1
|
Maestro S, Gomez-Echarte N, Camps G, Usai C, Olagüe C, Vales A, Aldabe R, Gonzalez-Aseguinolaza G. Deciphering the Role of Post-Translational Modifications and Cellular Location of Hepatitis Delta Virus (HDV) Antigens in HDV-Mediated Liver Damage in Mice. Viruses 2024; 16:379. [PMID: 38543745 PMCID: PMC10975000 DOI: 10.3390/v16030379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 05/23/2024] Open
Abstract
Hepatitis D virus (HDV) infection represents the most severe form of chronic viral hepatitis. We have shown that the delivery of HDV replication-competent genomes to the hepatocytes using adeno-associated virus (AAV-HDV) as gene delivery vehicles offers a unique platform to investigate the molecular aspects of HDV and associated liver damage. For the purpose of this study, we generated HDV genomes modified by site-directed mutagenesis aimed to (i) prevent some post-translational modifications of HDV antigens (HDAgs) such as large-HDAg (L-HDAg) isoprenylation or short-HDAg (S-HDAg) phosphorylation; (ii) alter the localization of HDAgs within the subcellular compartments; and (iii) inhibit the right conformation of the delta ribozyme. First, the different HDV mutants were tested in vitro using plasmid-transfected Huh-7 cells and then in vivo in C57BL/6 mice using AAV vectors. We found that Ser177 phosphorylation and ribozymal activity are essential for HDV replication and HDAg expression. Mutations of the isoprenylation domain prevented the formation of infectious particles and increased cellular toxicity and liver damage. Furthermore, altering HDAg intracellular localization notably decreased viral replication, though liver damage remained unchanged versus normal HDAg distribution. In addition, a mutation in the nuclear export signal impaired the formation of infectious viral particles. These findings contribute valuable insights into the intricate mechanisms of HDV biology and have implications for therapeutic considerations.
Collapse
Affiliation(s)
- Sheila Maestro
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
- IdiSNA—Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Nahia Gomez-Echarte
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
- IdiSNA—Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Gracian Camps
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
- IdiSNA—Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Carla Usai
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
- IdiSNA—Instituto de Investigación Sanitaria de Navarra, 31008 Pamplona, Spain
| | - Cristina Olagüe
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
| | - Africa Vales
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
| | - Rafael Aldabe
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
| | - Gloria Gonzalez-Aseguinolaza
- DNA & RNA Medicine Division, Centro de Investigación Médica Aplicada, University of Navarra, Avenida Pío XII, 31008 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (C.O.); (A.V.)
| |
Collapse
|
2
|
Yao T, Lv M, Ma S, Chen J, Zhang Y, Yu Y, Zang G, Chen X. Ubiquitinated Hepatitis D Antigen-Loaded Microvesicles Induce a Potent Specific Cellular Immune Response to Inhibit HDV Replication in Vivo. Microbiol Spectr 2021; 9:e0102421. [PMID: 34908456 PMCID: PMC8672902 DOI: 10.1128/spectrum.01024-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Hepatitis D is the most severe form of human viral hepatitis and currently lacks an efficient therapy. Dendritic cell-derived exosomes (Dexs) have been found to induce immune responses capable of eliminating viruses. However, the therapeutic potential of antigen-loaded exosomes in hepatitis D is still unknown. Recently, we designed exosomes loaded with ubiquitinated hepatitis delta virus (HDV) small delta antigen (Ub-S-HDAg) and then treated mice bearing replicating HDV with these exosomes to explore their antiviral effect and mechanism. Mature dendritic cell-derived exosomes (mDexs) were loaded with Ub-S-HDAg and their antivirus function was evaluated in mice with HDV viremia. Furthermore, the proportion of CD8+ cells, the ratio of Th1/Th2 cells, the postimmunization levels of cytokines were explored, and the Janus kinases (JAK)/signal transducer and activator of transcription (STAT) pathway was evaluated with a JAK2 inhibitor AG490. In Ub-S-HDAg-Dexs group, the HDV RNA viral load was significantly decreased compared with other groups by CD8+ cell enrichment and an increase Th1/Th2 cell ratio. Furthermore, lymphocyte infiltration was increased, while the HDAg level was decreased in mouse liver tissue. However, there were no significant differences in HBV surface antigen (HBsAg), alanine aminotransferase (ALT), or aspartate aminotransferase (AST) levels among the groups. Moreover, p-JAK2, p-STAT1, p-STAT4, STAT1, and STAT4 expression was increased in Ub-S-HDAg-Dexs group. In conclusion, Ub-S-HDAg-Dexs might be a potential immunotherapeutic agent for eradicating HDV by inducing specific cellular immune response via the JAK/STAT pathway. IMPORTANCE Hepatitis D is the most severe viral hepatitis with accelerating the process of liver cirrhosis and increasing the risk of hepatocellular carcinoma. However, there are no effective antiviral drugs. Exosomes derived from mature dendritic cells are used not only as immunomodulators, but also as biological carriers to deliver antigens to induce robust immune response. Based on these properties, exosomes could be used as a biological immunotherapy by enhancing adaptive immune response to inhibit hepatitis D virus replication. Our research may provide a new therapeutic strategy to eradicate HDV in the future.
Collapse
Affiliation(s)
- Ting Yao
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Mengjiao Lv
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Siyuan Ma
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Jinmei Chen
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yi Zhang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Yongsheng Yu
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Guoqing Zang
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Xiaohua Chen
- Department of Infectious Diseases, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
3
|
Netter HJ, Barrios MH, Littlejohn M, Yuen LKW. Hepatitis Delta Virus (HDV) and Delta-Like Agents: Insights Into Their Origin. Front Microbiol 2021; 12:652962. [PMID: 34234753 PMCID: PMC8256844 DOI: 10.3389/fmicb.2021.652962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/12/2021] [Indexed: 01/05/2023] Open
Abstract
Hepatitis delta virus (HDV) is a human pathogen, and the only known species in the genus Deltavirus. HDV is a satellite virus and depends on the hepatitis B virus (HBV) for packaging, release, and transmission. Extracellular HDV virions contain the genomic HDV RNA, a single-stranded negative-sense and covalently closed circular RNA molecule, which is associated with the HDV-encoded delta antigen forming a ribonucleoprotein complex, and enveloped by the HBV surface antigens. Replication occurs in the nucleus and is mediated by host enzymes and assisted by cis-acting ribozymes allowing the formation of monomer length molecules which are ligated by host ligases to form unbranched rod-like circles. Recently, meta-transcriptomic studies investigating various vertebrate and invertebrate samples identified RNA species with similarities to HDV RNA. The delta-like agents may be representatives of novel subviral agents or satellite viruses which share with HDV, the self-complementarity of the circular RNA genome, the ability to encode a protein, and the presence of ribozyme sequences. The widespread distribution of delta-like agents across different taxa with considerable phylogenetic distances may be instrumental in comprehending their evolutionary history by elucidating the transition from transcriptome to cellular circular RNAs to infectious subviral agents.
Collapse
Affiliation(s)
- Hans J Netter
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, VIC, Australia.,School of Science, Royal Melbourne Institute of Technology (RMIT) University, Melbourne, VIC, Australia
| | - Marilou H Barrios
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, VIC, Australia.,The Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Margaret Littlejohn
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, VIC, Australia
| | - Lilly K W Yuen
- Victorian Infectious Diseases Reference Laboratory (VIDRL), Melbourne Health, The Peter Doherty Institute, Melbourne, VIC, Australia
| |
Collapse
|
4
|
Maestro S, Gómez-Echarte N, Camps G, Usai C, Suárez L, Vales Á, Olagüe C, Aldabe R, González-Aseguinolaza G. AAV-HDV: An Attractive Platform for the In Vivo Study of HDV Biology and the Mechanism of Disease Pathogenesis. Viruses 2021; 13:v13050788. [PMID: 33925087 PMCID: PMC8145145 DOI: 10.3390/v13050788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatitis delta virus (HDV) infection causes the most severe form of viral hepatitis, but little is known about the molecular mechanisms involved. We have recently developed an HDV mouse model based on the delivery of HDV replication-competent genomes using adeno-associated vectors (AAV), which developed a liver pathology very similar to the human disease and allowed us to perform mechanistic studies. We have generated different AAV-HDV mutants to eliminate the expression of HDV antigens (HDAgs), and we have characterized them both in vitro and in vivo. We confirmed that S-HDAg is essential for HDV replication and cannot be replaced by L-HDAg or host cellular proteins, and that L-HDAg is essential to produce the HDV infectious particle and inhibits its replication. We have also found that lack of L-HDAg resulted in the increase of S-HDAg expression levels and the exacerbation of liver damage, which was associated with an increment in liver inflammation but did not require T cells. Interestingly, early expression of L-HDAg significantly ameliorated the liver damage induced by the mutant expressing only S-HDAg. In summary, the use of AAV-HDV represents a very attractive platform to interrogate in vivo the role of viral components in the HDV life cycle and to better understand the mechanism of HDV-induced liver pathology.
Collapse
Affiliation(s)
- Sheila Maestro
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
| | - Nahia Gómez-Echarte
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
| | - Gracián Camps
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
| | - Carla Usai
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
| | - Lester Suárez
- Suite 110 Research Triangle Park, 20 TW Alexander Drive, AskBio, NC 27709, USA;
| | - África Vales
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
| | - Cristina Olagüe
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
| | - Rafael Aldabe
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
- Correspondence: (R.A.); (G.G.-A.); Tel.: +34-948194700 (ext 4024) (R.A.); +34-948194700 (ext 4024) (G.G.-A.)
| | - Gloria González-Aseguinolaza
- Programa de Terapia Génica y Regulación de la Expresión Génica, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Avenida Pío XII, 31080 Pamplona, Spain; (S.M.); (N.G.-E.); (G.C.); (C.U.); (Á.V.); (C.O.)
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, 31080 Pamplona, Spain
- Correspondence: (R.A.); (G.G.-A.); Tel.: +34-948194700 (ext 4024) (R.A.); +34-948194700 (ext 4024) (G.G.-A.)
| |
Collapse
|
5
|
Abbaspour A, Esmaeilzadeh A, Sharafi A. Suicide gene therapy-mediated purine nucleoside phosphorylase/fludarabine system for in vitro breast cancer model with emphasis on evaluation of vascular endothelial growth factor promoter efficacy. 3 Biotech 2021; 11:140. [PMID: 33708463 DOI: 10.1007/s13205-021-02692-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
In this study, a suicide gene therapy approach was optimized by a non-viral polyplex system based on pEGFP-N1 vector harboring purine nucleoside phosphorylase gene conducted by vascular endothelial growth factor promoter for an in vitro breast cancer model (4T1 cell line). The VEGF promoter and purine nucleoside phosphorylase gene were cloned into the vector from the source of 4T1 and E. coli genomic DNA, respectively. A gene construct was developed by replacing VEGF promoter instead of CMV promoter in pEGFP-N1vector. PNP gene was integrated in to the multiple cloning site of the obtained vector. On the other hand, a construct from pEGFP-N1 harboring PNP gene under the control of the original CMV promoter was developed. The transfection method using cationic polymer was optimized based on N/P ratio, cell cytotoxicity, polyplex size, zeta potential and the green fluorescent protein (GFP) expression by fluorescent microscopy and flowcytometry. Also, the effect of hypoxia condition induced by 0.5 mM H2O2 on the promoter efficiency was investigated. The results showed that the performed gene delivery system is capable of the gene transfection to more than 30% of the cancer cells with both VEGF-PNP-pEGFP-N1 and PNP-pEGFP-N1 plasmids. The hypoxia condition did not show a significant effect on the VEGF promoter. But, it revealed that bystander effect can improve the efficacy of this system and reduce drug IC50 to 2 and fourfold for plasmids VEGF-PNP-pEGFP-N1 and PNP-pEGFP-N1, respectively. These results showed that the bystander effect could almost compensate the low efficiency of non-viral gene delivery systems. We suggest that the tumor-specific gene expression system mediated by the VEGF promoter can be especially useful in the present model of breast cancer gene therapy.
Collapse
Affiliation(s)
- Akbar Abbaspour
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ali Sharafi
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
6
|
Wang J, Qu B, Zhang F, Zhang C, Deng W, Dao Thi VL, Xia Y. Stem Cell-Derived Hepatocyte-Like Cells as Model for Viral Hepatitis Research. Stem Cells Int 2019; 2019:9605252. [PMID: 31281392 PMCID: PMC6594266 DOI: 10.1155/2019/9605252] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Viral hepatitis, the leading cause of liver diseases worldwide, is induced upon infection with hepatotropic viruses, including hepatitis A, B, C, D, and E virus. Due to their obligate intracellular lifestyles, culture systems for efficient viral replication are vital. Although basic and translational research on viral hepatitis has been performed for many years, conventional hepatocellular culture systems are not optimal. These studies have greatly benefited from recent efforts on improving cell culture models for virus replication and infection studies. Here we summarize the use of human stem cell-derived hepatocyte-like cells for hepatotropic virus infection studies, including the dissection of virus-host interactions and virus-induced pathogenesis as well as the identification and validation of novel antiviral agents.
Collapse
Affiliation(s)
- Jingjing Wang
- State Key Laboratory of Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Bingqian Qu
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fang Zhang
- Department of Translational Medicine, Baruch S. Blumberg Institute, Doylestown, PA, USA
| | - Cindy Zhang
- Schaller Research Group at Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, Heidelberg, Germany
- BioQuant Center, University of Heidelberg, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Wanyan Deng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Viet Loan Dao Thi
- Schaller Research Group at Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Cluster of Excellence CellNetworks, Heidelberg, Germany
| | - Yuchen Xia
- State Key Laboratory of Virology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Ye X, Tateno C, Thi EP, Kakuni M, Snead NM, Ishida Y, Barnard TR, Sofia MJ, Shimada T, Lee ACH. Hepatitis B Virus Therapeutic Agent ARB-1740 Has Inhibitory Effect on Hepatitis Delta Virus in a New Dually-Infected Humanized Mouse Model. ACS Infect Dis 2019; 5:738-749. [PMID: 30408957 DOI: 10.1021/acsinfecdis.8b00192] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Hepatitis delta virus (HDV) infects 10-20 million individuals worldwide and causes severe fulminant hepatitis with high likelihood of cirrhosis and hepatocellular carcinoma. HDV infection cannot occur in the absence of the surface antigen (HBsAg) of the hepatitis B virus. RNA interference is an effective mechanism by which to inhibit viral transcripts, and siRNA therapeutics sharing this mechanism have begun to demonstrate clinical efficacy. Here we assessed the outcome of HBV-targeting siRNA intervention against HDV and compared it to a direct anti-HDV siRNA approach in dually infected humanized mice. Treatment with ARB-1740, a clinical stage HBV-targeting siRNA agent delivered using lipid nanoparticle (LNP) technology, effectively reduced HBV viremia by 2.3 log10 and serum HBsAg by 2.6 log10, leading to 1.6 log10 reduction of HDV viremia. In contrast, HDV-targeting siRNA inhibited HDV in both blood and liver compartments without affecting HBV and PEGylated interferon-alpha reduced HBV viremia by 2.0 log10 but had no effect on HDV viremia under these study conditions. These results illustrate the inhibitory effects of siRNAs against these two viral infections and suggest that ARB-1740 may be of therapeutic benefit for hepatitis delta patients, a subpopulation with high unmet medical need.
Collapse
Affiliation(s)
- Xin Ye
- Arbutus Biopharma, 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Chise Tateno
- PhoenixBio Co., Ltd., 3-4-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, Japan
| | - Emily P. Thi
- Arbutus Biopharma, 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Masakazu Kakuni
- PhoenixBio Co., Ltd., 3-4-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, Japan
| | - Nicholas M. Snead
- Arbutus Biopharma, 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Yuji Ishida
- PhoenixBio Co., Ltd., 3-4-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, Japan
| | - Trisha R. Barnard
- Arbutus Biopharma, 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Michael J. Sofia
- Arbutus Biopharma, 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| | - Takashi Shimada
- PhoenixBio Co., Ltd., 3-4-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, Japan
| | - Amy C. H. Lee
- Arbutus Biopharma, 701 Veterans Circle, Warminster, Pennsylvania 18974, United States
| |
Collapse
|
8
|
Alfaiate D, Durantel D. [A new immune competent mouse model for the study of hepatitis D virus infection]. Med Sci (Paris) 2017; 33:570-573. [PMID: 28990549 DOI: 10.1051/medsci/20173306004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Dulce Alfaiate
- Département de pathologie et immunologie, université de Genève, 1211 Genève, Suisse
| | - David Durantel
- Centre de recherche en cancérologie de Lyon, UMR Inserm U1052, CNRS UMR-5286, 69424 Lyon Cedex 03, France - Université Claude-Bernard Lyon 1, Lyon, France
| |
Collapse
|
9
|
Suárez-Amarán L, Usai C, Di Scala M, Godoy C, Ni Y, Hommel M, Palomo L, Segura V, Olagüe C, Vales A, Ruiz-Ripa A, Buti M, Salido E, Prieto J, Urban S, Rodríguez-Frias F, Aldabe R, González-Aseguinolaza G. A new HDV mouse model identifies mitochondrial antiviral signaling protein (MAVS) as a key player in IFN-β induction. J Hepatol 2017; 67:669-679. [PMID: 28527664 DOI: 10.1016/j.jhep.2017.05.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Studying hepatitis delta virus (HDV) and developing new treatments is hampered by the limited availability of small animal models. Herein, a description of a robust mouse model of HDV infection that mimics several important characteristics of the human disease is presented. METHODS HDV and hepatitis B virus (HBV) replication competent genomes were delivered to the mouse liver using adeno-associated viruses (AAV; AAV-HDV and AAV-HBV). Viral load, antigen expression and genomes were quantified at different time points after AAV injection. Furthermore, liver pathology, genome editing, and the activation of the innate immune response were evaluated. RESULTS AAV-HDV infection initiated HDV replication in mouse hepatocytes. Genome editing was confirmed by the presence of small and large HDV antigens and sequencing. Viral replication was detected for 45days, even after the AAV-HDV vector had almost disappeared. In the presence of HBV, HDV infectious particles were detected in serum. Furthermore, as observed in patients, co-infection was associated with the reduction of HBV antigen expression and the onset of liver damage that included the alteration of genes involved in the development of liver pathologies. HDV replication induced a sustained type I interferon response, which was significantly reduced in immunodeficient mice and almost absent in mitochondrial antiviral signaling protein (MAVS)-deficient mice. CONCLUSION The animal model described here reproduces important characteristics of human HDV infection and provides a valuable tool for characterizing the viral infection and for developing new treatments. Furthermore, MAVS was identified as a main player in HDV detection and adaptive immunity was found to be involved in the amplification of the innate immune response. Lay summary: Co-infection with hepatitis B and D virus (HBV and HDV, respectively) often causes a more severe disease condition than HBV alone. Gaining more insight into HDV and developing new treatments is hampered by limited availability of adequate immune competent small animal models and new ones are needed. Here, a mouse model of HDV infection is described, which mimics several important characteristics of the human disease, such as the initiation and maintenance of replication in murine hepatocytes, genome editing and, in the presence of HBV, generation of infectious particles. Lastly, the involvement of an adaptive immunity and the intracellular signaling molecule MAVS in mounting a strong and lasting innate response was shown. Thus, our model serves as a useful tool for the investigation of HDV biology and new treatments.
Collapse
MESH Headings
- Adaptive Immunity
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Animals
- Cell Line
- Coinfection/immunology
- Coinfection/pathology
- Coinfection/virology
- Dependovirus/genetics
- Disease Models, Animal
- Genome, Viral
- Hepatitis B/complications
- Hepatitis B/immunology
- Hepatitis B/virology
- Hepatitis B Antigens/metabolism
- Hepatitis B virus/genetics
- Hepatitis B virus/immunology
- Hepatitis D/complications
- Hepatitis D/immunology
- Hepatitis D/virology
- Hepatitis Delta Virus/genetics
- Hepatitis Delta Virus/immunology
- Hepatitis Delta Virus/physiology
- Hepatitis delta Antigens/metabolism
- Humans
- Immunity, Innate
- Interferon-beta/biosynthesis
- Liver/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Models, Immunological
- Signal Transduction/immunology
- Virus Replication
Collapse
Affiliation(s)
- Lester Suárez-Amarán
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Carla Usai
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Marianna Di Scala
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Cristina Godoy
- Centro de Investigación Biomédica en red: Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain; Liver Pathology Unit, Departments of Biochemistry and Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain; Virology Unit, Department of Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Yi Ni
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mirja Hommel
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Laura Palomo
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Víctor Segura
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain; Bioinformatics Unit, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Cristina Olagüe
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Africa Vales
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Alicia Ruiz-Ripa
- Centro de Investigación Biomédica en red: Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain; Liver Pathology Unit, Departments of Biochemistry and Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain; Virology Unit, Department of Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Buti
- Centro de Investigación Biomédica en red: Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain; Liver Pathology Unit, Departments of Biochemistry and Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain; Virology Unit, Department of Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eduardo Salido
- Department of Pathology, Centre for Biomedical Research on Rare Diseases (CIBERER), La Laguna, S/C Tenerife, Spain
| | - Jesús Prieto
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain; Centro de Investigación Biomédica en red: Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Pamplona, Spain
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Francisco Rodríguez-Frias
- Centro de Investigación Biomédica en red: Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain; Liver Pathology Unit, Departments of Biochemistry and Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain; Virology Unit, Department of Microbiology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Rafael Aldabe
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain
| | - Gloria González-Aseguinolaza
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Calle Irunlarrea 3, Pamplona 31008, Spain.
| |
Collapse
|
10
|
Moreno D, Neri L, Vicente E, Vales A, Aldabe R. Use of Thymidine Kinase Recombinant Adenovirus and Ganciclovir Mediated Mouse Liver Preconditioning for Hepatocyte Xenotransplantation. Methods Mol Biol 2017; 1506:179-192. [PMID: 27830553 DOI: 10.1007/978-1-4939-6506-9_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Hepatocyte transplantation is the best approach to maintain and propagate differentiated hepatocytes from different species. Host liver has to be adapted for transplanted hepatocytes productive engraftment and proliferation being required a chronic liver injury to eliminate host hepatocytes and provide a proliferative advantage to the transplanted hepatocytes. Most valuable mouse models for xenograft hepatocyte transplantation are based on genetically modified animals to cause a chronic liver damage and to limit host hepatocyte regeneration potential. We present a methodology that generates a chronic liver damage and can be applied to any host mouse strain and animal species based on the inoculation of a recombinant adenovirus to express herpes simplex thymidine kinase in host hepatocytes sensitizing them to ganciclovir treatment. This causes a prolonged liver damage that allows hepatocyte transplantation and generation of regenerative nodules in recipient mouse liver integrated by transplanted cells and host sinusoidal. Obtained chimeric animals maintain functional chimeric nodules for several weeks, ready to be used in any study.
Collapse
Affiliation(s)
- Daniel Moreno
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Leire Neri
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Eva Vicente
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Africa Vales
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Rafael Aldabe
- Gene Therapy and Regulation of Gene Expression Program, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, 31008, Spain.
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
11
|
Sukowati CHC, El-Khobar KE, Ie SI, Anfuso B, Muljono DH, Tiribelli C. Significance of hepatitis virus infection in the oncogenic initiation of hepatocellular carcinoma. World J Gastroenterol 2016; 22:1497-1512. [PMID: 26819517 PMCID: PMC4721983 DOI: 10.3748/wjg.v22.i4.1497] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/06/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related death worldwide. Chronic infection of hepatitis B virus (HBV) and/or hepatitis C virus (HCV) is a major risk factor in the development of the HCC, independently from excessive alcohol abuse and metabolic disease. Since the biology of HBV and HCV is different, their oncogenic effect may go through different mechanisms, direct and/or indirect. Viral hepatitis infection is associated with cellular inflammation, oxidative stress, and DNA damage, that may lead to subsequent hepatic injuries such as chronic hepatitis, fibrosis, cirrhosis, and finally HCC. Direct oncogenic properties of these viruses are related with their genotypic characteristics and the ability of viral proteins to interact with host proteins, thus altering the molecular pathways balance of the cells. In addition, the integration of HBV DNA, especially the gene S and X, in a particular site of the host genome can disrupt chromosomal stability and may activate various oncogenic mechanisms, including those in hematopoietic cells. Recently, several studies also had demonstrated that viral hepatitis could trigger the population of hepatic cancer stem cells. This review summarize available pre-clinical and clinical data in literature regarding oncogenic properties of HBV and HCV in the early initiation of HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/epidemiology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Viral
- Gene Expression Regulation, Neoplastic
- Gene Expression Regulation, Viral
- Genotype
- Hepacivirus/genetics
- Hepacivirus/pathogenicity
- Hepatitis B virus/genetics
- Hepatitis B virus/pathogenicity
- Hepatitis B, Chronic/complications
- Hepatitis B, Chronic/virology
- Hepatitis C, Chronic/complications
- Hepatitis C, Chronic/virology
- Host-Pathogen Interactions
- Humans
- Liver Neoplasms/epidemiology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/virology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/virology
- Oncogenes
- Risk Factors
Collapse
|
12
|
Chen J, Wu M, Liu K, Zhang W, Li Y, Zhou X, Bai L, Yuan Z. New insights into hepatitis B virus biology and implications for novel antiviral strategies. Natl Sci Rev 2015. [DOI: 10.1093/nsr/nwv044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Abstract
Hepatitis B virus (HBV), a small DNA virus with a unique replication mode, can cause chronic hepatitis (CHB), which is characterized by the persistence of the viral covalently closed circular DNA that serves as the template for HBV replication and the production of large amounts of secreted HBV surface antigen (HBsAg) that is present in excess of the levels of infectious virus. Despite the success of currently approved antiviral treatments for CHB patients, including interferon and nucleotide analogs, which suppress HBV replication and reduce the risk of CHB-related liver diseases, these therapies fail to eradicate the virus in most of the patients. With the development of the cell and animal models for HBV study, a better understanding of the HBV life cycle has been achieved and a series of novel antiviral strategies that target different stages of HBV replication have been designed to overcome the viral factors that contribute to HBV persistence. Such basic HBV research advancements and therapeutic developments are the subject of this review.
Collapse
Affiliation(s)
- Jieliang Chen
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Min Wu
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Kuancheng Liu
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institutes of Medical Microbiology and Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Wen Zhang
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaohui Zhou
- Research Unit, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lu Bai
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology, Ministry of Education and Health, and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Institutes of Medical Microbiology and Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|