhang, X.; Xiao, Y.; Wang, J.; Yi, Z.; et al. Long-term functional maintenance of primary human hepatocytes in vitro. Science 2019, 364, 399–402.
  15. Zahmatkesh, E.; Vosough, M. A Quick update from the Past to Current Status of Human Pluripotent Stem Cell-derived Hepatocyte culture systems. Mod. Med. Lab. J. 2018, 2, 110–112.
  16. Galle, P.R.; Hagelstein, J.; Kommerell, B.; Volkmann, M.; Schranz, P.; Zentgraf, H. In vitro experimental infection of primary human hepatocytes with hepatitis B virus. Gastroenterology 1994, 106, 664–673.
  17. Fournier, C.; Pageaux, G.; Maurel, P.; Coste, J.; Larrey, D.; Ducos, J.; Sureau, C.; Domergue, J. In vitro infection of adult normal human hepatocytes in primary culture by hepatitis C virus. J. Gen. Virol. 1998, 79, 2367–2374.
  18. Yin, X.; Li, X.; Ambardekar, C.; Hu, Z.; Lhomme, S.; Feng, Z. Hepatitis E virus persists in the presence of a type III interferon response. PLoS Pathog. 2017, 13, e1006417.
  19. Gudima, S.; He, Y.; Meier, A.; Chang, J.; Chen, R.; Jarnik, M.; Nicolas, E.; Bruss, V.; Taylor, J. Assembly of Hepatitis Delta Virus: Particle Characterization, Including the Ability To Infect Primary Human Hepatocytes. J. Virol. 2007, 81, 3608–3617.
  20. Gudima, S.; He, Y.; Chai, N.; Bruss, V.; Urban, S.; Mason, W.; Taylor, J. Primary Human Hepatocytes Are Susceptible to Infection by Hepatitis Delta Virus Assembled with Envelope Proteins of Woodchuck Hepatitis Virus. J. Virol. 2008, 82, 7276–7283.
  21. Zhang, Z.; Filzmayer, C.; Ni, Y.; Sültmann, H.; Mutz, P.; Hiet, M.S.; Vondran, F.W.R.; Bartenschlager, R.; Urban, S. Hepatitis D virus replication is sensed by MDA5 and induces IFN-β/λ responses in hepatocytes. J. Hepatol. 2018, 69, 25–35.
  22. Taylor, J.M.; Han, Z. Purinergic receptor functionality is necessary for infection of human hepatocytes by hepatitis delta virus and hepatitis b virus. PLoS ONE 2010, 5, e15784.
  23. Marion, O.; Lhomme, S.; Nayrac, M.; Dubois, M.; Pucelle, M.; Requena, M.; Migueres, M.; Abravanel, F.; Peron, J.M.; Carrere, N.; et al. Hepatitis E virus replication in human intestinal cells. Gut 2020, 69, 901–910.
  24. El-mokhtar, M.A.; Othman, E.R.; Khashbah, M.Y.; Ismael, A.; Ghaliony, M.A.A.; Seddik, M.I.; Sayed, I.M. Evidence of the Extrahepatic Replication of Hepatitis. Pathogens 2020, 9, 295.
  25. Xia, Y.; Carpentier, A.; Cheng, X.; Block, P.D.; Zhao, Y.; Zhang, Z.; Protzer, U.; Liang, T.J. Human stem cell-derived hepatocytes as a model for hepatitis B virus infection, spreading and virus-host interactions. Physiol. Behav. 2018, 176, 139–148.
  26. PLoSs, A.; Khetani, S.R.; Jones, C.T.; Syder, A.J.; Trehan, K.; Gaysinskaya, V.A.; Mu, K.; Ritola, K.; Rice, C.M.; Bhatia, S.N. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc. Natl. Acad. Sci. USA 2010, 107, 3141–3145.
  27. Shlomai, A.; Schwartz, R.E.; Ramanan, V.; Bhatta, A.; De Jong, Y.P.; Bhatia, S.N.; Rice, C.M. Modeling host interactions with hepatitis B virus using primary and induced pluripotent stem cell-derived hepatocellular systems. Proc. Natl. Acad. Sci. USA 2014, 111, 12193–12198.
  28. March, S.; Ramanan, V.; Trehan, K.; Ng, S.; Galstian, A.; Gural, N.; Scull, M.A.; Shlomai, A.; Mota, M.M.; Fleming, H.E.; et al. Micropatterned coculture of primary human hepatocytes and supportive cells for the study of hepatotropic pathogens. Nat. Protoc. 2015, 10, 2027–2053.
  29. Zhou, M.; Zhao, F.; Li, J.; Cheng, Z.; Tian, X.; Zhi, X.; Huang, Y.; Hu, K. Long-term maintenance of human fetal hepatocytes and prolonged susceptibility to HBV infection by co-culture with non-parenchymal cells. J. Virol. Methods 2014, 195, 185–193.
  30. Winer, B.Y.; Huang, T.S.; Pludwinski, E.; Heller, B.; Wojcik, F.; Lipkowitz, G.E.; Parekh, A.; Cho, C.; Shrirao, A.; Muir, T.W.; et al. Long-term hepatitis B infection in a scalable hepatic co-culture system. Nat. Commun. 2017, 8, 1–11.
  31. Winer, B.Y.; Gaska, J.M.; Lipkowitz, G.; Bram, Y.; Parekh, A.; Parsons, L.; Leach, R.; Jindal, R.; Cho, C.H.; Shrirao, A.; et al. Analysis of Host Responses to Hepatitis B and Delta Viral Infections in a Micro-scalable Hepatic Co-culture System. Hepatology 2020, 71, 14–30.
  32. Sayed, I.M.; Seddik, M.I.; Gaber, M.A.; Saber, S.H.; Mandour, S.A.; El-Mokhtar, M.A. Replication of hepatitis e virus (HEV) in primary human-derived monocytes and macrophages in vitro. Vaccines 2020, 8, 239.
  33. El-Mokhtar, M.A.; Seddik, M.I.; Osman, A.; Adel, S.; Abdel Aziz, E.M.; Mandour, S.A.; Mohammed, N.; Zarzour, M.A.; Abdel-Wahid, L.; Radwan, E.; et al. Hepatitis e virus mediates renal injury via the interaction between the immune cells and renal epithelium. Vaccines 2020, 8, 454.
  34. Ladner, S.K.; Otto, M.J.; Barker, C.S.; Zaifert, K.; Wang, G.H.; Guo, J.U.T.; Seeger, C.; King, R.W. Inducible Expression of Human Hepatitis B Virus (HBV) in Stably Transfected Hepatoblastoma Cells: A Novel System for Screening Potential Inhibitors of HBV Replication. Antimicrob. Agents Chemother. 1997, 41, 1715–1720.
  35. Guo, H.; Jiang, D.; Zhou, T.; Cuconati, A.; Block, T.M.; Guo, J.-T. Characterization of the Intracellular Deproteinized Relaxed Circular DNA of Hepatitis B Virus: An Intermediate of Covalently Closed Circular DNA Formation. J. Virol. 2007, 81, 12472–12484.
  36. Yang, D.; Zuo, C.; Wang, X.; Meng, X.; Xue, B.; Liu, N.; Yu, R.; Qin, Y.; Gao, Y.; Wang, Q.; et al. Complete replication of hepatitis B virus and hepatitis C virus in a newly developed hepatoma cell line. Proc. Natl. Acad. Sci. USA 2014, 111, E1264–E1273.
  37. Yang, X.; Cai, W.; Sun, X.; Bi, Y.; Zeng, C.; Zhao, X.Y.; Zhou, Q.; Xu, T.; Xie, Q.; Sun, P.; et al. Defined host factors support HBV infection in non-hepatic 293T cells. J. Cell. Mol. Med. 2020, 24, 2507–2518.
  38. Lempp, F.A.; Schlund, F.; Rieble, L.; Nussbaum, L.; Link, C.; Zhang, Z.; Ni, Y.; Urban, S. Recapitulation of HDV infection in a fully permissive hepatoma cell line allows efficient drug evaluation. Nat. Commun. 2019, 10, 1–11.
  39. So, C.W.; Randall, G. Three-Dimensional Cell Culture Systems for Studying Hepatitis C Virus. Viruses 2021, 13, 211.
  40. Hantz, O.; Parent, R.; Durantel, D.; Gripon, P.; Guguen-Guillouzo, C.; Zoulim, F. Persistence of the hepatitis B virus covalently closed circular DNA in HepaRG human hepatocyte-like cells. J. Gen. Virol. 2009, 90, 127–135.
  41. Yuan, L.; Liu, X.; Zhang, L.; Zhang, Y.; Chen, Y.; Li, X.; Wu, K.; Cao, J.; Hou, W.; Que, Y.; et al. Optimized HepaRG is a suitable cell source to generate the human liver chimeric mouse model for the chronic hepatitis B virus infection. Emerg. Microbes Infect. 2018, 7, 1–17.
  42. Shen, F.; Li, Y.; Wang, Y.; Sozzi, V.; Revill, P.A.; Liu, J.; Gao, L.; Yang, G.; Lu, M.; Sutter, K.; et al. Hepatitis B Virus Sensitivity to interferon-α in Hepatocytes Is More Associated With Cellular Interferon Response Than with Viral Genotype. Hepatology 2018, 67, 1237–1252.
  43. Rogée, S.; Talbot, N.; Caperna, T.; Bouquet, J.; Barnaud, E.; Pavio, N. New models of hepatitis E virus replication in human and porcine hepatocyte cell lines. J. Gen. Virol. 2013, 94, 549–558.
  44. Pellerin, M.; Hirchaud, E.; Blanchard, Y.; Pavio, N.; Doceul, V. Characterization of a cell culture system of persistent hepatitis e virus infection in the human heparg hepatic cell line. Viruses 2021, 13, 406.
  45. Lucifora, J.; Michelet, M.; Salvetti, A.; Durantel, D. Fast Differentiation of HepaRG Cells Allowing Hepatitis B and Delta Virus Infections. Cells 2020, 9, 2288.
  46. Alfaiate, D.; Lucifora, J.; Abeywickrama-Samarakoon, N.; Michelet, M.; Testoni, B.; Cortay, J.C.; Sureau, C.; Zoulim, F.; Dény, P.; Durantel, D. HDV RNA replication is associated with HBV repression and interferon-stimulated genes induction in super-infected hepatocytes. Antivir. Res. 2016, 136, 19–31.
  47. Ni, Y.; Lempp, F.A.; Mehrle, S.; Nkongolo, S.; Kaufman, C.; Fälth, M.; Stindt, J.; Königer, C.; Nassal, M.; Kubitz, R.; et al. Hepatitis B and D viruses exploit sodium taurocholate co-transporting polypeptide for species-specific entry into hepatocytes. Gastroenterology 2014, 146, 1070–1083.e6.
  48. Verrier, E.R.; Colpitts, C.C.; Schuster, C.; Zeisel, M.B.; Baumert, T.F. Cell culture models for the investigation of Hepatitis B and D Virus infection. Viruses 2016, 8, 261.
  49. Farag, M.M.S.; Mansour, M.T.M. Characterization of Subviral Particles of Hepatitis B Virus Produced by HepG2.2.15 Cell Line—In vitro Study. Int. J. Virol. Mol. Biol. 2016, 5, 1–7.
  50. Yan, H.; Zhong, G.; Xu, G.; He, W.; Jing, Z.; Gao, Z.; Huang, Y.; Qi, Y.; Peng, B.; Wang, H.; et al. Sodium taurocholate cotransporting polypeptide is a functional receptor for human hepatitis B and D virus. Elife 2012, 2012.
  51. Otsuka, M.; Aizaki, H.; Kato, N.; Suzuki, T.; Miyamura, T.; Omata, M.; Seki, N. Differential cellular gene expression induced by hepatitis B and C viruses. Biochem. Biophys. Res. Commun. 2003, 300, 443–447.
  52. König, A.; Yang, J.; Jo, E.; Park, K.H.P.; Kim, H.; Than, T.T.; Song, X.; Qi, X.; Dai, X.; Park, S.; et al. Efficient long-term amplification of hepatitis B virus isolates after infection of slow proliferating HepG2-NTCP cells. J. Hepatol. 2019, 71, 289–300.
  53. Shukla, P.; Nguyen, H.T.; Torian, U.; Engle, R.E.; Faulk, K.; Dalton, H.R.; Bendall, R.P.; Keane, F.E.; Purcell, R.H.; Emerson, S.U. Cross-species infections of cultured cells by hepatitis E virus and discovery of an infectious virus-host recombinant. Proc. Natl. Acad. Sci. USA 2011, 108, 2438–2443.
  54. Todt, D.; Friesland, M.; Moeller, N.; Praditya, D.; Kinast, V.; Brüggemann, Y.; Knegendorf, L.; Burkard, T.; Steinmann, J.; Burm, R.; et al. Robust hepatitis e virus infection and transcriptional response in human hepatocytes. Proc. Natl. Acad. Sci. USA 2020, 117, 1731–1741.
  55. Jiang, W. A visualizable hepatitis A virus and hepatitis C virus coinfection model in vitro: Coexistence of two hepatic viruses under limited competition in viral RNA synthesis. bioRxivxiv 2019, 1–22.
  56. Sun, D.; Nassal, M. Stable HepG2- and Huh7-based human hepatoma cell lines for efficient regulated expression of infectious hepatitis B virus. J. Hepatol. 2006, 45, 636–645.
  57. Scarselli, E.; Ansuini, H.; Cerino, R.; Roccasecca, R.M.; Acali, S.; Filocamo, G.; Traboni, C.; Nicosia, A.; Cortese, R.; Vitelli, A. The human scavenger receptor class B type I is a novel candidate receptor for the hepatitis C virus. Embo J. 2002, 21, 5017–5025.
  58. Zhang, J.; Randall, G.; Higginbottom, A.; Monk, P.; Rice, C.M.; McKeating, J.A. CD81 Is Required for Hepatitis C Virus Glycoprotein-Mediated Viral Infection. J. Virol. 2004, 78, 1448–1455.
  59. Vosough, M.; Omidinia, E.; Kadivar, M.; Shokrgozar, M.A.; Pournasr, B.; Aghdami, N.; Baharvand, H. Generation of functional hepatocyte-like cells from human pluripotent stem cells in a scalable suspension culture. Stem Cells Dev. 2013, 22, 2693–2705.
  60. Arez, F.; Rodrigues, A.F.; Brito, C.; Alves, P.M. bioengineered liver cell models of hepatotropic infections. Viruses 2021, 13, 773.
  61. Schwartz, R.E.; Bram, Y.; Frankel, A. Pluripotent Stem Cell-Derived Hepatocyte-like Cells: A Tool to Study Infectious Disease. Curr. Pathobiol. Rep. 2016, 4, 147–156.
  62. Roelandt, P.; Obeid, S.; Paeshuyse, J.; Vanhove, J.; Van Lommel, A.; Nahmias, Y.; Nevens, F.; Neyts, J.; Verfaillie, C.M. Human pluripotent stem cell-derived hepatocytes support complete replication of hepatitis C virus. J. Hepatol. 2012, 57, 246–251.
  63. Ng, S.; Schwartz, R.E.; March, S.; Galstian, A.; Gural, N.; Shan, J.; Prabhu, M.; Mota, M.M.; Bhatia, S.N. Human iPSC-derived hepatocyte-like cells support plasmodium liver-stage infection in vitro. Stem Cell Rep. 2015, 4, 348–359.
  64. Yan, F.; Wang, Y.; Zhang, W.; Chang, M.; He, Z.; Xu, J.; Shang, C.; Chen, T.; Liu, J.; Wang, X.; et al. Human ES Cell-derived Hepatoblasts are an Optimal Lineage Stage for HCV Infection Fang. Hepatology 2017, 66, 717–735.
  65. Kaneko, S.; Kakinuma, S.; Asahina, Y.; Kamiya, A.; Miyoshi, M.; Tsunoda, T.; Nitta, S.; Asano, Y.; Nagata, H.; Otani, S.; et al. Human induced pluripotent stem cell-derived hepatic cell lines as a new model for host interaction with hepatitis B virus. Sci. Rep. 2016, 6, 29358.
  66. Sakurai, F.; Mitani, S.; Yamamoto, T.; Takayama, K.; Tachibana, M.; Watashi, K.; Wakita, T.; Iijima, S.; Tanaka, Y.; Mizuguchi, H. Human induced-pluripotent stem cell-derived hepatocyte-like cells as an in vitro model of human hepatitis B virus infection. Sci. Rep. 2017, 7, 45698.
  67. Schwartz, R.E.; Trehan, K.; Andrus, L.; Sheahan, T.P.; PLoSs, A.; Duncan, S.A.; Rice, C.M.; Bhatia, S.N. Modeling hepatitis C virus infection using human induced pluripotent stem cells. Proc. Natl. Acad. Sci. USA 2012, 109, 2544–2548.
  68. Wu, X.; Robotham, J.M.; Lee, E.; Dalton, S.; Kneteman, N.M.; Gilbert, D.M.; Tang, H. Productive hepatitis C virus infection of stem cell-derived hepatocytes reveals a critical transition to viral permissiveness during differentiation. PLoS Pathog. 2012, 8, e1002617.
  69. Zhou, X.; Sun, P.; Lucendo-Villarin, B.; Angus, A.G.N.; Szkolnicka, D.; Cameron, K.; Farnworth, S.L.; Patel, A.H.; Hay, D.C. Modulating innate immunity improves hepatitis C virus infection and replication in stem cell-derived hepatocytes. Stem Cell Rep. 2014, 3, 204–214.
  70. Dao Thi, V.L.; Debing, Y.; Wu, X.; Rice, C.M.; Neyts, J.; Moradpour, D.; Gouttenoire, J. Sofosbuvir Inhibits Hepatitis e Virus Replication in Vitro and Results in an Additive Effect When Combined with Ribavirin. Gastroenterology 2016, 150, 82–85.e4.
More
Information
Subjects: Virology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 473
Revisions: 2 times (View History)
Update Date: 27 Jul 2021
1000/1000