Next Article in Journal
Modification of Thin Film Composite PVA/PAN Membranes for Pervaporation Using Aluminosilicate Nanoparticles
Previous Article in Journal
The Therapeutic Potential of the Restoration of the p53 Protein Family Members in the EGFR-Mutated Lung Cancer
Previous Article in Special Issue
Evaluation of Potential Anti-Hepatitis A Virus 3C Protease Inhibitors Using Molecular Docking
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Hepatitis A Virus Infection and Molecular Research

1
Division of Gastroenterology and Hepatology, Department of Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-kamicho, Itabashi-ku, Tokyo 173-8610, Japan
2
Department of Gastroenterology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 7214; https://doi.org/10.3390/ijms23137214
Submission received: 23 June 2022 / Accepted: 27 June 2022 / Published: 29 June 2022
(This article belongs to the Special Issue Hepatitis Virus Infection and Molecular Research 2022)
Hepatitis A virus (HAV) infection is a major cause of acute viral hepatitis globally, which can occasionally lead to acute liver failure (ALF) and acute-on-chronic liver failure (ACLF), which often result in death without liver transplantation. Along with concomitant increases in alcohol misuse and metabolic syndrome in recent years, HAV plays a role as an acute condition in patients with ACLF [1]. Although HAV vaccination could prevent people from being infected with HAV, HAV vaccination frequency is too low to prevent HAV infection, at least in Japan. As the number of people without anti-HAV immunity is increasing in the Asian Pacific region, according to the improvement of public health, it is important to develop anti-HAV drugs and to distribute HAV vaccinations [2]. HAV RNA genome consists of a single, long, open reading frame (ORF) flanked by a 5′-untranslated region (UTR) and 3′-UTR. The HAV internal ribosomal entry site (IRES) is located in 5′-UTR, and translates as HAV proteins in a cap-independent manner. ORF encodes structural (VP4, VP2, VP3, VP1, and pX) and non-structural (2B, 2C, 3A, 3B, 3C, and 3D) proteins [3]. One large immature protein, encoded by ORF, is primarily cut into at least ten mature proteins by HAV 3C protease, and HAV 3D has an RNA-dependent RNA polymerase, which is important for HAV replication. HAV IRES, 3C protease, and 3D polymerase in HAV genome or HAV protein are attractive targets of anti-HAV drugs (Figure 1) [4,5,6,7,8,9,10,11].
Zinc compounds suppress HAV replication [13,14]. In this Special Issue, Kanda et al. [15] revealed that zinc chloride inhibits mitogen-activated protein kinase 3 (MAP2K3) expression and downregulates HAV replication in human hepatocytes. The inhibition of MAP2K3 may also prevent patients infected with HAV from developing ALF and ACLF.
Sasaki-Tanaka et al. [9] examined several novel therapeutic drugs through a drug repositioning approach, targeting RNA-dependent RNA polymerase and RNA-dependent DNA polymerase. They found that favipiravir effectively suppressed HAV replication through the introduction of mutagenesis into the HAV genome. Of interest is that favipiravir was effective in reducing the replication of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [16].
Sasaki-Tanaka et al. [10] performed the in silico screening of anti-HAV compounds targeting the HAV 3C protease enzyme, using the Schrodinger Glide molecular docking program to study the interactions between the ligand and HAV 3C protease enzyme at an atomic level, from an antiviral library of 25,000 compounds, to evaluate anti-HAV 3C protease inhibitors. They found the HAV 3C protease inhibitor Z10325150 (Enamine, Kyiv, Ukraine) and confirmed the HAV replication inhibitory activity in human hepatocytes.
In this Special Issue, Tréguier et al. [17] also reviewed the crucial role of apolipoprotein E (ApoE) in the lifecycles of hepatitis B virus (HBV) and hepatitis C virus (HCV) and discussed its potential role in the lifecycle of other hepatotropic viruses, including HAV. Hepatotropic viruses infect hepatocytes and spread throughout the liver, using ApoE, which seems one of the attractive targets of anti-HAV drugs.
HCV NS5A activates the glucokinase (GCK) isoenzyme of hexokinases through its D2 domain (NS5A-D2) [18]. HCV NS5A-D2 can reprogram central carbon metabolism towards a more energetic and glycolytic phenotype compatible with the needs of HCV for replication. Knockout of these host factor genes may impair HCV replication [19].
Lesnova et al. [20] demonstrated the first evidence of immunomodulatory activity of antioxidant N-acetylcysteine (NAC) and polyamine biosynthesis inhibitor 2-difluoromethylornithine (DFMO) during prophylactic immunization against infectious diseases. NAC and DFMO may be used as new adjuvant compounds that enhance the immune response, and it might be useful for the development of vaccines against hepatitis viruses.
In summary, this Special Issue offers a critical overview of recent research in hepatitis A virus and molecular research, and the related areas.

Funding

This research was partly funded by the Japan Agency for Medical Research and Development (AMED), grant number JP22fk0210075h0003.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data might be available from the authors of the cited papers.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kanda, T.; Sasaki, R.; Masuzaki, R.; Takahashi, H.; Mizutani, T.; Matsumoto, N.; Nirei, K.; Moriyama, M. Co-Occurrence of Hepatitis A Infection and Chronic Liver Disease. Int. J. Mol. Sci. 2020, 21, 6384. [Google Scholar] [CrossRef] [PubMed]
  2. Jindal, A.; Sarin, S.K. Epidemiology of liver failure in Asia-Pacific region. Liver Int. 2022. Online ahead of print. [Google Scholar] [CrossRef]
  3. Drexler, J.F.; Corman, V.M.; Lukashev, A.N.; van den Brand, J.M.; Gmyl, A.P.; Brünink, S.; Rasche, A.; Seggewiβ, N.; Feng, H.; Leijten, L.M.; et al. Evolutionary origins of hepatitis A virus in small mammals. Proc. Natl. Acad. Sci. USA 2015, 112, 15190–15195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kanda, T.; Zhang, B.; Kusov, Y.; Yokosuka, O.; Gauss-Müller, V. Suppression of hepatitis A virus genome translation and replication by siRNAs targeting the internal ribosomal entry site. Biochem. Biophys. Res. Commun. 2005, 330, 1217–1223. [Google Scholar] [CrossRef] [PubMed]
  5. Kanda, T.; Yokosuka, O.; Imazeki, F.; Fujiwara, K.; Nagao, K.; Saisho, H. Amantadine inhibits hepatitis A virus internal ribosomal entry site-mediated translation in human hepatoma cells. Biochem. Biophys. Res. Commun. 2005, 331, 619–621. [Google Scholar] [CrossRef] [PubMed]
  6. Kanda, T.; Gauss-Müller, V.; Cordes, S.; Tamura, R.; Okitsu, K.; Shuang, W.; Nakamoto, S.; Fujiwara, K.; Imazeki, F.; Yokosuka, O. Hepatitis A virus (HAV) proteinase 3C inhibits HAV IRES-dependent translation and cleaves the polypyrimidine tract-binding protein. J. Viral Hepat. 2010, 17, 618–623. [Google Scholar] [CrossRef] [PubMed]
  7. Kanda, T.; Imazeki, F.; Nakamoto, S.; Okitsu, K.; Fujiwara, K.; Yokosuka, O. Internal ribosomal entry-site activities of clinical isolate-derived hepatitis A virus and inhibitory effects of amantadine. Hepatol. Res. 2010, 40, 415–423. [Google Scholar] [CrossRef] [PubMed]
  8. Wu, S.; Nakamoto, S.; Kanda, T.; Jiang, X.; Nakamura, M.; Miyamura, T.; Shirasawa, H.; Sugiura, N.; Takahashi-Nakaguchi, A.; Gonoi, T.; et al. Ultra-deep sequencing analysis of the hepatitis A virus 5′-untranslated region among cases of the same outbreak from a single source. Int. J. Med. Sci. 2013, 11, 60–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Sasaki-Tanaka, R.; Shibata, T.; Okamoto, H.; Moriyama, M.; Kanda, T. Favipiravir Inhibits Hepatitis A Virus Infection in Human Hepatocytes. Int. J. Mol. Sci. 2022, 23, 2631. [Google Scholar] [CrossRef] [PubMed]
  10. Sasaki-Tanaka, R.; Nagulapalli Venkata, K.C.; Okamoto, H.; Moriyama, M.; Kanda, T. Evaluation of Potential Anti-Hepatitis A Virus 3C Protease Inhibitors Using Molecular Docking. Int. J. Mol. Sci. 2022, 23, 6044. [Google Scholar] [CrossRef] [PubMed]
  11. Kanda, T.; Sasaki, R.; Masuzaki, R.; Matsumoto, N.; Ogawa, M.; Moriyama, M. Cell Culture Systems and Drug Targets for Hepatitis A Virus Infection. Viruses 2020, 12, 533. [Google Scholar] [CrossRef] [PubMed]
  12. Kanda, T.; Jeong, S.H.; Imazeki, F.; Fujiwara, K.; Yokosuka, O. Analysis of 5′ nontranslated region of hepatitis A viral RNA genotype I from South Korea: Comparison with disease severities. PLoS ONE 2010, 5, e15139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Ogawa, M.; Kanda, T.; Suganami, A.; Nakamoto, S.; Win, N.N.; Tamura, Y.; Nakamura, M.; Matsuoka, S.; Yokosuka, O.; Kato, N.; et al. Antiviral activity of zinc sulfate against hepatitis A virus replication. Future Virol. 2019, 14, 399–406. [Google Scholar] [CrossRef]
  14. Kanda, T.; Sasaki, R.; Masuzaki, R.; Takahashi, H.; Fujisawa, M.; Matsumoto, N.; Okamoto, H.; Moriyama, M. Additive Effects of Zinc Chloride on the Suppression of Hepatitis A Virus Replication by Interferon in Human Hepatoma Huh7 Cells. In Vivo 2020, 34, 3301–3308. [Google Scholar] [CrossRef] [PubMed]
  15. Kanda, T.; Sasaki-Tanaka, R.; Masuzaki, R.; Matsumoto, N.; Okamoto, H.; Moriyama, M. Knockdown of Mitogen-Activated Protein Kinase 3 Negatively Regulates Hepatitis A Virus Replication. Int. J. Mol. Sci. 2021, 22, 7420. [Google Scholar] [CrossRef]
  16. Ghasemnejad-Berenji, M.; Pashapour, S. Favipiravir and COVID-19: A Simplified Summary. Drug Res. 2021, 71, 166–170. [Google Scholar] [CrossRef] [PubMed]
  17. Tréguier, Y.; Bull-Maurer, A.; Roingeard, P. Apolipoprotein E, a Crucial Cellular Protein in the Lifecycle of Hepatitis Viruses. Int. J. Mol. Sci. 2022, 23, 3676. [Google Scholar] [CrossRef] [PubMed]
  18. Perrin-Cocon, L.; Kundlacz, C.; Jacquemin, C.; Hanoulle, X.; Aublin-Gex, A.; Figl, M.; Manteca, J.; André, P.; Vidalain, P.-O.; Lotteau, V.; et al. Domain 2 of Hepatitis C Virus Protein NS5A Activates Glucokinase and Induces Lipogenesis in Hepatocytes. Int. J. Mol. Sci. 2022, 23, 919. [Google Scholar] [CrossRef] [PubMed]
  19. Kulsuptrakul, J.; Wang, R.; Meyers, N.L.; Ott, M.; Puschnik, A.S. A genome-wide CRISPR screen identifies UFMylation and TRAMP-like complexes as host factors required for hepatitis A virus infection. Cell Rep. 2021, 34, 108859. [Google Scholar] [CrossRef] [PubMed]
  20. Lesnova, E.I.; Masalova, O.V.; Permyakova, K.Y.; Kozlov, V.V.; Nikolaeva, T.N.; Pronin, A.V.; Valuev-Elliston, V.T.; Ivanov, A.V.; Kushch, A.A. Difluoromethylornithine (DFMO), an Inhibitor of Polyamine Biosynthesis, and Antioxidant N-Acetylcysteine Potentiate Immune Response in Mice to the Recombinant Hepatitis C Virus NS5B Protein. Int. J. Mol. Sci. 2021, 22, 6892. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Hepatitis A virus (HAV) infection into hepatocytes of the liver. HAV internal ribosomal entry-site (IRES), 3C protease, and 3D ribonucleic acid (RNA)-dependent RNA polymerase are attractive targets of anti-HAV drugs [11]. HAV 5′-untranslated region (UTR) has conservative RNA sequences among different strains [8,12].
Figure 1. Hepatitis A virus (HAV) infection into hepatocytes of the liver. HAV internal ribosomal entry-site (IRES), 3C protease, and 3D ribonucleic acid (RNA)-dependent RNA polymerase are attractive targets of anti-HAV drugs [11]. HAV 5′-untranslated region (UTR) has conservative RNA sequences among different strains [8,12].
Ijms 23 07214 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kanda, T.; Sasaki-Tanaka, R.; Nakamoto, S. Hepatitis A Virus Infection and Molecular Research. Int. J. Mol. Sci. 2022, 23, 7214. https://doi.org/10.3390/ijms23137214

AMA Style

Kanda T, Sasaki-Tanaka R, Nakamoto S. Hepatitis A Virus Infection and Molecular Research. International Journal of Molecular Sciences. 2022; 23(13):7214. https://doi.org/10.3390/ijms23137214

Chicago/Turabian Style

Kanda, Tatsuo, Reina Sasaki-Tanaka, and Shingo Nakamoto. 2022. "Hepatitis A Virus Infection and Molecular Research" International Journal of Molecular Sciences 23, no. 13: 7214. https://doi.org/10.3390/ijms23137214

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop