51
|
Kirstgen M, Lowjaga KAAT, Müller SF, Goldmann N, Lehmann F, Glebe D, Baringhaus KH, Geyer J. Hepatitis D Virus Entry Inhibitors Based on Repurposing Intestinal Bile Acid Reabsorption Inhibitors. Viruses 2021; 13:v13040666. [PMID: 33921515 PMCID: PMC8068820 DOI: 10.3390/v13040666] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Identification of Na+/taurocholate co-transporting polypeptide (NTCP) as high-affinity hepatic entry receptor for the Hepatitis B and D viruses (HBV/HDV) opened the field for target-based development of cell-entry inhibitors. However, most of the HBV/HDV entry inhibitors identified so far also interfere with the physiological bile acid transporter function of NTCP. The present study aimed to identify more virus-selective inhibitors of NTCP by screening of 87 propanolamine derivatives from the former development of intestinal bile acid reabsorption inhibitors (BARIs), which interact with the NTCP-homologous intestinal apical sodium-dependent bile acid transporter (ASBT). In NTCP-HEK293 cells, the ability of these compounds to block the HBV/HDV-derived preS1-peptide binding to NTCP (virus receptor function) as well as the taurocholic acid transport via NTCP (bile acid transporter function) were analyzed in parallel. Hits were subsequently validated by performing in vitro HDV infection experiments in NTCP-HepG2 cells. The most potent compounds S985852, A000295231, and S973509 showed in vitro anti-HDV activities with IC50 values of 15, 40, and 70 µM, respectively, while the taurocholic acid uptake inhibition occurred at much higher IC50 values of 24, 780, and 490 µM, respectively. In conclusion, repurposing of compounds from the BARI class as novel HBV/HDV entry inhibitors seems possible and even enables certain virus selectivity based on structure-activity relationships.
Collapse
Affiliation(s)
- Michael Kirstgen
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg (BFS), Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany; (M.K.); (K.A.A.T.L.); (S.F.M.)
| | - Kira Alessandra Alicia Theresa Lowjaga
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg (BFS), Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany; (M.K.); (K.A.A.T.L.); (S.F.M.)
| | - Simon Franz Müller
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg (BFS), Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany; (M.K.); (K.A.A.T.L.); (S.F.M.)
| | - Nora Goldmann
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University Giessen, 35392 Giessen, Germany; (N.G.); (F.L.); (D.G.)
| | - Felix Lehmann
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University Giessen, 35392 Giessen, Germany; (N.G.); (F.L.); (D.G.)
| | - Dieter Glebe
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University Giessen, 35392 Giessen, Germany; (N.G.); (F.L.); (D.G.)
- German Center for Infection Research (DZIF), Giessen-Marburg-Langen Partner Site, 35392 Giessen, Germany
| | | | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg (BFS), Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392 Giessen, Germany; (M.K.); (K.A.A.T.L.); (S.F.M.)
- Correspondence: ; Tel.: +49-641-99-38404; Fax: +49-641-99-38409
| |
Collapse
|
52
|
Chothe PP, Pemberton R, Hariparsad N. Function and Expression of Bile Salt Export Pump in Suspension Human Hepatocytes. Drug Metab Dispos 2021; 49:314-321. [PMID: 33472814 DOI: 10.1124/dmd.120.000057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 01/07/2021] [Indexed: 11/22/2022] Open
Abstract
The mechanistic understanding of bile salt disposition is not well established in suspension human hepatocytes (SHH) because of the limited information on the expression and function of bile salt export protein (BSEP) in this system. We investigated the transport function of BSEP in SHH using a method involving in situ biosynthesis of bile salts from their precursor bile acids, cholic acid (CA) and chenodeoxycholic acid (CDCA). Our data indicated that glycine- and taurine-conjugated CA and CDCA were generated efficiently and transported out of hepatocytes in a concentration- and time-dependent manner. We also observed that the membrane protein abundance of BSEP was similar between SHH and sandwich-cultured human hepatocytes. Furthermore, known cholestatic agents significantly inhibited G-CA and G-CDCA efflux in SHH. Interestingly, cyclosporine A, troglitazone, itraconazole, loratadine, and lovastatin inhibited G-CA efflux more potently than G-CDCA efflux (3- to 5-fold). Because of these significant differential effects on G-CA and G-CDCA efflux inhibition, we determined the IC50 values of troglitazone for G-CA (9.9 µM) and for G-CDCA (43.1 µM) efflux. The observed discrepancy in the IC50 was attributed to the fact that troglitazone also inhibits organic anion transporting polypeptides and Na+/taurocholate cotransporting polypeptide in addition to BSEP. The hepatocyte uptake study suggested that both active uptake and passive diffusion contribute to the liver uptake of CA, whereas CDCA primarily undergoes passive diffusion into the liver. In summary, these data demonstrated the expression and function of BSEP and its major role in transport of bile salts in cryopreserved SHH. SIGNIFICANCE STATEMENT: BSEP transport function and protein abundance was evident in SHH in the present study. The membrane abundance of BSEP protein was similar between SHH and sandwich-cultured human hepatocytes. The study also illustrated the major role of BSEP relative to basolateral MRP3 and MRP4 in transport of bile salts in SHH. Understanding of BSEP function in SHH may bolster the utility of this platform in mechanistic understanding of bile salt disposition and potentially in the assessment of drugs for BSEP inhibition.
Collapse
Affiliation(s)
- Paresh P Chothe
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Rachel Pemberton
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Niresh Hariparsad
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| |
Collapse
|
53
|
Wu MR, Huang YY, Hsiao JK. Role of Sodium Taurocholate Cotransporting Polypeptide as a New Reporter and Drug-Screening Platform: Implications for Preventing Hepatitis B Virus Infections. Mol Imaging Biol 2021; 22:313-323. [PMID: 31140111 DOI: 10.1007/s11307-019-01373-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Sodium taurocholate cotransporting polypeptide (NTCP) is a transmembrane protein responsible for delivering indocyanine green (ICG), an ideal infrared fluorescent dye, from extracellular space into the cytoplasm. Additionally, NTCP located in the hepatocyte membrane is the portal for hepatitis B and D virus (HBV/HDV) infections. This study verified the feasibility of NTCP as a reporter and further established a drug-screening platform for HBV/HDV infections. PROCEDURES NTCP was transduced into HT-29, a colorectal cancer cell line. To examine the use of NTCP as a reporter, NTCP-expressing cells were treated with ICG and examined through flow cytometry, an in vivo imaging system (IVIS), and confocal microscopy. Furthermore, ICG was administrated to NTCP-expressing tumor-bearing nude mice and examined using the IVIS. To study the drug-screening platform, NTCP-expressing cells were treated with cyclosporin A, an NTCP inhibitor, and ICG, and examined using a multimode detection platform. Moreover, nude mice were injected with NTCP inhibitors and ICG, and subsequently, their ICG signal was examined in vivo and in the blood. RESULTS In the reporter study, the ICG signal was higher in NTCP-expressing cells/tumors than in control cells/tumors after ICG treatment. In the drug-screening platform study, NTCP-expressing cells had decreased ICG intensity after treatment with NTCP inhibitors and ICG. Nude mice that were administered cyclosporin A had lower ICG intensity in the liver and higher intensity in the peripheral tissue and blood. CONCLUSIONS NTCP and ICG form an ideal reporter system with extensive applications in cancer biology, robust drug-drug interactions, and drug screening in HBV/HDV infections.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan.,Department of Medical Imaging, Taipei Tzuchi General Hospital, Buddhist Tzu-Chi Medical Foundation, No.289, Jianguo Rd., Xindian Dist., New Taipei City, 23142, Taiwan
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzuchi General Hospital, Buddhist Tzu-Chi Medical Foundation, No.289, Jianguo Rd., Xindian Dist., New Taipei City, 23142, Taiwan. .,School of Medicine, Tzu Chi University, No.701, Sec. 3, Zhongyang Rd., Hualien, 97004, Taiwan.
| |
Collapse
|
54
|
Müller JP, Keufgens L, Gründemann D. Hyperosmolarity stimulates transporter-mediated insertion of estrone sulfate into the plasma membrane, but inhibits the uptake by SLC10A1 (NTCP). Biochem Pharmacol 2021; 186:114484. [PMID: 33617845 DOI: 10.1016/j.bcp.2021.114484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Many drugs are largely hydrophobic molecules; a transporter might conceivably insert these into the plasma membrane. At least 18 transporters from diverse families have been reported to transport the model compound estrone sulfate alias estrone-3-sulfate (E3S). Out of these, we recently examined SLC22A11 (OAT4). We concluded from a comparison of E3S and uric acid transport that SLC22A11 does not translocate E3S into the cytosol, but into the plasma membrane. Here we present a hyperosmolarity alias hypertonicity assay to differentiate transport mechanisms. Human transporters were expressed heterologously in 293 cells. Solute uptake into intact cells was measured by LC-MS. Addition of mannitol or sucrose led to rapid cell shrinkage, but cell viability after 60 min in hyperosmolar buffer was not impaired. A decrease in substrate accumulation with increasing osmolarity as observed here for several substrates and the transporters SLC22A11, ETT (SLC22A4), OCT2 (SLC22A2), OAT3 (SLC22A8), and MATE1 (SLC47A1) suggests regular substrate translocation into the cytosol. An increase as observed for E3S transport by SLC22A11, OAT3, MATE1, SLC22A9, and SLC10A6 implies insertion into the membrane. In marked contrast to the other E3S transporters, the bile acid transporter SLC10A1 (NTCP, Na+ taurocholate co-transporting polypeptide) showed a decrease in the accumulation of E3S in hyperosmolar buffer; the same was observed with taurocholic acid. Indeed, our data from several functional assays strongly suggest that the transport mechanism is identical for both substrates. Apparently, a unique transport mechanism has been established for SLC10A1 by evolution that ensures the transport of amphipathic, detergent-like molecules into the cytosol.
Collapse
Affiliation(s)
- Julian Peter Müller
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, Cologne 50931, Germany
| | - Lena Keufgens
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, Cologne 50931, Germany
| | - Dirk Gründemann
- Department of Pharmacology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Gleueler Straße 24, Cologne 50931, Germany.
| |
Collapse
|
55
|
Lowjaga KAAT, Kirstgen M, Müller SF, Goldmann N, Lehmann F, Glebe D, Geyer J. Long-term trans-inhibition of the hepatitis B and D virus receptor NTCP by taurolithocholic acid. Am J Physiol Gastrointest Liver Physiol 2021; 320:G66-G80. [PMID: 33174454 DOI: 10.1152/ajpgi.00263.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human hepatic bile acid transporter Na+/taurocholate cotransporting polypeptide (NTCP) represents the liver-specific entry receptor for the hepatitis B and D viruses (HBV/HDV). Chronic hepatitis B and D affect several million people worldwide, but treatment options are limited. Recently, HBV/HDV entry inhibitors targeting NTCP have emerged as promising novel drug candidates. Nevertheless, the exact molecular mechanism that NTCP uses to mediate virus binding and entry into hepatocytes is still not completely understood. It is already known that human NTCP mRNA expression is downregulated under cholestasis. Furthermore, incubation of rat hepatocytes with the secondary bile acid taurolithocholic acid (TLC) triggers internalization of the rat Ntcp protein from the plasma membrane. In the present study, the long-term inhibitory effect of TLC on transport function, HBV/HDV receptor function, and membrane expression of human NTCP were analyzed in HepG2 and human embryonic kidney (HEK293) cells stably overexpressing NTCP. Even after short-pulse preincubation, TLC had a significant long-lasting inhibitory effect on the transport function of NTCP, but the NTCP protein was still present at the plasma membrane. Furthermore, binding of the HBV/HDV myr-preS1 peptide and susceptibility for in vitro HDV infection were significantly reduced by TLC preincubation. We hypothesize that TLC rapidly accumulates in hepatocytes and mediates long-lasting trans-inhibition of the transport and receptor function of NTCP via a particular TLC-binding site at an intracellularly accessible domain of NTCP. Physiologically, this trans-inhibition might protect hepatocytes from toxic overload of bile acids. Pharmacologically, it provides an interesting novel NTCP target site for potential long-acting HBV/HDV entry inhibitors.NEW & NOTEWORTHY The hepatic bile acid transporter NTCP is a high-affinity receptor for hepatitis B and D viruses. This study shows that TLC rapidly accumulates in NTCP-expressing hepatoma cells and mediates long-lasting trans-inhibition of NTCP's transporter and receptor function via an intracellularly accessible domain, without substantially affecting its membrane expression. This domain is a promising novel NTCP target site for pharmacological long-acting HBV/HDV entry inhibitors.
Collapse
Affiliation(s)
- Kira A A T Lowjaga
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Michael Kirstgen
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Simon F Müller
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| | - Nora Goldmann
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Felix Lehmann
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Dieter Glebe
- Institute of Medical Virology, National Reference Center for Hepatitis B Viruses and Hepatitis D Viruses, Justus Liebig University, Giessen, Germany
| | - Joachim Geyer
- Faculty of Veterinary Medicine, Institute of Pharmacology and Toxicology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
56
|
Liu Y, Ruan H, Li Y, Sun G, Liu X, He W, Mao F, He M, Yan L, Zhong G, Yan H, Li W, Zhang Z. Potent and Specific Inhibition of NTCP-Mediated HBV/HDV Infection and Substrate Transporting by a Novel, Oral-Available Cyclosporine A Analogue. J Med Chem 2020; 64:543-565. [PMID: 33369415 DOI: 10.1021/acs.jmedchem.0c01484] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Analogues of the natural product cyclosporine A (CsA) were developed and assessed as antivirals against infection of hepatitis B virus (HBV) and its satellite hepatitis D virus (HDV). An analogue termed 27A exhibits potent inhibition of HBV/HDV infection by specifically blocking viral engagement to its cellular receptor NTCP, while it lacks immunosuppressive activity found in natural CsA. Intraperitoneal injection or oral intake of 27A protects HDV-susceptible mouse model from HDV infection. 27A serves as a promising lead for the development of novel anti-HDV/HBV agents.
Collapse
Affiliation(s)
- Yang Liu
- National Institute of Biological Sciences, Beijing 102206, China.,Graduate Program, Tsinghua University, Beijing 100084, China
| | - Hanying Ruan
- National Institute of Biological Sciences, Beijing 102206, China
| | - Ying Li
- National Institute of Biological Sciences, Beijing 102206, China
| | - Guoliang Sun
- National Institute of Biological Sciences, Beijing 102206, China.,Graduate Program, Peking University, Beijing 100080, China
| | - Xiao Liu
- National Institute of Biological Sciences, Beijing 102206, China
| | - Wenhui He
- National Institute of Biological Sciences, Beijing 102206, China
| | - Fengfeng Mao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Miaomiao He
- National Institute of Biological Sciences, Beijing 102206, China.,Graduate Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Liwei Yan
- National Institute of Biological Sciences, Beijing 102206, China
| | - Guocai Zhong
- National Institute of Biological Sciences, Beijing 102206, China
| | - Huan Yan
- National Institute of Biological Sciences, Beijing 102206, China
| | - Wenhui Li
- National Institute of Biological Sciences, Beijing 102206, China
| | - Zhiyuan Zhang
- National Institute of Biological Sciences, Beijing 102206, China
| |
Collapse
|
57
|
Kirstgen M, Lowjaga KAAT, Müller SF, Goldmann N, Lehmann F, Alakurtti S, Yli-Kauhaluoma J, Glebe D, Geyer J. Selective hepatitis B and D virus entry inhibitors from the group of pentacyclic lupane-type betulin-derived triterpenoids. Sci Rep 2020; 10:21772. [PMID: 33303817 PMCID: PMC7729925 DOI: 10.1038/s41598-020-78618-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
Current treatment options against hepatitis B and D virus (HBV/HDV) infections have only limited curative effects. Identification of Na+/taurocholate co-transporting polypeptide (NTCP) as the high-affinity hepatic receptor for both viruses in 2012 enables target-based development of HBV/HDV cell-entry inhibitors. Many studies already identified appropriate NTCP inhibitors. However, most of them interfere with NTCP’s physiological function as a hepatic bile acid transporter. To overcome this drawback, the present study aimed to find compounds that specifically block HBV/HDV binding to NTCP without affecting its transporter function. A novel assay was conceptualized to screen for both in parallel; virus binding to NTCP (measured via binding of a preS1-derived peptide of the large HBV/HDV envelope protein) and bile acid transport via NTCP. Hits were subsequently validated by in vitro HDV infection studies using NTCP-HepG2 cells. Derivatives of the birch-derived pentacyclic lupane-type triterpenoid betulin revealed clear NTCP inhibitory potency and selectivity for the virus receptor function of NTCP. Best performing compounds in both aspects were 2, 6, 19, and 25. In conclusion, betulin derivatives show clear structure–activity relationships for potent and selective inhibition of the HBV/HDV virus receptor function of NTCP without tackling its physiological bile acid transport function and therefore are promising drug candidates.
Collapse
Affiliation(s)
- Michael Kirstgen
- Biomedical Research Center Seltersberg (BFS), Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392, Giessen, Germany
| | - Kira Alessandra Alicia Theresa Lowjaga
- Biomedical Research Center Seltersberg (BFS), Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392, Giessen, Germany
| | - Simon Franz Müller
- Biomedical Research Center Seltersberg (BFS), Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392, Giessen, Germany
| | - Nora Goldmann
- National Reference Center for Hepatitis B Viruses and D Viruses, Institute of Medical Virology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Felix Lehmann
- National Reference Center for Hepatitis B Viruses and D Viruses, Institute of Medical Virology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Sami Alakurtti
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, 00014, Helsinki, Finland.,VTT Technical Research Centre of Finland, Biologinkuja 7, P.O. Box 1000, 02044, Espoo, Finland
| | - Jari Yli-Kauhaluoma
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, P.O. Box 56, 00014, Helsinki, Finland
| | - Dieter Glebe
- National Reference Center for Hepatitis B Viruses and D Viruses, Institute of Medical Virology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Joachim Geyer
- Biomedical Research Center Seltersberg (BFS), Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, 35392, Giessen, Germany.
| |
Collapse
|
58
|
Association of the Hepatitis B Virus Large Surface Protein with Viral Infectivity and Endoplasmic Reticulum Stress-mediated Liver Carcinogenesis. Cells 2020; 9:cells9092052. [PMID: 32911838 PMCID: PMC7563867 DOI: 10.3390/cells9092052] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B is the most prevalent viral hepatitis worldwide, affecting approximately one-third of the world’s population. Among HBV factors, the surface protein is the most sensitive biomarker for viral infection, given that it is expressed at high levels in all viral infection phases. The large HBV surface protein (LHBs) contains the integral pre-S1 domain, which binds to the HBV receptor sodium taurocholate co transporting polypeptide on the hepatocyte to facilitate viral entry. The accumulation of viral LHBs and its prevalent pre-S mutants in chronic HBV carriers triggers a sustained endoplasmic reticulum (ER) overload response, leading to ER stress-mediated cell proliferation, metabolic switching and genomic instability, which are associated with pro-oncogenic effects. Ground glass hepatocytes identified in HBV-related hepatocellular carcinoma (HCC) patients harbor pre-S deletion variants that largely accumulate in the ER lumen due to mutation-induced protein misfolding and are associated with increased risks of cancer recurrence and metastasis. Moreover, in contrast to the major HBs, which is decreased in tumors to a greater extent than it is in peritumorous regions, LHBs is continuously expressed during tumorigenesis, indicating that LHBs serves as a promising biomarker for HCC in people with CHB. Continuing efforts to delineate the molecular mechanisms by which LHBs regulates pathological changes in CHB patients are important for establishing a correlation between LHBs biomarkers and HCC development.
Collapse
|
59
|
Wettengel JM, Burwitz BJ. Innovative HBV Animal Models Based on the Entry Receptor NTCP. Viruses 2020; 12:E828. [PMID: 32751581 PMCID: PMC7472226 DOI: 10.3390/v12080828] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
Hepatitis B is a major global health problem, with an estimated 257 million chronically infected patients and almost 1 million deaths per year. The causative agent is hepatitis B virus (HBV), a small, enveloped, partially double-stranded DNA virus. HBV has a strict species specificity, naturally infecting only humans and chimpanzees. Sodium taurocholate co-transporting polypeptide (NTCP), a bile acid transporter expressed on hepatocytes, has been shown to be one of the key factors in HBV infection, playing a crucial role in the HBV entry process in vitro and in vivo. Variations in the amino acid sequence of NTCP can inhibit HBV infection and, therefore, contributes, in part, to the species barrier. This discovery has revolutionized the search for novel animal models of HBV. Indeed, it was recently shown that variations in the amino acid sequence of NTCP represent the sole species barrier for HBV infection in macaques. Here, we review what is known about HBV entry through the NTCP receptor and highlight how this knowledge has been harnessed to build new animal models for the study of HBV pathogenesis and curative therapies.
Collapse
Affiliation(s)
- Jochen M. Wettengel
- Institute of Virology, Technische Universität München/Helmholtz Zentrum München, Trogerstr. 30, 81675 Munich, Germany;
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 N.W. 185th Avenue Beaverton, Tanasbourne, OR 97006, USA
| | - Benjamin J. Burwitz
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, 505 N.W. 185th Avenue Beaverton, Tanasbourne, OR 97006, USA
| |
Collapse
|
60
|
Guo WL, Guo JB, Liu BY, Lu JQ, Chen M, Liu B, Bai WD, Rao PF, Ni L, Lv XC. Ganoderic acid A from Ganoderma lucidum ameliorates lipid metabolism and alters gut microbiota composition in hyperlipidemic mice fed a high-fat diet. Food Funct 2020; 11:6818-6833. [PMID: 32686808 DOI: 10.1039/d0fo00436g] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Ganoderic acid A (GA) is one of the most abundant triterpenoids in Ganoderma lucidum, and has been proved to possess a wide range of beneficial health effects. The aim of the current study is to investigate the amelioration effects and mechanism of GA on improving hyperlipidemia in mice fed a high-fat diet (HFD). The results showed that GA intervention significantly inhibited the abnormal growth of body weight and epididymal white adipose tissue (eWAT), prevented the hypertrophy of epididymal adipocytes, and ameliorated the biochemical parameters of serum and liver related to lipid metabolism in HFD-fed mice. Histological analysis also showed that the excessive accumulation of lipid droplets in the liver induced by HFD-feeding was greatly alleviated by GA intervention. In addition, GA intervention also increased the level of short chain fatty acids (SCFAs) in the intestine and promoted the excretion of bile acids (BAs) through feces. High-throughput sequencing of bacterial full-length 16S rDNA revealed that daily supplementation with GA made significant structural changes in the gut microbial population of mice fed with HFD, in particular modulating the relative abundance of some function related microbial phylotypes. The relationships between lipid metabolic parameters and gut microbial phylotypes were also revealed by correlation analysis based on a heatmap and network. The result showed that 46 key gut microbial phylotypes (OTUs) were markedly correlated with at least one lipid metabolic parameter. Moreover, UPLC-QTOF/MS-based liver metabolomics showed that 111 biomarkers (47 up-regulated metabolites and 64 down-regulated metabolites) were significantly changed after high-dose GA intervention (75 mg kg-1 day-1), compared with the HFD-fed hyperlipidemic mice. Metabolic pathway enrichment analysis of the differential hepatic metabolites demonstrated that GA intervention had significant regulatory effects on primary bile acid biosynthesis, fatty acid biosynthesis, amino sugar and nucleotide sugar metabolism, inositol phosphate metabolism, and so on. In addition, GA intervention regulated the mRNA levels of hepatic genes involved in fatty acid metabolism and bile acid homeostasis. These findings present new evidence supporting that GA from G. lucidum has the potential to alleviate lipid metabolic disorders and ameliorate the imbalance of gut microflora in a positive way.
Collapse
Affiliation(s)
- Wei-Ling Guo
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou, Fujian 350108, P. R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Herrscher C, Roingeard P, Blanchard E. Hepatitis B Virus Entry into Cells. Cells 2020; 9:cells9061486. [PMID: 32570893 PMCID: PMC7349259 DOI: 10.3390/cells9061486] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatitis B virus (HBV), an enveloped partially double-stranded DNA virus, is a widespread human pathogen responsible for more than 250 million chronic infections worldwide. Current therapeutic strategies cannot eradicate HBV due to the persistence of the viral genome in a special DNA structure (covalently closed circular DNA, cccDNA). The identification of sodium taurocholate co-transporting polypeptide (NTCP) as an entry receptor for both HBV and its satellite virus hepatitis delta virus (HDV) has led to great advances in our understanding of the life cycle of HBV, including the early steps of infection in particular. However, the mechanisms of HBV internalization and the host factors involved in this uptake remain unclear. Improvements in our understanding of HBV entry would facilitate the design of new therapeutic approaches targeting this stage and preventing the de novo infection of naïve hepatocytes. In this review, we provide an overview of current knowledge about the process of HBV internalization into cells.
Collapse
Affiliation(s)
- Charline Herrscher
- Inserm U1259, Morphogénèse et Antigénicité du VIH et des Virus des Hépatites (MAVIVH), Université de Tours and CHRU de Tours, 37032 Tours, France;
| | - Philippe Roingeard
- Inserm U1259, Morphogénèse et Antigénicité du VIH et des Virus des Hépatites (MAVIVH), Université de Tours and CHRU de Tours, 37032 Tours, France;
- Plate-Forme IBiSA des Microscopies, PPF ASB, Université de Tours and CHRU de Tours, 37032 Tours, France
- Correspondence: (P.R.); (E.B.); Tel.: +33-2-3437-9646 (E.B.)
| | - Emmanuelle Blanchard
- Inserm U1259, Morphogénèse et Antigénicité du VIH et des Virus des Hépatites (MAVIVH), Université de Tours and CHRU de Tours, 37032 Tours, France;
- Plate-Forme IBiSA des Microscopies, PPF ASB, Université de Tours and CHRU de Tours, 37032 Tours, France
- Correspondence: (P.R.); (E.B.); Tel.: +33-2-3437-9646 (E.B.)
| |
Collapse
|
62
|
Lin R, Zhan M, Yang L, Wang H, Shen H, Huang S, Huang X, Xu S, Zhang Z, Li W, Liu Q, Shi Y, Chen W, Yu J, Wang J. Deoxycholic acid modulates the progression of gallbladder cancer through N 6-methyladenosine-dependent microRNA maturation. Oncogene 2020; 39:4983-5000. [PMID: 32514152 PMCID: PMC7314665 DOI: 10.1038/s41388-020-1349-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 02/07/2023]
Abstract
Bile acids (BAs), well-defined signaling molecules with diverse metabolic functions, play important roles in cellular processes associated with many cancers. As one of the most common BAs, deoxycholic acid (DCA) is originally synthesized in the liver, stored in the gallbladder, and processed in the gut. DCA plays crucial roles in various tumors; however, functions and molecular mechanisms of DCA in gallbladder cancer (GBC) still remain poorly characterized. Here, we analyzed human GBC samples and found that DCA was significantly downregulated in GBC, and reduced levels of DCA was associated with poor clinical outcome in patients with GBC. DCA treatment impeded tumor progression by halting cell proliferation. DCA decreased miR-92b-3p expression in an m6A-dependent posttranscriptional modification manner by facilitating dissociation of METTL3 from METTL3-METTL14-WTAP complex, which increased the protein level of the phosphatase and tensin homolog, a newly identified target of miR-92b-3p, and subsequently inactivated the PI3K/AKT signaling pathway. Our findings revealed that DCA might function as a tumor suppressive factor in GBC at least by interfering with miR-92b-3p maturation, and suggested that DCA treatment could provide a new therapeutic strategy for GBC.
Collapse
Affiliation(s)
- Ruirong Lin
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Ming Zhan
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Linhua Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hui Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hui Shen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shuai Huang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xince Huang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sunwang Xu
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zijie Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Weijian Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qiang Liu
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yongsheng Shi
- Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Chen
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jianxiu Yu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Basic Clinical Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Jian Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Basic Clinical Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
63
|
Xiao Q, Zhang S, Ren H, Du R, Li J, Zhao J, Gao Y, Zhu Y, Huang W. Ginsenoside Rg1 alleviates ANIT-induced intrahepatic cholestasis in rats via activating farnesoid X receptor and regulating transporters and metabolic enzymes. Chem Biol Interact 2020; 324:109062. [PMID: 32198087 DOI: 10.1016/j.cbi.2020.109062] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 12/12/2022]
Abstract
Ginsenoside Rg1 is an active ingredient extracted from the roots of ginsenoside, and an α-naphthylisothiocyanate (ANIT)-induced rat model of intrahepatic cholestasis was used to investigate the protective effect of Rg1 on cholestasis. 48 SD male rats were randomly divided into 6 groups: control group, model group, UDCA group (ursodeoxycholic acid), low-dose Rg1 group (10 mg/kg), medium-dose Rg1 group (20 mg/kg) and high-dose Rg1 group (40 mg/kg). The model group, the UDCA group and all the Rg1 group were then intragastrically administered with 80 mg/kg ANIT, and the control group were given equal volume of olive oil. Then the pathological changes in liver tissue were observed, the secretion of bile in the bile duct was measured, and the biochemical markers in serum were quantified, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), glutamyl transfer peptidase (GTP) and the content of total bilirubin (TBIL), direct bilirubin (DBIL), total bile acid (TBA). The contents of inflammatory mediators in serum were quantified, including tumor necrosis factor (TNF-α), γ-interferon (IFN-γ) and interleukin-1β (IL-1β). The contents of superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) in liver homogenate were quantified. Expression of farnesoid X receptor (FXR), transporters and metabolic enzymes in liver tissue was monitored. Rg1 treatment improved liver tissue pathological damage, promoted bile secretion and significantly reduced serum levels of the intrahepatic cholestasis markers ALT, AST, ALP, GTP, TBIL, DBIL and TBA. Rg1 increased the activity of SOD and GSH-Px in liver homogenate, while, reducing the serum levels of MDA and inflammatory mediators. Rg1 also regulated the expression of FXR, bile acid transporters and metabolic enzymes. Overall, Rg1 alleviated liver injury by improving secretion of bile and normalizing the activity of enzymes in the serum. The protective mechanism appeared to be related to the activation of FXR and regulation of liver transporters and metabolic enzymes.
Collapse
Affiliation(s)
- Qing Xiao
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shujun Zhang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huina Ren
- Department of General Medicine, People's Hospital of Chongqing Bishan District, Chongqing, China
| | - Ruoyang Du
- Department of Urology, Chongqing Emergency Medical Center, Chongqing, China
| | - Jiajun Li
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinqiu Zhao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Gao
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yali Zhu
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wenxiang Huang
- Chongqing Key Laboratory of Infectious Diseases and Parasitic Diseases, Department of Infectious Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
64
|
Noppes S, Müller SF, Bennien J, Holtemeyer M, Palatini M, Leidolf R, Alber J, Geyer J. Homo- and heterodimerization is a common feature of the solute carrier family SLC10 members. Biol Chem 2020; 400:1371-1384. [PMID: 31256060 DOI: 10.1515/hsz-2019-0148] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022]
Abstract
The solute carrier family SLC10 consists of seven members, including the bile acid transporters Na+/taurocholate co-transporting polypeptide (NTCP) and apical sodium-dependent bile acid transporter (ASBT), the steroid sulfate transporter SOAT as well as four orphan carriers (SLC10A3, SLC10A4, SLC10A5 and SLC10A7). Previously, homodimerization of NTCP, ASBT and SOAT was described and there is increasing evidence that carrier oligomerization is an important regulatory factor for protein sorting and transport function. In the present study, homo- and heterodimerization were systematically analyzed among all SLC10 carriers (except for SLC10A3) using the yeast-two-hybrid membrane protein system. Strong homodimerization occurred for NTCP/NTCP, ASBT/ASBT and SLC10A7/SLC10A7. Heterodimerization was observed for most of the SLC10 carrier combinations. Heterodimerization of NTCP was additionally investigated by co-localization of NTCP-GFP and NTCP-mScarlet with respective SLC10 carrier constructs. NTCP co-localized with SLC10A4, SLC10A5, SOAT and SLC10A7. This co-localization was most pronounced for SLC10A4 and was additionally confirmed by co-immunoprecipitation. Interestingly, SLC10 carrier co-expression decreased the taurocholate transport function of NTCP for most of the analyzed constructs, indicating that SLC10 carrier heterodimerization is of functional relevance. In conclusion, homo- and heterodimerization is a common feature of the SLC10 carriers. The relevance of this finding for regulation and transport function of the SLC10 carriers in vivo needs further investigation.
Collapse
Affiliation(s)
- Saskia Noppes
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Simon Franz Müller
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Josefine Bennien
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Matthias Holtemeyer
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Massimo Palatini
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Regina Leidolf
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Jörg Alber
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Schubertstr. 81, D-35392 Giessen, Germany
| |
Collapse
|
65
|
Abbas Z, Saad M, Asim M, Abbas M, Samejo SA. The effect of twelve weeks of treatment with ezetimibe on HDV RNA level in patients with chronic hepatitis D. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2020; 31:136-141. [PMID: 32141822 PMCID: PMC7062136 DOI: 10.5152/tjg.2020.18846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/29/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND/AIMS Sodium taurocholate co-transporting polypeptide (NTCP) is the receptor for the hepatitis B virus (HBV) and hepatitis D virus (HDV) entry into hepatocytes. Ezetimibe is a cholesterol-lowering drug that possesses the pharmacophore features to inhibit NTCP. This study evaluates the efficacy of ezetimibe in patients with chronic HDV infection in a nonrandomized trial. MATERIALS AND METHODS This proof of concept phase 2 trial evaluated the efficacy and safety of ezetimibe 10 mg daily in (interferon treatment-experienced or interferon ineligible) patients with chronic hepatitis D (CHD). Forty-four patients with CHD were recruited, 38 male and 6 female patients, mean age 35.2±8.7 (range 19-64). Fifteen (34%) patients were on concomitant nucleoside therapy, and cirrhosis was present in 14 subjects. The primary therapeutic endpoint was a decline in HDV RNA at one log or more from the baseline at week 12. RESULTS The mean HDV RNA level was 5.4±1.3 log10 IU/mL. HBeAg was non-reactive in 43 (98%). HBV DNA was undetectable in 28 (64%). One patient stopped treatment at week 4, and one patient did not follow-up. One log or more reduction in the HDV RNA levels was observed in 18/44 (41%) patients. No log reduction occurred in 16 patients, and 8 experienced a log increase. No adverse effects from the concomitant nucleoside analogue use or clinical cirrhosis were observed. The drug exhibited a positive safety profile. CONCLUSION Treatment of CHD patients with ezetimibe resulted in a one log reduction of viral load in 43% (18/42) of the patients who completed the 12 weeks of therapy.
Collapse
Affiliation(s)
- Zaigham Abbas
- Department of Hepato-Gastroenterology, Dr. Ziauddin University Hospital Clifton, Karachi, Pakistan
| | - Muhammad Saad
- Department of Hepato-Gastroenterology, Dr. Ziauddin University Hospital Clifton, Karachi, Pakistan
| | - Muhammad Asim
- Department of Hepato-Gastroenterology, Dr. Ziauddin University Hospital Clifton, Karachi, Pakistan
| | - Minaam Abbas
- Department of Hepato-Gastroenterology, Dr. Ziauddin University Hospital Clifton, Karachi, Pakistan
| | - Shoukat Ali Samejo
- Department of Hepato-Gastroenterology, Dr. Ziauddin University Hospital Clifton, Karachi, Pakistan
| |
Collapse
|
66
|
Iwamoto M, Saso W, Nishioka K, Ohashi H, Sugiyama R, Ryo A, Ohki M, Yun JH, Park SY, Ohshima T, Suzuki R, Aizaki H, Muramatsu M, Matano T, Iwami S, Sureau C, Wakita T, Watashi K. The machinery for endocytosis of epidermal growth factor receptor coordinates the transport of incoming hepatitis B virus to the endosomal network. J Biol Chem 2020. [DOI: 10.1016/s0021-9258(17)49936-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
67
|
Shuai T, Zhou Y, Shao G, Yang R, Wang L, Wang J, Sun J, Ren L, Wang J, Liao Y, Wei M, Xu Q, Li Y, Zhao L. Bimodal Molecule as NIR-CT Contrast Agent for Hepatoma Specific Imaging. Anal Chem 2019; 92:1138-1146. [PMID: 31820637 DOI: 10.1021/acs.analchem.9b04212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
With currently available molecular imaging techniques, hepatocellular carcinoma (HCC), a liver cancer with high mortality rates and poor treatment responses, is mostly diagnosed at its late stage. This is largely due to the lack of highly sensitive contrast agents with high liver specificity. Herein, we report a novel bimodal contrast agent molecule CNCI-1 for the effective detection of HCC at its early stage both in vitro and in vivo. The agent has high liver specificity with effective X-ray computed tomography (CT)/near-infrared (NIR) imaging functions. It has been successfully applied to in vivo NIR imaging with high sensitivity and high selectivity to the HCC region of the HepG2 tumor-xenografted mice model and LM3 orthotopic hepatoma mice model. Moreover, the agent was found to be noninvasive and hepatocarcinoma cells preferential. Furthermore, it also enhanced the tumor imaging by revealing the blood vessels nearby for the CT image acquisition in the VX2 orthotopic hepatoma rabbit model. Our design strategy provides a new avenue to develop the medical relevant bimodal contrast agents for diagnosis of HCC at its early stage.
Collapse
Affiliation(s)
- Tianbai Shuai
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211100 , China
| | - Yizhou Zhou
- School of Basic Medicine and Clinical Pharmacology , China Pharmaceutical University , Nanjing 211100 , China
| | - Guoqiang Shao
- Department of Nuclear Medicine , Nanjing First Hospital, Affiliated to Nanjing Medical University , Nanjing 210006 , China
| | - Rui Yang
- Department of Nuclear Medicine , Nanjing First Hospital, Affiliated to Nanjing Medical University , Nanjing 210006 , China
| | - Letian Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211100 , China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery , Nanjing Drum Tower Hospital, Affiliated to Medical College of Nanjing University , Nanjing 210008 , China
| | - Jie Sun
- School of Basic Medicine and Clinical Pharmacology , China Pharmaceutical University , Nanjing 211100 , China
| | - Longfei Ren
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211100 , China
| | - Jintao Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211100 , China
| | - Yan Liao
- School of Basic Medicine and Clinical Pharmacology , China Pharmaceutical University , Nanjing 211100 , China
| | - Mian Wei
- School of Basic Medicine and Clinical Pharmacology , China Pharmaceutical University , Nanjing 211100 , China
| | - Qingxiang Xu
- Department of Hepatobiliary Surgery , Nanjing Drum Tower Hospital, Affiliated to Medical College of Nanjing University , Nanjing 210008 , China
| | - Yuyan Li
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry , China Pharmaceutical University , Nanjing 211100 , China
| | - Li Zhao
- School of Basic Medicine and Clinical Pharmacology , China Pharmaceutical University , Nanjing 211100 , China
| |
Collapse
|
68
|
Iwamoto M, Saso W, Nishioka K, Ohashi H, Sugiyama R, Ryo A, Ohki M, Yun JH, Park SY, Ohshima T, Suzuki R, Aizaki H, Muramatsu M, Matano T, Iwami S, Sureau C, Wakita T, Watashi K. The machinery for endocytosis of epidermal growth factor receptor coordinates the transport of incoming hepatitis B virus to the endosomal network. J Biol Chem 2019; 295:800-807. [PMID: 31836663 DOI: 10.1074/jbc.ac119.010366] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 12/10/2019] [Indexed: 12/27/2022] Open
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is expressed at the surface of human hepatocytes and functions as an entry receptor of hepatitis B virus (HBV). Recently, we have reported that epidermal growth factor receptor (EGFR) is involved in NTCP-mediated viral internalization during the cell entry process. Here, we analyzed which function of EGFR is essential for mediating HBV internalization. In contrast to the reported crucial function of EGFR-downstream signaling for the entry of hepatitis C virus (HCV), blockade of EGFR-downstream signaling proteins, including mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K), and signal transducer and activator of transcription (STAT), had no or only minor effects on HBV infection. Instead, deficiency of EGFR endocytosis resulting from either a deleterious mutation in EGFR or genetic knockdown of endocytosis adaptor molecules abrogated internalization of HBV via NTCP and prevented viral infection. EGFR activation triggered a time-dependent relocalization of HBV preS1 to the early and late endosomes and to lysosomes in concert with EGFR transport. Suppression of EGFR ubiquitination by site-directed mutagenesis or by knocking down two EGFR-sorting molecules, signal-transducing adaptor molecule (STAM) and lysosomal protein transmembrane 4β (LAPTM4B), suggested that EGFR transport to the late endosome is critical for efficient HBV infection. Cumulatively, these results support the idea that the EGFR endocytosis/sorting machinery drives the translocation of NTCP-bound HBV from the cell surface to the endosomal network, which eventually enables productive viral infection.
Collapse
Affiliation(s)
- Masashi Iwamoto
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,Mathematical Biology Laboratory, Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan
| | - Wakana Saso
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Kazane Nishioka
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,Department of Applied Biological Science, Tokyo University of Science, Noda 278-8510, Japan
| | - Hirofumi Ohashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,Department of Applied Biological Science, Tokyo University of Science, Noda 278-8510, Japan
| | - Ryuichi Sugiyama
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama 236-0004, Japan
| | - Mio Ohki
- Drug Design Laboratory, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Ji-Hye Yun
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, South Korea
| | - Sam-Yong Park
- Drug Design Laboratory, Yokohama City University Graduate School of Medical Life Science, Yokohama 230-0045, Japan
| | - Takayuki Ohshima
- Faculty of Pharmaceutical Science at Kagawa Campus, Tokushima Bunri University, Kagawa 769-2193, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.,The Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Shingo Iwami
- Mathematical Biology Laboratory, Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan.,Core Research for Evolutional Science and Technology (CREST) Japan Science and Technology Agency (JST), Saitama 332-0012, Japan.,MIRAI, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, Paris 75739, France
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan .,Department of Applied Biological Science, Tokyo University of Science, Noda 278-8510, Japan.,Core Research for Evolutional Science and Technology (CREST) Japan Science and Technology Agency (JST), Saitama 332-0012, Japan.,MIRAI, Japan Science and Technology Agency (JST), Saitama 332-0012, Japan.,Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
69
|
Xiao Y, Liu C, Tang W, Zhang H, Chen X. Evans Blue Inhibits HBV Replication Through a Dual Antiviral Mechanism by Targeting Virus Binding and Capsid Assembly. Front Microbiol 2019; 10:2638. [PMID: 31798562 PMCID: PMC6868041 DOI: 10.3389/fmicb.2019.02638] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022] Open
Abstract
Chronic hepatitis B (CHB) is a global health problem caused by human hepatitis B virus (HBV). Current treatment with interferons and nucleos(t)ide analogs (NAs) can cause population tolerance and drug resistance. Therefore, new antiviral drugs, especially those targeting host factors, are urgently needed. Here, we identified Evans blue as a new HBV inhibitor by screening an FDA drug library using Huh7DhNTCP cells and confirmed the antiviral activity in primary human hepatocytes and human sodium taurocholate cotransporting polypeptide (hNTCP)-transfected porcine primary hepatocytes. Our efficacy study showed that Evans blue has an IC50 of 2 μM against HBV infection in Huh7DhNTCP cells, and no apparent toxicity at up to 1000 μM. The IC50 of Evans blue against HBV in primary human hepatocytes was approximately 5 μM. Mechanism studies revealed that Evans blue has a dual anti-HBV effect. It inhibits both the binding of viral preS1 to host cells through the host factor NTCP and the virus capsid assembly by targeting the host factor BK channel. The KD of the direct interaction between Evans blue and NTCP is 8.82E-8 M. Evans blue can suppress capsid assembly at micromolar concentrations by reducing the cytosolic calcium ion concentration. Since the antiviral effects on HBV binding and assembly are both achieved through targeting host factors, Evans blue inhibits the infection of nucleos(t)ide analog drug-resistant HBV strains in Huh7DhNTCP cells. Taken together, our results suggest that Evans blue may be a promising anti-HBV drug candidate in the classes of both entry and assembly inhibitors.
Collapse
Affiliation(s)
- Yu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunlan Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Wei Tang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Haiwei Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xulin Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,Guangdong Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
70
|
Goh B, Choi J, Kang JA, Park SG, Seo J, Kim TY. Development of a mass spectrometric screening assay for hepatitis B virus entry inhibitors. J Pharm Biomed Anal 2019; 178:112959. [PMID: 31722821 DOI: 10.1016/j.jpba.2019.112959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/24/2019] [Accepted: 10/27/2019] [Indexed: 12/24/2022]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) involved in bile acid transport in the liver is an entry receptor of hepatitis B virus (HBV). In the present study, we introduce a mass spectrometric screening assay for targeting HBV entry inhibitors that can reduce NTCP transporter activity by employing taurocholic acid (TCA) labeled with stable isotope (2,2,4,4-d4-TCA, d4-TCA) and NTCP-overexpressing human liver cancer cell lines such as HepG2 and Huh-7. The accuracy and reliability of the proposed mass spectrometric NTCP activity assay have been validated with known HBV inhibitors including cyclosporine A (CsA) and pre-S1 peptide (PreS/2-48Myr or myrcludex B analog) that suppress the entry of HBV into hepatocytes by targeting NTCP. For the inhibitor screening assay, NTCP-overexpressing HepG2 or Huh-7 cells are treated with either a combination of TCA and an inhibitor (CsA or PreS/2-48Myr) or d4-TCA alone to serve as a reference. The activity of an HBV inhibitor is determined by relative quantification between TCA and d4-TCA in a 1:1 mixture of inhibitor-treated cells and untreated control cells using liquid chromatography-mass spectrometry. With our new approach, the half maximal inhibitory concentration (IC50) values for CsA and PreS/2-48Myr have been determined at micromolar and nanomolar concentrations, respectively, which is consistent with the previous results obtained with other conventional HBV entry inhibitor assay methods. Our assay method does not require HBV infection or radioactive 3H-TCA and provides a facile way to identify viral entry inhibitors via measuring bile acid transport activity of NTCP.
Collapse
Affiliation(s)
- Byoungsook Goh
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jieun Choi
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jung-Ah Kang
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea
| | - Jiwon Seo
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| | - Tae-Young Kim
- Department of Chemistry, School of Physics and Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea; School of Earth Sciences and Environmental Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, South Korea.
| |
Collapse
|
71
|
Sa-Ngiamsuntorn K, Thongsri P, Pewkliang Y, Wongkajornsilp A, Kongsomboonchoke P, Suthivanich P, Borwornpinyo S, Hongeng S. An Immortalized Hepatocyte-like Cell Line (imHC) Accommodated Complete Viral Lifecycle, Viral Persistence Form, cccDNA and Eventual Spreading of a Clinically-Isolated HBV. Viruses 2019; 11:E952. [PMID: 31623162 PMCID: PMC6832882 DOI: 10.3390/v11100952] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
More than 350 million people worldwide have been persistently infected with the hepatitis B virus (HBV). Chronic HBV infection could advance toward liver cirrhosis and hepatocellular carcinoma. The intervention with prophylactic vaccine and conventional treatment could suppress HBV, but could not completely eradicate it. The major obstacle for investigating curative antiviral drugs are the incompetence of hepatocyte models that should have closely imitated natural human infection. Here, we demonstrated that an immortalized hepatocyte-like cell line (imHC) could accommodate for over 30 days the entire life cycle of HBV prepared from either established cultured cells or clinically-derived fresh isolates. Normally, imHCs had intact interferon signaling with anti-viral action. Infected imHCs responded to treatments with direct-acting antiviral drugs (DAAs) and interferons (IFNs) by diminishing HBV DNA, the covalently closed circular DNA (cccDNA) surface antigen of HBV (HBsAg, aka the Australia antigen) and the hepatitis B viral protein (HBeAg). Notably, we could observe and quantify HBV spreading from infected cells to naïve cells using an imHC co-culture model. In summary, this study constructed a convenient HBV culture model that allows the screening for novel anti-HBV agents with versatile targets, either HBV entry, replication or cccDNA formation. Combinations of agents aiming at different targets should achieve a complete HBV eradication.
Collapse
Affiliation(s)
- Khanit Sa-Ngiamsuntorn
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Piyanoot Thongsri
- Department of Biochemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand.
| | - Yongyut Pewkliang
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Adisak Wongkajornsilp
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| | | | - Phichaya Suthivanich
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Suradej Hongeng
- Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
72
|
Yan Y, Allweiss L, Yang D, Kang J, Wang J, Qian X, Zhang T, Liu H, Wang L, Liu S, Sui J, Chen X, Dandri M, Zhao J, Lu F. Down-regulation of cell membrane localized NTCP expression in proliferating hepatocytes prevents hepatitis B virus infection. Emerg Microbes Infect 2019; 8:879-894. [PMID: 31179847 PMCID: PMC6567113 DOI: 10.1080/22221751.2019.1625728] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocyte proliferation could result in the loss of covalently closed circular DNA (cccDNA) and the emergence of cccDNA-cleared nascent hepatocytes, which appear refractory to hepatitis B virus (HBV) reinfection with unknown mechanism(s). Sodium taurocholate cotransporting polypeptide (NTCP) is the functional receptor for HBV entry. In this study, down-regulation of cell membrane localized NTCP expression in proliferating hepatocytes was found to prevent HBV infection in HepG2-NTCP-tet cells and in liver-humanized mice. In patients, lower NTCP protein expression was correlated well with higher levels of hepatocyte proliferation and less HBsAg expression in HBV-related focal nodular hyperplasia (FNH) tissues. Clinically, significantly lower NTCP protein expression was correlated with more active hepatocyte proliferation in CHB patients with severe active necroinflammation and better antiviral treatment outcome. Mechanistically, the activation of cell cycle regulatory genes p53, S-phase kinase-associated protein 2 (SKP2) and cyclin D1 during cell proliferation, as well as proliferative and inflammatory cytokine Interleukin-6 (IL-6) could transcriptionally down-regulate NTCP expression. From these aspects, we conclude that within the milieu of hepatocyte proliferation, down-regulation of cell membrane localized NTCP expression level renders nascent hepatocytes resistant to HBV reinfection. This may accelerate virus clearance during immune-mediated cell death and compensatory proliferation of survival hepatocytes.
Collapse
Affiliation(s)
- Ying Yan
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Lena Allweiss
- b Department of Medicine, Center for Internal Medicine , University Medical Center Hamburg-Eppendorf , Hamburg , Germany
| | - Danli Yang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Jingting Kang
- c Institute of Basic Medical Sciences Chinese Academy of Medical Sciences , School of Basic Medicine Peking Union Medical College , Beijing , People's Republic of China
| | - Jianwen Wang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Xiangjun Qian
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Ting Zhang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Hui Liu
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Lu Wang
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Shuhong Liu
- d Department of Pathology and Hepatology , The 5th Medical Centre, Chinese PLA General Hospital , Beijing , People's Republic of China
| | - Jianhua Sui
- e Biologics Research Center , National Institute of Biological Sciences , Beijing , People's Republic of China
| | - Xiangmei Chen
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| | - Maura Dandri
- b Department of Medicine, Center for Internal Medicine , University Medical Center Hamburg-Eppendorf , Hamburg , Germany.,f German Center for Infection Research (DZIF) , Hamburg-Lübeck-Borstel-Riems Partner Site , Hamburg , Germany
| | - Jingmin Zhao
- d Department of Pathology and Hepatology , The 5th Medical Centre, Chinese PLA General Hospital , Beijing , People's Republic of China
| | - Fengmin Lu
- a State Key Laboratory of Natural and Biomimetic Drugs, Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences , Peking University Health Science Center , Beijing , People's Republic of China
| |
Collapse
|
73
|
König A, Yang J, Jo E, Park KHP, Kim H, Than TT, Song X, Qi X, Dai X, Park S, Shum D, Ryu WS, Kim JH, Yoon SK, Park JY, Ahn SH, Han KH, Gerlich WH, Windisch MP. Efficient long-term amplification of hepatitis B virus isolates after infection of slow proliferating HepG2-NTCP cells. J Hepatol 2019; 71:289-300. [PMID: 31077792 DOI: 10.1016/j.jhep.2019.04.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS As hepatitis B virus (HBV) spreads through the infected liver it is simultaneously secreted into the blood. HBV-susceptible in vitro infection models do not efficiently amplify viral progeny or support cell-to-cell spread. We sought to establish a cell culture system for the amplification of infectious HBV from clinical specimens. METHODS An HBV-susceptible sodium-taurocholate cotransporting polypeptide-overexpressing HepG2 cell clone (HepG2-NTCPsec+) producing high titers of infectious progeny was selected. Secreted HBV progeny were characterized by native gel electrophoresis and electron microscopy. Comparative RNA-seq transcriptomics was performed to quantify the expression of host proviral and restriction factors. Viral spread routes were evaluated using HBV entry- or replication inhibitors, visualization of viral cell-to-cell spread in reporter cells, and nearest neighbor infection determination. Amplification kinetics of HBV genotypes B-D were analyzed. RESULTS Infected HepG2-NTCPsec+ secreted high levels of large HBV surface protein-enveloped infectious HBV progeny with typical appearance under electron microscopy. RNA-seq transcriptomics revealed that HBV does not induce significant gene expression changes in HepG2-NTCPsec+, however, transcription factors favoring HBV amplification were more strongly expressed than in less permissive HepG2-NTCPsec-. Upon inoculation with HBV-containing patient sera, rates of infected cells increased from 10% initially to 70% by viral spread to adjacent cells, and viral progeny and antigens were efficiently secreted. HepG2-NTCPsec+ supported up to 1,300-fold net amplification of HBV genomes depending on the source of virus. Viral spread and amplification were abolished by entry and replication inhibitors; viral rebound was observed after inhibitor discontinuation. CONCLUSIONS The novel HepG2-NTCPsec+ cells efficiently support the complete HBV life cycle, long-term viral spread and amplification of HBV derived from patients or cell culture, resembling relevant features of HBV-infected patients. LAY SUMMARY Currently available laboratory systems are unable to reproduce the dynamics of hepatitis B virus (HBV) spread through the infected liver and release into the blood. We developed a slowly dividing liver-derived cell line which multiplies infectious viral particles upon inoculation with patient- or cell culture-derived HBV. This new infection model can improve therapy by measuring, in advance, the sensitivity of a patient's HBV strain to specific antiviral drugs.
Collapse
Affiliation(s)
- Alexander König
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Jaewon Yang
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Eunji Jo
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Kyu Ho Paul Park
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Hyun Kim
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea; Division of Bio-Medical Science and Technology, University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon, South Korea
| | - Thoa Thi Than
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Xiyong Song
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Xiaoxuan Qi
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Xinghong Dai
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Soonju Park
- Screening Discovery Platform, Institut Pasteur Korea, Seongnam-si, South Korea
| | - David Shum
- Screening Discovery Platform, Institut Pasteur Korea, Seongnam-si, South Korea
| | - Wang-Shick Ryu
- Department of Biochemistry, Yonsei University, Seoul, South Korea
| | - Jung-Hee Kim
- Catholic University Liver Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Seung Kew Yoon
- Catholic University Liver Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Jun Yong Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Marc Peter Windisch
- Applied Molecular Virology Laboratory, Institut Pasteur Korea, Seongnam-si, South Korea; Division of Bio-Medical Science and Technology, University of Science and Technology, 217, Gajeong-ro, Yuseong-gu, Daejeon, South Korea.
| |
Collapse
|
74
|
Bi YA, Costales C, Mathialagan S, West M, Eatemadpour S, Lazzaro S, Tylaska L, Scialis R, Zhang H, Umland J, Kimoto E, Tess DA, Feng B, Tremaine LM, Varma MVS, Rodrigues AD. Quantitative Contribution of Six Major Transporters to the Hepatic Uptake of Drugs: "SLC-Phenotyping" Using Primary Human Hepatocytes. J Pharmacol Exp Ther 2019; 370:72-83. [PMID: 30975793 DOI: 10.1124/jpet.119.257600] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 04/08/2019] [Indexed: 11/22/2022] Open
Abstract
Hepatic uptake transporters [solute carriers (SLCs)], including organic anion transporting polypeptide (OATP) 1B1, OATP1B3, OATP2B1, sodium-dependent taurocholate cotransporting polypeptide (NTCP), and organic anion (OAT2) and organic cation (OCT1) transporters, play a key role in determining the systemic and liver exposure of chemically diverse drugs. Here, we established a phenotyping approach to quantify the contribution of the six SLCs, and passive diffusion, to the overall uptake using plated human hepatocytes (PHHs). First, selective inhibitor conditions were identified by screening about 20 inhibitors across the six SLCs using single-transfected human embryonic kidney 293 cells. Data implied rifamycin SV (20 µM) inhibits three OATPs, while rifampicin (5 µM) inhibits OATP1B1/1B3 only. Further, hepatitis B virus myristoylated-preS1 peptide (0.1 µM), quinidine (100 µM), and ketoprofen (100-300 µM) are relatively selective against NTCP, OCT1, and OAT2, respectively. Second, using these inhibitory conditions, the fraction transported (ft ) by the individual SLCs was characterized for 20 substrates with PHH. Generally, extended clearance classification system class 1A/3A (e.g., warfarin) and 1B/3B compounds (e.g., statins) showed predominant OAT2 and OATP1B1/1B3 contribution, respectively. OCT1-mediated uptake was prominent for class 2/4 compounds (e.g., metformin). Third, in vitro ft values were corrected using quantitative proteomics data to obtain "scaled ft " Fourth, in vitro-in vivo extrapolation of the scaled OATP1B1/1B3 ft was assessed, leveraging statin clinical drug-drug interaction data with rifampicin as the perpetrator. Finally, we outlined a novel stepwise strategy to implement phenotypic characterization of SLC-mediated hepatic uptake for new molecular entities and drugs in a drug discovery and development setting.
Collapse
Affiliation(s)
- Yi-An Bi
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Chester Costales
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Mark West
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Soraya Eatemadpour
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sarah Lazzaro
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Laurie Tylaska
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Renato Scialis
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Hui Zhang
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - John Umland
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Emi Kimoto
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - David A Tess
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Bo Feng
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Larry M Tremaine
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Manthena V S Varma
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - A David Rodrigues
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
75
|
Wu MR, Huang YY, Hsiao JK. Use of Indocyanine Green (ICG), a Medical Near Infrared Dye, for Enhanced Fluorescent Imaging-Comparison of Organic Anion Transporting Polypeptide 1B3 (OATP1B3) and Sodium-Taurocholate Cotransporting Polypeptide (NTCP) Reporter Genes. Molecules 2019; 24:molecules24122295. [PMID: 31234288 PMCID: PMC6630416 DOI: 10.3390/molecules24122295] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 12/13/2022] Open
Abstract
Molecular and cellular imaging in living organisms have ushered in an era of comprehensive understanding of intracellular and intercellular events. Currently, more efforts have been focused on the infrared fluorescent dyes that facilitate deeper tissue visualization. Both sodium taurocholate cotransporting polypeptide (NTCP) and organic-anion-transporting polypeptide 1B3 (OATP1B3) are capable of carrying indocyanine green (ICG) into the cytoplasm. We compared the feasibility of NTCP and OATP1B3 as reporter genes in combination with ICG. NTCP and OATP1B3 were transduced into HT-29 cells. Genetically modified HT-29 cells were inoculated into nude mice. ICG was administered in vitro and in vivo and the signals were observed under confocal microscopy, flow cytometry, multimode microplate reader, and an in vivo imaging system. Both NTCP- and OATP1B3-expressing cells and xenografts had higher ICG intensities. The OATP1B3-expressing xenograft has a higher ICG uptake than the NTCP-expressing xenograft. NTCP or OATP1B3 combined with ICG could serve as a noninvasive imaging modality for molecular and cellular imaging. OATP1B3 outperforms NTCP in terms of in vivo imaging.
Collapse
Affiliation(s)
- Menq-Rong Wu
- Institute of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan.
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, No.289, Jianguo Rd., Xindian Dist., New Taipei city 23142, Taiwan.
| | - Yi-You Huang
- Institute of Biomedical Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei 10617, Taiwan.
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei TzuChi General Hospital, Buddhist Tzu-Chi Medical Foundation, No.289, Jianguo Rd., Xindian Dist., New Taipei city 23142, Taiwan.
- School of Medicine, Tzu Chi University, No. 701, Sec. 3, Zhongyang Rd. Hualien 97004, Taiwan.
| |
Collapse
|
76
|
Kostyusheva AP, Kostyushev DS, Brezgin SA, Zarifyan DN, Volchkova EV, Chulanov VP. Small Molecular Inhibitors of DNA Double Strand Break Repair Pathways Increase the ANTI-HBV Activity of CRISPR/Cas9. Mol Biol 2019. [DOI: 10.1134/s0026893319010072] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
77
|
Iwamoto M, Saso W, Sugiyama R, Ishii K, Ohki M, Nagamori S, Suzuki R, Aizaki H, Ryo A, Yun JH, Park SY, Ohtani N, Muramatsu M, Iwami S, Tanaka Y, Sureau C, Wakita T, Watashi K. Epidermal growth factor receptor is a host-entry cofactor triggering hepatitis B virus internalization. Proc Natl Acad Sci U S A 2019; 116:8487-8492. [PMID: 30952782 PMCID: PMC6486715 DOI: 10.1073/pnas.1811064116] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is a host cell receptor required for hepatitis B virus (HBV) entry. However, the susceptibility of NTCP-expressing cells to HBV is diverse depending on the culture condition. Stimulation with epidermal growth factor (EGF) was found to potentiate cell susceptibility to HBV infection. Here, we show that EGF receptor (EGFR) plays a critical role in HBV virion internalization. In EGFR-knockdown cells, HBV or its preS1-specific fluorescence peptide attached to the cell surface, but its internalization was attenuated. PreS1 internalization and HBV infection could be rescued by complementation with functional EGFR. Interestingly, the HBV/preS1-NTCP complex at the cell surface was internalized concomitant with the endocytotic relocalization of EGFR. Molecular interaction between NTCP and EGFR was documented by immunoprecipitation assay. Upon dissociation from functional EGFR, NTCP no longer functioned to support viral infection, as demonstrated by either (i) the introduction of NTCP point mutation that disrupted its interaction with EGFR, (ii) the detrimental effect of decoy peptide interrupting the NTCP-EGFR interaction, or (iii) the pharmacological inactivation of EGFR. Together, these data support the crucial role of EGFR in mediating HBV-NTCP internalization into susceptible cells. EGFR thus provides a yet unidentified missing link from the cell-surface HBV-NTCP attachment to the viral invasion beyond the host cell membrane.
Collapse
Affiliation(s)
- Masashi Iwamoto
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
- Mathematical Biology Laboratory, Department of Biology, Faculty of Sciences, Kyushu University, 812-8581 Fukuoka, Japan
| | - Wakana Saso
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
- The Institute of Medical Science, The University of Tokyo, 108-8639 Tokyo, Japan
| | - Ryuichi Sugiyama
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Koji Ishii
- Department of Quality Assurance and Radiological Protection, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Mio Ohki
- Drug Design Laboratory, Yokohama City University Graduate School of Medical Life Science, 230-0045 Yokohama, Japan
| | - Shushi Nagamori
- Department of Collaborative Research, Nara Medical University, 634-8521 Kashihara, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, 236-0004 Yokohama, Japan
| | - Ji-Hye Yun
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, 03722 Seoul, South Korea
| | - Sam-Yong Park
- Drug Design Laboratory, Yokohama City University Graduate School of Medical Life Science, 230-0045 Yokohama, Japan
| | - Naoko Ohtani
- Graduate School of Medicine, Osaka City University, 545-8585 Osaka, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Shingo Iwami
- Mathematical Biology Laboratory, Department of Biology, Faculty of Sciences, Kyushu University, 812-8581 Fukuoka, Japan
- Core Research for Evolutional Science and Technology (CORE), Japan Science and Technology Agency (JST), 332-0012 Saitama, Japan
- MIRAI, Japan Science and Technology, 332-0012 Saitama, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medicinal Sciences, 467-8601 Nagoya, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, 75739 Paris, France
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, 162-8640 Tokyo, Japan;
- Core Research for Evolutional Science and Technology (CORE), Japan Science and Technology Agency (JST), 332-0012 Saitama, Japan
- MIRAI, Japan Science and Technology, 332-0012 Saitama, Japan
- Department of Applied Biological Science, Tokyo University of Science, 278-8510 Noda, Japan
| |
Collapse
|
78
|
Fukano K, Watashi K. [From the Establishment of Hepatitis B Virus Cell Culture Systems to Drug Discovery]. YAKUGAKU ZASSHI 2019; 139:81-87. [PMID: 30606935 DOI: 10.1248/yakushi.18-00164-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The development of antiviral agents enables the control of chronic infectious diseases caused by infection with herpesviruses, human immunodeficiency virus, and hepatitis C virus. In contrast, antiviral treatment against hepatitis B virus (HBV) infection remains a significant area for improvement. One of the main barriers hampering the progress of HBV research has been a lack of cell culture systems efficiently reproducing the viral proliferation process. Recently, cell line-based HBV infection systems have been developed which are useful to analyze the mechanisms of HBV replication and to screen for new anti-HBV agents. In this article, we summarize the establishment of such cell models and the identification of small molecules that inhibit the HBV entry process and discuss their future potential as a novel class of anti-HBV agents.
Collapse
Affiliation(s)
- Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases.,Department of Analytical Biochemistry, Meiji Pharmaceutical University
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases
| |
Collapse
|
79
|
Rasche A, Sander AL, Corman VM, Drexler JF. Evolutionary biology of human hepatitis viruses. J Hepatol 2019; 70:501-520. [PMID: 30472320 PMCID: PMC7114834 DOI: 10.1016/j.jhep.2018.11.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/09/2018] [Accepted: 11/10/2018] [Indexed: 02/06/2023]
Abstract
Hepatitis viruses are major threats to human health. During the last decade, highly diverse viruses related to human hepatitis viruses were found in animals other than primates. Herein, we describe both surprising conservation and striking differences of the unique biological properties and infection patterns of human hepatitis viruses and their animal homologues, including transmission routes, liver tropism, oncogenesis, chronicity, pathogenesis and envelopment. We discuss the potential for translation of newly discovered hepatitis viruses into preclinical animal models for drug testing, studies on pathogenesis and vaccine development. Finally, we re-evaluate the evolutionary origins of human hepatitis viruses and discuss the past and present zoonotic potential of their animal homologues.
Collapse
Affiliation(s)
- Andrea Rasche
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany,German Center for Infection Research (DZIF), Germany
| | - Anna-Lena Sander
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany
| | - Victor Max Corman
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany,German Center for Infection Research (DZIF), Germany
| | - Jan Felix Drexler
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, 10117 Berlin, Germany; German Center for Infection Research (DZIF), Germany.
| |
Collapse
|
80
|
Fukano K, Tsukuda S, Oshima M, Suzuki R, Aizaki H, Ohki M, Park SY, Muramatsu M, Wakita T, Sureau C, Ogasawara Y, Watashi K. Troglitazone Impedes the Oligomerization of Sodium Taurocholate Cotransporting Polypeptide and Entry of Hepatitis B Virus Into Hepatocytes. Front Microbiol 2019; 9:3257. [PMID: 30671048 PMCID: PMC6331526 DOI: 10.3389/fmicb.2018.03257] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022] Open
Abstract
Current anti-hepatitis B virus (HBV) agents, which include nucleos(t)ide analogs and interferons, can significantly suppress HBV infection. However, there are limitations in the therapeutic efficacy of these agents, indicating the need to develop anti-HBV agents with different modes of action. In this study, through a functional cell-based chemical screening, we found that a thiazolidinedione, troglitazone, inhibits HBV infection independently of the compound's ligand activity for peroxisome proliferator-activated receptor γ (PPARγ). Analog analysis suggested chemical moiety required for the anti-HBV activity and identified ciglitazone as an analog having higher anti-HBV potency. Whereas, most of the reported HBV entry inhibitors target viral attachment to the cell surface, troglitazone blocked a process subsequent to viral attachment, i.e., internalization of HBV preS1 and its receptor, sodium taurocholate cotransporting polypeptide (NTCP). We also found that NTCP was markedly oligomerized in the presence of HBV preS1, but such NTCP oligomerization was abrogated by treatment with troglitazone, but not with pioglitazone, correlating with inhibition activity to viral internalization. Also, competitive peptides that blocked NTCP oligomerization impeded viral internalization and infection. This work represents the first report identifying small molecules and peptides that specifically inhibit the internalization of HBV. This study is also significant in proposing a possible role for NTCP oligomerization in viral entry, which will shed a light on a new aspect of the cellular mechanisms regulating HBV infection.
Collapse
Affiliation(s)
- Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Senko Tsukuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.,Liver Cancer Prevention Research Unit, Center for Integrative Medical Sciences, RIKEN, Wako, Japan
| | - Mizuki Oshima
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Applied Biological Science, Tokyo University of Science, Noda, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mio Ohki
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, CNRS, INSERM U1134, Paris, France
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Applied Biological Science, Tokyo University of Science, Noda, Japan.,JST CREST, Saitama, Japan
| |
Collapse
|
81
|
Hu J, Lin YY, Chen PJ, Watashi K, Wakita T. Cell and Animal Models for Studying Hepatitis B Virus Infection and Drug Development. Gastroenterology 2019; 156:338-354. [PMID: 30243619 PMCID: PMC6649672 DOI: 10.1053/j.gastro.2018.06.093] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 06/21/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
Abstract
Many cell culture and animal models have been used to study hepatitis B virus (HBV) replication and its effects in the liver; these have facilitated development of strategies to control and clear chronic HBV infection. We discuss the advantages and limitations of systems for studying HBV and developing antiviral agents, along with recent advances. New and improved model systems are needed. Cell culture systems should be convenient, support efficient HBV infection, and reproduce responses of hepatocytes in the human body. We also need animals that are fully permissive to HBV infection, convenient for study, and recapitulate human immune responses to HBV and effects in the liver. High-throughput screening technologies could facilitate drug development based on findings from cell and animal models.
Collapse
Affiliation(s)
- Jianming Hu
- The Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| | - You-Yu Lin
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Jer Chen
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Hepatitis Research Center, National Taiwan University Hospital, National Taiwan University.
| | | | - Takaji Wakita
- National Institute of Infectious Diseases, Tokyo, Japan.
| |
Collapse
|
82
|
Zhao K, Liu S, Chen Y, Yao Y, Zhou M, Yuan Y, Wang Y, Pei R, Chen J, Hu X, Zhou Y, Zhao H, Lu M, Wu C, Chen X. Upregulation of HBV transcription by sodium taurocholate cotransporting polypeptide at the postentry step is inhibited by the entry inhibitor Myrcludex B. Emerg Microbes Infect 2018; 7:186. [PMID: 30459339 PMCID: PMC6246608 DOI: 10.1038/s41426-018-0189-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/30/2018] [Accepted: 10/26/2018] [Indexed: 02/07/2023]
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is a functional receptor for hepatitis B virus (HBV) entry. However, little is known regarding whether NTCP is involved in regulating the postentry steps of the HBV life cycle. Here, we found that NTCP expression upregulated HBV transcription at the postentry step and that the NTCP-targeting entry inhibitor Myrcludex B (MyrB) effectively suppressed HBV transcription both in an HBV in vitro infection system and in mice hydrodynamically injected with an HBV expression plasmid. Mechanistically, NTCP upregulated HBV transcription via farnesoid X receptor α (FxRα)-mediated activation of the HBV EN2/core promoter at the postentry step in a manner that was dependent on the bile acid (BA)-transport function of NTCP, which was blocked by MyrB. Our findings uncover a novel role for NTCP in the HBV life cycle and provide a reference for the use of novel NTCP-targeting entry inhibitors to suppress HBV infection and replication.
Collapse
Affiliation(s)
- Kaitao Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shuhui Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yingshan Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yongxuan Yao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Ming Zhou
- Shenzhen Xenotransplantation Research and Development Center, State and Local Joint Cancer Genome Clinical Application of Key Technology Laboratory, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, 518035, Shenzhen, China
| | - Yifei Yuan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yun Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Rongjuan Pei
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Jizheng Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Xue Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Yuan Zhou
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - He Zhao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China
| | - Mengji Lu
- Institute of Virology, University Hospital of Essen, 45147, Essen, Germany
| | - Chunchen Wu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China.
| | - Xinwen Chen
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, 430071, Wuhan, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China.
| |
Collapse
|
83
|
Eller C, Heydmann L, Colpitts CC, Verrier ER, Schuster C, Baumert TF. The functional role of sodium taurocholate cotransporting polypeptide NTCP in the life cycle of hepatitis B, C and D viruses. Cell Mol Life Sci 2018; 75:3895-3905. [PMID: 30097692 PMCID: PMC7613421 DOI: 10.1007/s00018-018-2892-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/02/2018] [Accepted: 07/25/2018] [Indexed: 12/11/2022]
Abstract
Chronic hepatitis B, C and D virus (HBV, HCV and HDV) infections are a major cause of liver disease and cancer worldwide. Despite employing distinct replication strategies, the three viruses are exclusively hepatotropic, and therefore depend on hepatocyte-specific host factors. The sodium taurocholate co-transporting polypeptide (NTCP), a transmembrane protein highly expressed in human hepatocytes that mediates the transport of bile acids, plays a key role in HBV and HDV entry into hepatocytes. Recently, NTCP has been shown to modulate HCV infection of hepatocytes by regulating innate antiviral immune responses in the liver. Here, we review the current knowledge of the functional role and the molecular and cellular biology of NTCP in the life cycle of the three major hepatotropic viruses, highlight the impact of NTCP as an antiviral target and discuss future avenues of research.
Collapse
Affiliation(s)
- Carla Eller
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Laura Heydmann
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Che C Colpitts
- Division of Infection and Immunity, University College London, London, UK
| | - Eloi R Verrier
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Catherine Schuster
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France
- Université de Strasbourg, 67000, Strasbourg, France
| | - Thomas F Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 3 Rue Koeberlé, 67000, Strasbourg, France.
- Université de Strasbourg, 67000, Strasbourg, France.
- Institut Hospitalo-Universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, 67000, Strasbourg, France.
| |
Collapse
|
84
|
Expansion and differentiation of human hepatocyte-derived liver progenitor-like cells and their use for the study of hepatotropic pathogens. Cell Res 2018; 29:8-22. [PMID: 30361550 PMCID: PMC6318298 DOI: 10.1038/s41422-018-0103-x] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/10/2018] [Accepted: 09/25/2018] [Indexed: 12/12/2022] Open
Abstract
The study of pathophysiological mechanisms in human liver disease has been constrained by the inability to expand primary hepatocytes in vitro while maintaining proliferative capacity and metabolic function. We and others have previously shown that mouse mature hepatocytes can be converted to liver progenitor-like cells in vitro with defined chemical factors. Here we describe a protocol achieving efficient conversion of human primary hepatocytes into liver progenitor-like cells (HepLPCs) through delivery of developmentally relevant cues, including NAD + -dependent deacetylase SIRT1 signaling. These HepLPCs could be expanded significantly during in vitro passage. The expanded cells can readily be converted back into metabolically functional hepatocytes in vitro and upon transplantation in vivo. Under three-dimensional culture conditions, differentiated cells generated from HepLPCs regained the ability to support infection or reactivation of hepatitis B virus (HBV). Our work demonstrates the utility of the conversion between hepatocyte and liver progenitor-like cells for studying HBV biology and antiviral therapies. These findings will facilitate the study of liver diseases and regenerative medicine.
Collapse
|
85
|
Kostyusheva A, Kostyushev D, Brezgin S, Volchkova E, Chulanov V. Clinical Implications of Hepatitis B Virus RNA and Covalently Closed Circular DNA in Monitoring Patients with Chronic Hepatitis B Today with a Gaze into the Future: The Field Is Unprepared for a Sterilizing Cure. Genes (Basel) 2018; 9:E483. [PMID: 30301171 PMCID: PMC6210151 DOI: 10.3390/genes9100483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/30/2018] [Accepted: 10/03/2018] [Indexed: 12/12/2022] Open
Abstract
. Chronic hepatitis B virus (HBV) infection has long remained a critical global health issue. Covalently closed circular DNA (cccDNA) is a persistent form of the HBV genome that maintains HBV chronicity. Decades of extensive research resulted in the two therapeutic options currently available: nucleot(s)ide analogs and interferon (IFN) therapy. A plethora of reliable markers to monitor HBV patients has been established, including the recently discovered encapsidated pregenomic RNA in serum, which can be used to determine treatment end-points and to predict the susceptibility of patients to IFN. Additionally, HBV RNA splice variants and cccDNA and its epigenetic modifications are associated with the clinical course and risks of hepatocellular carcinoma (HCC) and liver fibrosis. However, new antivirals, including CRISPR/Cas9, APOBEC-mediated degradation of cccDNA, and T-cell therapies aim at completely eliminating HBV, and it is clear that the diagnostic arsenal for defining the long-awaited sterilizing cure is missing. In this review, we discuss the currently available tools for detecting and measuring HBV RNAs and cccDNA, as well as the state-of-the-art in clinical implications of these markers, and debate needs and goals within the context of the sterilizing cure that is soon to come.
Collapse
Affiliation(s)
| | | | - Sergey Brezgin
- Central Research Institute of Epidemiology, Moscow, 111123, Russia.
- National Research Centre, Institute of Immunology, Federal Medical Biological Agency, Moscow, 115478, Russia.
| | - Elena Volchkova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119146, Russia.
| | - Vladimir Chulanov
- Central Research Institute of Epidemiology, Moscow, 111123, Russia.
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of the Russian Federation, Moscow, 119146, Russia.
| |
Collapse
|
86
|
Yu Y, Li S, Liang W. Bona fide receptor for hepatitis B and D viral infections: Mechanism, research models and molecular drug targets. Emerg Microbes Infect 2018; 7:134. [PMID: 30050063 PMCID: PMC6062556 DOI: 10.1038/s41426-018-0137-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
Abstract
Hepatitis B infections have become a serious public health issue globally, and the current first-line antiviral treatment for this disease is not a true cure. Recently, sodium taurocholate cotransporting polypeptide (NTCP), a liver-specific bile acid transporter, was identified as a bona fide receptor for hepatitis B virus (HBV) and its satellite virus, hepatitis delta virus (HDV). Identification of the HBV receptor has led to the development of robust cell cultures and provides a potential target for new treatments. This review summarizes the process by which NTCP was discovered and describes its clinical significance as the receptor for HBV and HDV entry.
Collapse
Affiliation(s)
- Yueran Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.,Shengzhou People's Hospital, Shengzhou Branch of the First Affiliated Hospital of Zhejiang University, Shengzhou, 312400, China
| | - Shangda Li
- Department of Psychiatry, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Weifeng Liang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China. .,Shengzhou People's Hospital, Shengzhou Branch of the First Affiliated Hospital of Zhejiang University, Shengzhou, 312400, China.
| |
Collapse
|
87
|
Passioura T, Watashi K, Fukano K, Shimura S, Saso W, Morishita R, Ogasawara Y, Tanaka Y, Mizokami M, Sureau C, Suga H, Wakita T. De Novo Macrocyclic Peptide Inhibitors of Hepatitis B Virus Cellular Entry. Cell Chem Biol 2018; 25:906-915.e5. [PMID: 29779957 DOI: 10.1016/j.chembiol.2018.04.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 02/01/2018] [Accepted: 04/11/2018] [Indexed: 12/19/2022]
Abstract
Hepatitis B virus (HBV) constitutes a significant public health burden, and currently available treatment options are not generally curative, necessitating the development of new therapeutics. Here we have applied random non-standard peptide integrated discovery (RaPID) screening to identify small macrocyclic peptide inhibitors of HBV entry that target the cell-surface receptor for HBV, sodium taurocholate cotransporting polypeptide (NTCP). In addition to their anti-HBV activity, these molecules also inhibit cellular entry by the related hepatitis D virus (HDV), and are active against diverse strains of HBV (including clinically relevant nucleos(t)ide analog-resistant and vaccine escaping strains). Importantly, these macrocyclic peptides, in contrast to other NTCP-binding HBV entry inhibitors, exhibited no inhibition of NTCP-mediated bile acid uptake, making them appealing candidates for therapeutic development.
Collapse
Affiliation(s)
- Toby Passioura
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Department of Applied Biological Sciences, Tokyo University of Science, Noda 278-8510, Japan; JST CREST, Saitama 332-0012, Japan
| | - Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose 204-8588, Japan
| | - Satomi Shimura
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Wakana Saso
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ryo Morishita
- CellFree Sciences Co., Ltd., Matsuyama 790-8577, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose 204-8588, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University, Graduate School of Medicinal Sciences, Nagoya 467-8601, Japan
| | - Masashi Mizokami
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa 272-8516, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, INSERM U1134, Paris 75015, France
| | - Hiroaki Suga
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Tokyo 113-0033, Japan; JST CREST, The University of Tokyo, Tokyo 113-0033, Japan.
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.
| |
Collapse
|
88
|
Müller SF, König A, Döring B, Glebe D, Geyer J. Characterisation of the hepatitis B virus cross-species transmission pattern via Na+/taurocholate co-transporting polypeptides from 11 New World and Old World primate species. PLoS One 2018; 13:e0199200. [PMID: 29912972 PMCID: PMC6005513 DOI: 10.1371/journal.pone.0199200] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/04/2018] [Indexed: 12/18/2022] Open
Abstract
The hepatic Na+/taurocholate co-transporting polypeptide (NTCP in man, Ntcp in animals) is the high-affinity receptor for the hepatitis B (HBV) and hepatitis D (HDV) viruses. Species barriers for human HBV/HDV within the order Primates were previously attributed to Ntcp sequence variations that disable virus-receptor interaction. However, only a limited number of primate Ntcps have been analysed so far. In the present study, a total of 11 Ntcps from apes, Old and New World monkeys were cloned and expressed in vitro to characterise their interaction with HBV and HDV. All Ntcps showed intact bile salt transport. Human NTCP as well as the Ntcps from the great apes chimpanzee and orangutan showed transport-competing binding of HBV derived myr-preS1-peptides. In contrast, all six Ntcps from the group of Old World monkeys were insensitive to HBV myr-preS1-peptide binding and HBV/HDV infection. This is basically predetermined by the amino acid arginine at position 158 of all studied Old World monkey Ntcps. An exchange from arginine to glycine (as present in humans and great apes) at this position (R158G) alone was sufficient to achieve full transport-competing HBV myr-preS1-peptide binding and susceptibility for HBV/HDV infection. New World monkey Ntcps showed higher sequence heterogeneity, but in two cases with 158G showed transport-competing HBV myr-preS1-peptide binding, and in one case (Saimiri sciureus) even susceptibility for HBV/HDV infection. In conclusion, amino acid position 158 of NTCP/Ntcp is sufficient to discriminate between the HBV/HDV susceptible group of humans and great apes (158G) and the non-susceptible group of Old World monkeys (158R). In the case of the phylogenetically more distant New World monkey Ntcps amino acid 158 plays a significant, but not exclusive role.
Collapse
Affiliation(s)
- Simon F. Müller
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Alexander König
- Institute of Medical Virology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Barbara Döring
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Dieter Glebe
- Institute of Medical Virology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| | - Joachim Geyer
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
89
|
A novel hepatitis B virus species discovered in capuchin monkeys sheds new light on the evolution of primate hepadnaviruses. J Hepatol 2018; 68:1114-1122. [PMID: 29428874 DOI: 10.1016/j.jhep.2018.01.029] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 01/19/2018] [Accepted: 01/27/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS All known hepatitis B virus (HBV) genotypes occur in humans and hominoid Old World non-human primates (NHPs). The divergent woolly monkey HBV (WMHBV) forms another orthohepadnavirus species. The evolutionary origins of HBV are unclear. METHODS We analysed sera from 124 Brazilian monkeys collected during 2012-2016 for hepadnaviruses using molecular and serological tools, and conducted evolutionary analyses. RESULTS We identified a novel orthohepadnavirus species in capuchin monkeys (capuchin monkey hepatitis B virus [CMHBV]). We found CMHBV-specific antibodies in five animals and high CMHBV concentrations in one animal. Non-inflammatory, probably chronic infection was consistent with an intact preCore domain, low genetic variability, core deletions in deep sequencing, and no elevated liver enzymes. Cross-reactivity of antisera against surface antigens suggested antigenic relatedness of HBV, CMHBV, and WMHBV. Infection-determining CMHBV surface peptides bound to the human HBV receptor (human sodium taurocholate co-transporting polypeptide), but preferentially interacted with the capuchin monkey receptor homologue. CMHBV and WMHBV pseudotypes infected human hepatoma cells via the human sodium taurocholate co-transporting polypeptide, and were poorly neutralised by HBV vaccine-derived antibodies, suggesting that cross-species infections may be possible. Ancestral state reconstructions and sequence distance comparisons associated HBV with humans, whereas primate hepadnaviruses as a whole were projected to NHP ancestors. Co-phylogenetic analyses yielded evidence for co-speciation of hepadnaviruses and New World NHP. Bayesian hypothesis testing yielded strong support for an association of the HBV stem lineage with hominoid ancestors. Neither CMHBV nor WMHBV was likely the ancestor of the divergent human HBV genotypes F/H found in American natives. CONCLUSIONS Our data suggest ancestral co-speciation of hepadnaviruses and NHP, and an Old World origin of the divergent HBV genotypes F/H. The identification of a novel primate hepadnavirus offers new perspectives for urgently needed animal models of chronic hepatitis B. LAY SUMMARY The origins of HBV are unclear. The new orthohepadnavirus species from Brazilian capuchin monkeys resembled HBV in elicited infection patterns and could infect human liver cells using the same receptor as HBV. Evolutionary analyses suggested that primate HBV-related viruses might have emerged in African ancestors of New World monkeys millions of years ago. HBV was associated with hominoid primates, including humans and apes, suggesting evolutionary origins of HBV before the formation of modern humans. HBV genotypes found in American natives were divergent from those found in American monkeys, and likely introduced along prehistoric human migration. Our results elucidate the evolutionary origins and dispersal of primate HBV, identify a new orthohepadnavirus reservoir, and enable new perspectives for animal models of hepatitis B.
Collapse
|
90
|
Virus entry and its inhibition to prevent and treat hepatitis B and hepatitis D virus infections. Curr Opin Virol 2018; 30:68-79. [DOI: 10.1016/j.coviro.2018.04.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/19/2018] [Accepted: 04/20/2018] [Indexed: 12/13/2022]
|
91
|
Saso W, Tsukuda S, Ohashi H, Fukano K, Morishita R, Matsunaga S, Ohki M, Ryo A, Park SY, Suzuki R, Aizaki H, Muramatsu M, Sureau C, Wakita T, Matano T, Watashi K. A new strategy to identify hepatitis B virus entry inhibitors by AlphaScreen technology targeting the envelope-receptor interaction. Biochem Biophys Res Commun 2018; 501:374-379. [PMID: 29730285 DOI: 10.1016/j.bbrc.2018.04.187] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022]
Abstract
Current anti-hepatitis B virus (HBV) agents have limited effect in curing HBV infection, and thus novel anti-HBV agents with different modes of action are in demand. In this study, we applied AlphaScreen assay to high-throughput screening of small molecules inhibiting the interaction between HBV large surface antigen (LHBs) and the HBV entry receptor, sodium taurocholate cotransporting polypeptide (NTCP). From the chemical screening, we identified that rapamycin, an immunosuppressant, strongly inhibited the LHBs-NTCP interaction. Rapamycin inhibited hepatocyte infection with HBV without significant cytotoxicity. This activity was due to impaired attachment of the LHBs preS1 domain to cell surface. Pretreatment of target cells with rapamycin remarkably reduced their susceptibility to preS1 attachment, while rapamycin pretreatment to preS1 did not affect its attachment activity, suggesting that rapamycin targets the host side. In support of this, a surface plasmon resonance analysis showed a direct interaction of rapamycin with NTCP. Consistently, rapamycin also prevented hepatitis D virus infection, whose entry into cells is also mediated by NTCP. We also identified two rapamycin derivatives, everolimus and temsirolimus, which possessed higher anti-HBV potencies than rapamycin. Thus, this is the first report for application of AlphaScreen technology that monitors a viral envelope-receptor interaction to identify viral entry inhibitors.
Collapse
Affiliation(s)
- Wakana Saso
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan; The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Senko Tsukuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan; Liver Cancer Prevention Research Unit, RIKEN Center for Integrative Medical Sciences (IMS), Wako, Japan
| | - Hirofumi Ohashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan; Department of Applied Biological Sciences, Tokyo University of Science, Noda, Japan
| | - Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan; Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | | | - Satoko Matsunaga
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mio Ohki
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masamichi Muramatsu
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, INSERM U1134, Paris, France
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tetsuro Matano
- AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan; The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan; Department of Applied Biological Sciences, Tokyo University of Science, Noda, Japan; CREST, JST, Saitama, Japan.
| |
Collapse
|
92
|
Yuan L, Liu X, Zhang L, Li X, Zhang Y, Wu K, Chen Y, Cao J, Hou W, Zhang J, Zhu H, Yuan Q, Tang Q, Cheng T, Xia N. A Chimeric Humanized Mouse Model by Engrafting the Human Induced Pluripotent Stem Cell-Derived Hepatocyte-Like Cell for the Chronic Hepatitis B Virus Infection. Front Microbiol 2018; 9:908. [PMID: 29867819 PMCID: PMC5952038 DOI: 10.3389/fmicb.2018.00908] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/18/2018] [Indexed: 12/18/2022] Open
Abstract
Humanized mouse model generated by grafting primary human hepatocytes (PHHs) to immunodeficient mouse has contributed invaluably to understanding the pathogenesis of hepatitis B virus (HBV). However, the source of PHHs is limited, which necessitates the search for alternatives. Recently, hepatocyte-like cells (HLCs) generated from human induced pluripotent stem cells (hiPSCs) have been used for in vitro HBV infection. Herein, we developed a robust human liver chimeric animal model to study in vivo HBV infection by engrafting the hiPSC-HLCs to Fah-/-Rag2-/-IL-2Rγc-/-SCID (FRGS) mice. After being optimized by a small molecule, XMU-MP-1, the hiPSC-HLCs engrafted FRGS (hHLC-FRGS) mice displayed approximately 40% liver chimerism at week 6 after engraftment and maintained at this level for at least 14 weeks. Viremia and HBV infection markers include antigens, RNA, DNA, and covalently closed circular DNA were detectable in HBV infected hHLC-FRGS mice. Furthermore, hiPSC-HLCs and hHLC-FRGS mice were successfully used to evaluate different antivirals. Therefore, we established a humanized mouse model for not only investigating HBV pathogenesis but also testing the effects of the anti-HBV drugs. Highlights: (1) The implanted hiPSC-HLCs established a long-term chimerism in FRGS mice liver. (2) hHLC-FRGS mice are adequate to support chronic HBV infection with a full viral life cycle. (3) hiPSC-HLCs and hHLC-FRGS mice are useful tools for evaluation of antivirals against HBV infection in vitro and in vivo. Research in Context To overcome the disadvantages of using primary human hepatocytes, we induced human pluripotent stem cells to hepatocyte-like cells (hiPSC-HLCs) that developed the capability to express important liver functional markers and critical host factors for HBV infection. The hiPSC-HLCs were permissive for the HBV infection and supported a full HBV replication. The hiPSC-HLCs were then engrafted to immunodeficient mouse to establish a chimeric liver mouse model, which was capable of supporting HBV infection in vivo and evaluating the effects of antiviral drugs. Our results shed light into improving the cellular and animal models for studying HBV and other hepatotropic viruses.
Collapse
Affiliation(s)
- Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Xuan Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Liang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Xiaoling Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Yali Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Kun Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Yao Chen
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Jiali Cao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Wangheng Hou
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Hua Zhu
- Department of Microbiology and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, NJ, United States
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, United States
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, Xiamen University, Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
93
|
Miyakawa K, Matsunaga S, Yamaoka Y, Dairaku M, Fukano K, Kimura H, Chimuro T, Nishitsuji H, Watashi K, Shimotohno K, Wakita T, Ryo A. Development of a cell-based assay to identify hepatitis B virus entry inhibitors targeting the sodium taurocholate cotransporting polypeptide. Oncotarget 2018; 9:23681-23694. [PMID: 29805766 PMCID: PMC5955094 DOI: 10.18632/oncotarget.25348] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 04/24/2018] [Indexed: 12/18/2022] Open
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) is a major entry receptor of hepatitis B virus (HBV) and one of the most attractive targets for anti-HBV drugs. We developed a cell-mediated drug screening method to monitor NTCP expression on the cell surface by generating a HepG2 cell line with tetracycline-inducible expression of NTCP and a monoclonal antibody that specifically detects cell-surface NTCP. Using this system, we screened a small molecule library for compounds that protected against HBV infection by targeting NTCP. We found that glabridin, a licorice-derived isoflavane, could suppress viral infection by inducing caveolar endocytosis of cell-surface NTCP with an IC50 of ~40 μM. We also found that glabridin could attenuate the inhibitory effect of taurocholate on type I interferon signaling by depleting the level of cell-surface NTCP. These results demonstrate that our screening system could be a powerful tool for discovering drugs targeting HBV entry.
Collapse
Affiliation(s)
- Kei Miyakawa
- Department of Microbiology, Yokohama City University School of Medicine, Kanagawa 236-0004, Japan
| | - Satoko Matsunaga
- Department of Microbiology, Yokohama City University School of Medicine, Kanagawa 236-0004, Japan
| | - Yutaro Yamaoka
- Department of Microbiology, Yokohama City University School of Medicine, Kanagawa 236-0004, Japan.,Isehara Research Laboratory, Technology and Development Division, Kanto Chemical Co., Inc., Kanagawa 259-1146, Japan
| | - Mina Dairaku
- Department of Microbiology, Yokohama City University School of Medicine, Kanagawa 236-0004, Japan
| | - Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Hirokazu Kimura
- School of Medical Technology, Faculty of Health Sciences, Gunma Paz University, Gunma 370-0006, Japan
| | - Tomoyuki Chimuro
- Isehara Research Laboratory, Technology and Development Division, Kanto Chemical Co., Inc., Kanagawa 259-1146, Japan
| | - Hironori Nishitsuji
- Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba 272-8516, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Kunitada Shimotohno
- Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Chiba 272-8516, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Kanagawa 236-0004, Japan
| |
Collapse
|
94
|
Feng S, Gao L, Han X, Hu T, Hu Y, Liu H, Thomas AW, Yan Z, Yang S, Young JAT, Yun H, Zhu W, Shen HC. Discovery of Small Molecule Therapeutics for Treatment of Chronic HBV Infection. ACS Infect Dis 2018; 4:257-277. [PMID: 29369612 DOI: 10.1021/acsinfecdis.7b00144] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The chronic infection of hepatitis B virus (HBV) inflicts 250 million people worldwide representing a major public health threat. A significant subpopulation of patients eventually develop cirrhosis and hepatocellular carcinoma (HCC). Unfortunately, none of the current standard therapies for chronic hepatitis B (CHB) result in a satisfactory clinical cure rate. Driven by a highly unmet medical need, multiple pharmaceutical companies and research institutions have been engaged in drug discovery and development to improve the CHB functional cure rate, defined by sustainable viral suppression and HBsAg clearance after a finite treatment. This Review summarizes the recent advances in the discovery and development of novel anti-HBV small molecules. It is believed that an improved CHB functional cure rate may be accomplished via the combination of molecules with distinct MoAs. Thus, certain molecules may evolve into key components of a suitable combination therapy leading to superior outcome of clinical efficacy in the future.
Collapse
Affiliation(s)
- Song Feng
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Lu Gao
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Xingchun Han
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Taishan Hu
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Yimin Hu
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Haixia Liu
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Andrew W. Thomas
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Zhipeng Yan
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Song Yang
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - John A. T. Young
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Hongying Yun
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Wei Zhu
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| | - Hong C. Shen
- Roche Innovation Center Shanghai, Roche Pharma Research & Early Development, Building 5, 720 Cailun Road, Shanghai, 201203, China
| |
Collapse
|
95
|
Bi YA, Mathialagan S, Tylaska L, Fu M, Keefer J, Vildhede A, Costales C, Rodrigues AD, Varma MVS. Organic Anion Transporter 2 Mediates Hepatic Uptake of Tolbutamide, a CYP2C9 Probe Drug. J Pharmacol Exp Ther 2018; 364:390-398. [PMID: 29326367 DOI: 10.1124/jpet.117.245951] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/02/2018] [Indexed: 03/08/2025] Open
Abstract
Tolbutamide is primarily metabolized by CYP2C9, and, thus, is frequently applied as a clinical probe substrate for CYP2C9 activity. However, there is a marked discrepancy in the in vitro-in vivo extrapolation of its metabolic clearance, implying a potential for additional clearance mechanisms. The goal of this study was to evaluate the role of hepatic uptake transport in the pharmacokinetics of tolbutamide and to identify the molecular mechanism thereof. Transport studies using singly transfected cells expressing six major hepatic uptake transporters showed that tolbutamide is a substrate to organic anion transporter 2 (OAT2) alone with transporter affinity [Michaelis-Menten constant (Km)] of 19.5 ± 4.3 µM. Additionally, OAT2-specific transport was inhibited by ketoprofen (an OAT2 inhibitor) and 1 mM rifamycin SV (pan inhibitor), but not by cyclosporine and rifampicin (OAT polypeptides/Na+-taurocholate cotransporting polypeptide inhibitors). Uptake studies in primary human hepatocytes confirmed the predominant role of OAT2 in the active uptake with significant inhibition by rifamycin SV and ketoprofen, but not by the other inhibitors. Concentration-dependent uptake was noted in human hepatocytes with active transport characterized by Km and Vmax values of 39.3 ± 6.6 µM and 426 ± 30 pmol/min per milligram protein, respectively. Bottom-up physiologically based pharmacokinetic modeling was employed to verify the proposed role of OAT2-mediated hepatic uptake. In contrast to the rapid equilibrium (CYP2C9-only) model, the permeability-limited (OAT2-CYP2C9 interplay) model better described the plasma concentration-time profiles of tolbutamide. Additionally, the latter well described tolbutamide pharmacokinetics in carriers of CYP2C9 genetic variants and quantitatively rationalized its known drug-drug interactions. Our results provide first-line evidence for the role of OAT2-mediated hepatic uptake in the pharmacokinetics of tolbutamide, and imply the need for additional clinical studies in this direction.
Collapse
Affiliation(s)
- Yi-An Bi
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Laurie Tylaska
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Myra Fu
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Julie Keefer
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Anna Vildhede
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Chester Costales
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - A David Rodrigues
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| | - Manthena V S Varma
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
96
|
Kaneko M, Futamura Y, Tsukuda S, Kondoh Y, Sekine T, Hirano H, Fukano K, Ohashi H, Saso W, Morishita R, Matsunaga S, Kawai F, Ryo A, Park SY, Suzuki R, Aizaki H, Ohtani N, Sureau C, Wakita T, Osada H, Watashi K. Chemical array system, a platform to identify novel hepatitis B virus entry inhibitors targeting sodium taurocholate cotransporting polypeptide. Sci Rep 2018; 8:2769. [PMID: 29426822 PMCID: PMC5807303 DOI: 10.1038/s41598-018-20987-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
Current anti-hepatitis B virus (HBV) agents including interferons and nucleos(t)ide analogs efficiently suppress HBV infection. However, as it is difficult to eliminate HBV from chronically infected liver, alternative anti-HBV agents targeting a new molecule are urgently needed. In this study, we applied a chemical array to high throughput screening of small molecules that interacted with sodium taurocholate cotransporting polypeptide (NTCP), an entry receptor for HBV. From approximately 30,000 compounds, we identified 74 candidates for NTCP interactants, and five out of these were shown to inhibit HBV infection in cell culture. One of such compound, NPD8716, a coumarin derivative, interacted with NTCP and inhibited HBV infection without causing cytotoxicity. Consistent with its NTCP interaction capacity, this compound was shown to block viral attachment to host hepatocytes. NPD8716 also prevented the infection with hepatitis D virus, but not hepatitis C virus, in agreement with NPD8716 specifically inhibiting NTCP-mediated infection. Analysis of derivative compounds showed that the anti-HBV activity of compounds was apparently correlated with the affinity to NTCP and the capacity to impair NTCP-mediated bile acid uptake. These results are the first to show that the chemical array technology represents a powerful platform to identify novel viral entry inhibitors.
Collapse
Affiliation(s)
- Manabu Kaneko
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.,Department of Applied Biological Sciences, Tokyo University of Science, Noda, 278-8510, Japan
| | - Yushi Futamura
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science (CSRS), Wako, 351-0198, Japan
| | - Senko Tsukuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.,Micro-signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies (CLST), Wako, 351-0198, Japan
| | - Yasumitsu Kondoh
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science (CSRS), Wako, 351-0198, Japan
| | - Tomomi Sekine
- Bio-Active Compounds Discovery Research Unit, RIKEN CSRS, Wako, 351-0198, Japan
| | - Hiroyuki Hirano
- Chemical Resource Development Research Unit, RIKEN CSRS, Wako, 351-0198, Japan
| | - Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.,Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, 204-8588, Japan
| | - Hirofumi Ohashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.,Department of Applied Biological Sciences, Tokyo University of Science, Noda, 278-8510, Japan
| | - Wakana Saso
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan.,The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Ryo Morishita
- CellFree Sciences Co., Ltd, Matsuyama, 790-8577, Japan
| | - Satoko Matsunaga
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0027, Japan
| | - Fumihiro Kawai
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0027, Japan
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, 230-0045, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Naoko Ohtani
- Department of Applied Biological Sciences, Tokyo University of Science, Noda, 278-8510, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine, INSERM U1134, Paris, 75015, France
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Hiroyuki Osada
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science (CSRS), Wako, 351-0198, Japan.
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan. .,Department of Applied Biological Sciences, Tokyo University of Science, Noda, 278-8510, Japan. .,CREST, JST, Saitama, 332-0012, Japan.
| |
Collapse
|
97
|
Thanapirom K, Suksawatamnuay S, Sukeepaisarnjaroen W, Treeprasertsuk S, Tanwandee T, Charatcharoenwitthaya P, Thongsawat S, Leerapun A, Piratvisuth T, Boonsirichan R, Bunchorntavakul C, Pattanasirigool C, Pornthisarn B, Tuntipanichteerakul S, Sripariwuth E, Jeamsripong W, Sanpajit T, Poovorawan Y, Komolmit P. Association of the S267F variant on NTCP gene and treatment response to pegylated interferon in patients with chronic hepatitis B: a multicentre study. Antivir Ther 2018; 23:67-75. [PMID: 28635613 DOI: 10.3851/imp3179] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Sodium taurocholate co-transporting polypeptide (NTCP) is a cell receptor for HBV. The S267F variant on the NTCP gene is inversely associated with the chronicity of HBV infection, progression to cirrhosis and hepatocellular carcinoma in East Asian populations. The aim of this study was to determine whether the S267F variant was associated with response to pegylated interferon (PEG-IFN) in patients with chronic HBV infection. METHODS A total of 257 patients with chronic HBV, treated with PEG-IFN for 48 weeks, were identified from 13 tertiary hospitals included in the hepatitis B database of the Thai Association for the Study of the Liver (THASL). RESULTS Of these, 202 patients were infected with HBV genotype C (84.9%); 146 patients were hepatitis B e antigen (HBeAg)-positive (56.8%). Genotypic frequencies of the S267F polymorphism were 85.2%, 14.8% and 0% for the GG, GA and AA genotypes, respectively. S267F GA was associated with sustained alanine aminotransferase (ALT) normalization (OR = 3.25, 95% CI 1.23, 8.61; P=0.02) in HBeAg-positive patients. Patients with S267F variant tended to have more virological response, sustained response with hepatitis B surface antigen (HBsAg) loss at 24 weeks following PEG-IFN treatment. There was no association between the S267F variant and improved patient outcomes in HBeAg-negative patients. CONCLUSIONS The S267F variant on the NTCP gene is independently associated with sustained normalization of ALT following treatment with PEG-IFN in patients with HBV infection who are HBeAg-positive. The findings of this study provide additional support for the clinical significance of the S267F variant of NTCP beyond HBV entry.
Collapse
Affiliation(s)
- Kessarin Thanapirom
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Liver Diseases, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Sirinporn Suksawatamnuay
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Liver Diseases, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | | | - Sombat Treeprasertsuk
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Liver Diseases, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Tawesak Tanwandee
- Division of Gastroenterology, Department of Medicine, Siriraj Hospital, Bangkok, Thailand
| | | | - Satawat Thongsawat
- Department of Internal Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apinya Leerapun
- Department of Internal Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | | | | | | | | | | | | | | | | | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Piyawat Komolmit
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
- Center of Excellence in Liver Diseases, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
98
|
Lee HW, Park HJ, Jin B, Dezhbord M, Kim DY, Han KH, Ryu WS, Kim S, Ahn SH. Effect of S267F variant of NTCP on the patients with chronic hepatitis B. Sci Rep 2017; 7:17634. [PMID: 29247233 PMCID: PMC5732244 DOI: 10.1038/s41598-017-17959-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/02/2017] [Indexed: 01/05/2023] Open
Abstract
Sodium taurocholate cotransporting polypeptide (NTCP) was identified as an entry receptor for hepatitis B virus (HBV) infection. The substitution of serine at position 267 of NTCP with phenylalanine (S267F) is an Asian-specific variation that hampers HBV entry in vitro. In this study, we aimed to evaluate the prevalence of S267F polymorphism in Korean patients with chronic hepatitis B (CHB) and its association with disease progression and potential viral evolution in the preS1 domain of HBV. We found that the frequency of the S267F variant of NTCP in CHB patients and controls was 2.7% and 5.7% (P = 0.031), respectively, and that those who had S267F variant were less susceptible to chronic HBV infection. The frequency of the S267F variant in CHB, cirrhosis and hepatocellular carcinoma (HCC) patients was 3.3%, 0.9%, and 3.5%, respectively. Thus, the S267F variant correlated significantly with a lower risk for cirrhosis (P = 0.036). Sequencing preS1 domain of HBV from the patients who had S267F variant revealed no significant sequence change compared to the wild type. In conclusion, the S267F variant of NTCP is clinically associated with a lower risk of chronic HBV infection and cirrhosis development, which implicates suppressing HBV entry could reduce the disease burden.
Collapse
Affiliation(s)
- Hye Won Lee
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Jung Park
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Bora Jin
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | | | - Do Young Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Wang-Shick Ryu
- Department of Biochemistry, Yonsei University, Seoul, Korea.,Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, Korea
| | - Seungtaek Kim
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea. .,Institut Pasteur Korea, Seongnam-si, Gyeonggi-do, Korea.
| | - Sang Hoon Ahn
- Department of Internal Medicine, Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
99
|
Verrier ER, Colpitts CC, Bach C, Heydmann L, Zona L, Xiao F, Thumann C, Crouchet E, Gaudin R, Sureau C, Cosset FL, McKeating JA, Pessaux P, Hoshida Y, Schuster C, Zeisel MB, Baumert TF. Solute Carrier NTCP Regulates Innate Antiviral Immune Responses Targeting Hepatitis C Virus Infection of Hepatocytes. Cell Rep 2017; 17:1357-1368. [PMID: 27783949 PMCID: PMC5098118 DOI: 10.1016/j.celrep.2016.09.084] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 08/10/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022] Open
Abstract
Chronic hepatitis B, C, and D virus (HBV, HCV, and HDV) infections are the leading causes of liver disease and cancer worldwide. Recently, the solute carrier and sodium taurocholate co-transporter NTCP has been identified as a receptor for HBV and HDV. Here, we uncover NTCP as a host factor regulating HCV infection. Using gain- and loss-of-function studies, we show that NTCP mediates HCV infection of hepatocytes and is relevant for cell-to-cell transmission. NTCP regulates HCV infection by augmenting the bile-acid-mediated repression of interferon-stimulated genes (ISGs), including IFITM3. In conclusion, our results uncover NTCP as a mediator of innate antiviral immune responses in the liver, and they establish a role for NTCP in the infection process of multiple viruses via distinct mechanisms. Collectively, our findings suggest a role for solute carriers in the regulation of innate antiviral responses, and they have potential implications for virus-host interactions and antiviral therapies. NTCP is involved in hepatocyte infection by multiple viruses via distinct mechanisms NTCP facilitates HCV infection by modulating innate antiviral responses Solute carrier NTCP is a regulator of antiviral immune responses in the liver This function is relevant for infection and therapies for hepatotropic viruses
Collapse
Affiliation(s)
- Eloi R Verrier
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Che C Colpitts
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Charlotte Bach
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Laura Heydmann
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Laetitia Zona
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Fei Xiao
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Christine Thumann
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Emilie Crouchet
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Raphaël Gaudin
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Camille Sureau
- INTS, Laboratoire de Virologie Moléculaire, 75015 Paris, France
| | - François-Loïc Cosset
- CIRI-International Center for Infectiology Research, 69364 Lyon Cedex 07, France; INSERM, U1111, 69007 Lyon, France; Ecole Normale Supérieure, 69007 Lyon, France; Centre National de la Recherche Scientifique (CNRS) UMR 5308, 69007 Lyon, France; LabEx Ecofect, University of Lyon, 69007 Lyon, France
| | - Jane A McKeating
- Centre for Human Virology, University of Birmingham, Birmingham, UK; NIHR Liver Biomedical Research Unit, University of Birmingham, Birmingham, UK
| | - Patrick Pessaux
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France; Institut Hospitalo-universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, 67000 Strasbourg, France
| | - Yujin Hoshida
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Catherine Schuster
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France
| | - Mirjam B Zeisel
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France.
| | - Thomas F Baumert
- INSERM, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, 67000 Strasbourg, France; Université de Strasbourg, 67000 Strasbourg, France; Institut Hospitalo-universitaire, Pôle Hépato-digestif, Nouvel Hôpital Civil, 67000 Strasbourg, France.
| |
Collapse
|
100
|
Shimura S, Watashi K, Fukano K, Peel M, Sluder A, Kawai F, Iwamoto M, Tsukuda S, Takeuchi JS, Miyake T, Sugiyama M, Ogasawara Y, Park SY, Tanaka Y, Kusuhara H, Mizokami M, Sureau C, Wakita T. Cyclosporin derivatives inhibit hepatitis B virus entry without interfering with NTCP transporter activity. J Hepatol 2017; 66:685-692. [PMID: 27890789 PMCID: PMC7172969 DOI: 10.1016/j.jhep.2016.11.009] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 10/25/2016] [Accepted: 11/14/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS The sodium taurocholate co-transporting polypeptide (NTCP) is the main target of most hepatitis B virus (HBV) specific entry inhibitors. Unfortunately, these agents also block NTCP transport of bile acids into hepatocytes, and thus have the potential to cause adverse effects. We aimed to identify small molecules that inhibit HBV entry while maintaining NTCP transporter function. METHODS We characterized a series of cyclosporine (CsA) derivatives for their anti-HBV activity and NTCP binding specificity using HepG2 cells overexpressing NTCP and primary human hepatocytes. The four most potent derivatives were tested for their capacity to prevent HBV entry, but maintain NTCP transporter function. Their antiviral activity against different HBV genotypes was analysed. RESULTS We identified several CsA derivatives that inhibited HBV infection with a sub-micromolar IC50. Among them, SCY446 and SCY450 showed low activity against calcineurin (CN) and cyclophilins (CyPs), two major CsA cellular targets. This suggested that instead, these compounds interacted directly with NTCP to inhibit viral attachment to host cells, and have no immunosuppressive function. Importantly, we found that SCY450 and SCY995 did not impair the NTCP-dependent uptake of bile acids, and inhibited multiple HBV genotypes including a clinically relevant nucleoside analog-resistant HBV isolate. CONCLUSIONS This is the first example of small molecule selective inhibition of HBV entry with no decrease in NTCP transporter activity. It suggests that the anti-HBV activity can be functionally separated from bile acid transport. These broadly active anti-HBV molecules are potential candidates for developing new drugs with fewer adverse effects. LAY SUMMARY In this study, we identified new compounds that selectively inhibited hepatitis B virus (HBV) entry, and did not impair bile acid uptake. Our evidence offers a new strategy for developing anti-HBV drugs with fewer side effects.
Collapse
Affiliation(s)
- Satomi Shimura
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; SCYNEXIS, Inc., Durham, NC 27713, USA
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Department of Applied Biological Science, Tokyo University of Sciences, Noda 278-8510, Japan; CREST, Japan Science and Technology Agency (J.S.T.), Saitama 332-0012, Japan.
| | - Kento Fukano
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose 204-8588, Japan
| | | | | | - Fumihiro Kawai
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Masashi Iwamoto
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Department of Applied Biological Science, Tokyo University of Sciences, Noda 278-8510, Japan
| | - Senko Tsukuda
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan; Micro-signaling Regulation Technology Unit, RIKEN Center for Life Science Technologies, Wako 351-0198, Japan
| | - Junko S Takeuchi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Takeshi Miyake
- The University of Tokyo, Graduate School of Pharmaceutical Sciences, Tokyo 113-0033, Japan
| | - Masaya Sugiyama
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa 272-8516, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose 204-8588, Japan
| | - Sam-Yong Park
- Drug Design Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medicinal Sciences, Nagoya 467-8601, Japan
| | - Hiroyuki Kusuhara
- The University of Tokyo, Graduate School of Pharmaceutical Sciences, Tokyo 113-0033, Japan
| | - Masashi Mizokami
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa 272-8516, Japan
| | - Camille Sureau
- Laboratoire de Virologie Moléculaire, Institut National de la Transfusion Sanguine (INTS), Paris, France
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|