Browse > Article
http://dx.doi.org/10.5352/JLS.2020.30.4.402

Intracellular Signaling Pathway for Host Defense Mechanisms against Piscine Nervous Necrosis Virus (NNV)  

Kim, Jong-Oh (Institute of Marine Biotechnology, Pukyong National University)
Publication Information
Journal of Life Science / v.30, no.4, 2020 , pp. 402-409 More about this Journal
Abstract
Nervous necrosis virus (NNV) contains a bi-segmented viral genome, RNA1 (3.4 kb, RdRp), and RNA2 (1.4 kb, capsid protein) in a small particle (25 nm). Despite its extremely compact size, NNV has caused serious damage by infecting approximately 120 fish species worldwide since it was first reported in the late 1980s. In order to minimize the damage caused by NNV infection and develop effective vaccines, it is necessary to understand the intra cellular signaling system according to NNV infection. NNV infection induces cell cycle arrest at the G1 phase via the p53-dependent pathway to use the cellular system for its replication. Otherwise, host cells recognize NNV infection through the RIG-1-like receptor (RLR) signaling pathway to control the virus and infected cells, and then ISGs required for antiviral action are activated via the IFN signaling pathway. Moreover, apoptosis of infected cells is triggered by the unfolded protein response (UPR) through ER stress and mitochondria-mediated cell death. Cell signaling studies on the NNV infection mechanisms are still at an early stage and many pathways have yet to be identified. Understanding the various disease-specific cellular signaling systems associated with NNV infection is essential for rapid and accurate diagnosis and vaccine development.
Keywords
Cell signaling pathway; defense mechanisms; nervous necrosis virus, NNV;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Davy, C. and Doorbar, J. 2007. G2/M cell cycle arrest in the life cycle of viruses. Virology 368, 219-226.   DOI
2 Egusa, S., Wakabayashi, H. and Muroga, K. 2006. Infectious and parasitic diseases of fish and shellfish. Life Science Publishing Co., Seoul, Korea.
3 Fenner, B. J., Thiagarajan, R., Chua, H. K. and Kwang, J. 2006a. Betanodavirus B2 is an RNA interference antagonist that facilitates intracellular viral RNA accumulation. J. Virol. 80, 85-94.   DOI
4 Fuchs, Y. and Steller, H. 2015. Live to die another way: modes of programmed cell death and the signals emanating from dying cells. Nat. Rev. Mol. Cell. Biol. 16, 329-344.   DOI
5 Fukuda, Y., Nguyen, H. D., Furuhashi, M. and Nakai, T. 1996 Mass mortality of cultured sevenband grouper, Epinephelus septemfasciatus, associated with viral nervous necrosis. Fish Pathol. 31, 165-170.   DOI
6 Gan, Z., Chen, S. N., Huang, B., Zou, J. and Nie, P. 2019. Fish type I and type II interferons: composition, receptor usage, production and function. Rev. Aquacult. 49, 1-32.
7 Garcia-Sastre, A. 2017. Ten strategies of interferon evasion by viruses. Cell Host Microbe. 22, 176-184.   DOI
8 Ge, H., Lin, K., Zhou, C., Lin, Q., Zhang, Z, Wu, J, Zheng, L., Yang, Q., Wu, S., Chen, W. and Wang, Y. 2020. A multi-omic analysis of orange-spotted grouper larvae infected with nervous necrosis virus identifies increased adhesion molecules and collagen synthesis in the persistent state. Fish Shellfish Immunol. 98, 595-604.   DOI
9 Johansen, R., Sommerset, I., Torud, B., Korsnes, K., Hjortaas, M. J., Nilsen, F., Nerland, A. H. and Dannevig, B. H. 2004. Characterization of nodavirus and viral encephalopathy and retinopathy in farmed turbot, Scophthalmus maximus (L.). J. Fish Dis. 27, 591-601.   DOI
10 Gomez, D. K., Baeck, G. W., Kim, J. H., Choresca, C. H. Jr. and Park, S. C. 2008. Molecular detection of betanodavirus in wild marine fish populations in South Korea. J. Vet. Diagn. Invest. 20, 38-44.   DOI
11 Kim, C. S., Kim, W. S., Nishizawa, T. and Oh, M. J. 2012. Prevalence of viral nervous necrosis (VNN) in sevenband grouper, Epinephelus septemfasciatus farms. J. Fish Pathol. 25, 111-116.   DOI
12 Kim, J. O., Kim, J. O., Kim, W. S. and Oh, M. J. 2017. Characterization of the transcriptome and gene expression of brain tissue in sevenband grouper (Hyporthodus septemfasciatus) in response to NNV infection. Genes 8, 31-43.   DOI
13 Krishnan, R., Girish, B. P., Jeena, K., Tripathi, G. and Pani, P. K. 2018. Molecular characterization, ontogeny and expression profiling of mitochondrial antiviral signaling adapter, MAVS from Asian seabass Lates calcarifer, Bloch (1790). Dev. Comp. Immunol. 79, 175-185.   DOI
14 Krishnan, R., Jeena, K., Mushtaq, Z., Shyam, K. U. and Kurcheti, P. P. 2018. Antiviral activity of transiently expressed mitochondrial antiviral signaling adapter, MAVS orthologue from Asian seabass. Fish Shellfish Immunol. 76, 183-186.   DOI
15 Loo, Y. M. and Gale, M. Jr. 2011. Immune signaling by RIG-I-like receptors. Immunity 34, 680-692.   DOI
16 Moore, C. B. and Ting, J. Y. 2008. Regulation of mitochondrial antiviral signaling pathways. Immunity 28, 735-739.   DOI
17 Maekawa, S., Aoki, T. and Wang, H. C. 2017. Constitutive overexpressed type I interferon induced downregulation of antiviral activity in medaka fish (Oryzias latipes). Dev. Comp. Immunol. 68, 12-20.   DOI
18 Mai, W., Liu, H., Chen, H., Zhou, Y. and Chen, Y. 2018. RGNNV-induced cell cycle arrest at G1/S phase enhanced viral replication via p53-dependent pathway in GS cells. Virus Res. 256, 142-152.   DOI
19 Michael, P., Brabant, D., Bleiblo, F., Ramana, C. V., Rutherford, M., Khurana, S., Tai, T. C., Kurmar, A. and Kumar, A. 2013. Influenza A induced cellular signal transduction pathways. J. Thorac. Dis. 5, S132-141.
20 Munday, B. L. and Nakai, T. 1997. Special topic review: nodaviruses as pathogens in larval and juvenile marine finfish. World J. Microb. Biot. 13, 375-381.   DOI
21 Munday, B. L., Kwang, J. and Moody, N. 2002. Betanodavirus infections of teleost fish: a review. J. Fish Dis. 25, 127-142.   DOI
22 Nishizawa, T., Mori, K. I., Furuhashi, M., Nakai, T., Furusawa, I. and Muroga, K. 1995. Five fish nodaviruses, the causative agents of viral nervous necrosis in marine fish. J. Gen. Virol. 76, 1563-1569.   DOI
23 Paladino, P., Marcon, E., Greenblatt, J. and Frappier, L. 2014. Identification of herpesvirus proteins that contribute to G1/S arrest. J. Virol. 88, 4480-4492.   DOI
24 Platanias, L. C. 2003. The p38 mitogen-activated protein kinase. Pharmacol. Ther. 98, 129-142.   DOI
25 Su, Y. C., Wu, J. L. and Hong, J. R. 2009. Betanodavirus non-structural protein B2: A novel necrotic death factor that induces mitochondria-mediated cell death in fish cells. Virology 385, 143-154.   DOI
26 Platanias, L. C. 2005. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 5, 375-386.   DOI
27 Ranjan, P., Bowzard, J. B., Schwerzmann, J. W., Jeisy-Scott, V., Fujita, T. and Sambhara, S. 2009. Cytoplasmic nucleic acid sensors in antiviral immunity. Trends Mol. Med. 15, 359-368.   DOI
28 Schneemann, A., Ball, L. A., Delsert, C., Johnson, J. E. and Nishizawa, T. 2005. Family Nodaviridae. Virus taxonomy: Eighth report of the international committee on taxonomy of viruses. CM, Mayo MA, Maniloff J, Desselberger U and Ball LA, eds. Elsevier/Academic Press, London, United Kingdom. Fauquet 869-872.
29 Sommerset, I. and Nerland, A. H. 2004. Complete sequence of RNA1 and subgenomic RNA3 of Atlantic halibut nodavirus (AHNV). Dis. Aquat. Organ. 58, 117-125.   DOI
30 Su, Y. C., Chiu, H. W., Hung, J. C. and Hong, J. R. 2014. Beta-nodavirus B2 protein induces hydrogen peroxide production, leading to Drp1-recruited mitochondrial fragmentation and cell death via mitochondrial targeting. Apoptosis 19, 1457-1470.   DOI
31 Su, Y. C., Wu, J. L. and Hong, J. R. 2011. Betanodavirus up-regulates chaperone GRP78 via ER stress: roles of GRP78 in viral replication and host mitochondria- mediated cell death. Apoptosis 16, 272-287.   DOI
32 Tan, C., Huang, B., Chang, S. F., Ngoh, G. H., Munday, B., Chen, S. C. and Kwang, J. 2001. Determination of the complete nucleotide sequences of RNA1 and RNA2 from greasy grouper (Epinephelus tauvina) nervous necrosis virus, Singapore strain. J. Gen Virol. 82, 647-653.   DOI
33 Akira, S., Uematsu, S. and Takeuchi, O. 2006. Pathogen recognition and innate immunity. Cell 124, 783-801.   DOI
34 Tso, C. H., Hung, Y. F., Tan, S. P. and Lu, M. W. 2013. Identification of the STAT1 gene and the characterisation of its immune response to immunostimulants, including nervous necrosis virus (NNV) infection, in Malabar grouper (Epinephelus malabaricus). Fish Shellfish Immunol. 35, 1339-1348.   DOI
35 Uddin, S., Yenush, L., Sun, X. J., Sweet, M. E., White, M. F. and Platanias, L. C. 1995. Interferon-$\alpha$ engages the insulin receptor substrate-1 to associate with the phosphatidylinositol 3'-kinase. J. Biol. Chem. 270, 15938-159341.   DOI
36 Valero, Y., Morcillo, P., Meseguer, J., Buonocore, F., Esteban, M. A., Chaves-Pozo, E. and Cuesta, A. 2015. Characterization of the IFN pathway in the teleost fish gonad against vertically transmitted viral nervous necrosis virus. J. Gen. Virol. 96, 2176-2187.   DOI
37 Wu, H. C., Wu, J. L., Chu, H. L., Su, Y. C. and Hong, J. R. 2010. RGNNV induces mitochondria-mediated cell death via newly synthesized protein dependent pathway in fish cells. Fish Shellfish Immunol. 29, 451-463.   DOI
38 Wu, Y. C., Lu, Y. F. and Chi, S. C. 2010. Anti-viral mechanism of barramundi Mx against beta-nodavirus involves the inhibition of viral RNA synthesis through the interference of RdRp. Fish Shellfish Immunol. 28, 467-475.   DOI
39 Xiang, Y., Jia, P., Liu, W., Yi, M. and Jia, K. 2019. Comparative transcriptome analysis reveals the role of p53 signalling pathway during red-spotted grouper nervous necrosis virus infection in Lateolabrax japonicus brain cells. J. Fish Dis. 42, 585-595.   DOI
40 Adams, J. M. and Cory, S. 1998. The Bcl-2 protein family: arbiters of cell survival. Science 281, 1322-136   DOI
41 Bondad-Reantaso, M. G., Subasinghe, R. P., Arthur, J. R., Ogawa, K., Chinabut, S., Adlard, R., Tan, Z. and Shariff, M. 2005. Disease and health management in Asian aquaculture. Vet. Parasitol. 132, 249-272.   DOI
42 Chen, S. P., Yang, H. L., Her, G. M., Lin, H. Y., Jeng, M. F., Wu, J. L. and Hong, J. R. 2006. Betanodavirus induces phosphatidylserine exposure and loss of mitochondrial membrane potential in secondary necrotic cells, both of which are blocked by bongkrekic acid. Virology 347, 379-391.   DOI
43 Bruns, A. M. and Horvath, C. M. 2014. Antiviral RNA recognition and assembly by RLR family innate immune sensors. Cytokine Growth F. R. 25, 507-512.   DOI
44 Chaves-Pozo, E., Bandin, I., Olveira, J. G., Esteve-Codina, A., Gomez-Garrido, J., Dabad, M., Alioto, T., Esteban, M. A. and Cuesta, A. 2019. European sea bass brain DLB-1 cell line is susceptible to nodavirus: A transcriptomic study. Fish Shellfish Immunol. 86, 14-24.   DOI
45 Chen, L. J., Su, Y. C. and Hong, J. R. 2009. Betanodavirus non-structural protein B1: A novel anti-necrotic death factor that modulates cell death in early replication cycle in fish cells. Virology 385, 444-454.   DOI
46 Darnell, J. E., Kerr, I. M. and Stark, G. R. 1994. A comprehensive review of the IFN-activated JAK- STAT-signalling pathways. Science 264, 1415-1420.   DOI
47 Costa, J. Z. and Thompson, K. D. 2016. Understanding the interaction between Betanodavirus and its host for the development of prophylactic measures for viral encephalopathy and retinopathy. Fish Shellfish Immun. 53, 35-49.   DOI