DMO-CAP inhibits influenza virus replication by activating heme oxygenase-1-mediated IFN response

Ming Zhong, Huiqiang Wang, Linlin Ma, Haiyan Yan, Shuo Wu, Zhengyi Gu, Yuhuan Li, Ming Zhong, Huiqiang Wang, Linlin Ma, Haiyan Yan, Shuo Wu, Zhengyi Gu, Yuhuan Li

Abstract

Background: As a leading cause of respiratory disease, influenza A virus (IAV) infection remains a pandemic threat in annual seasonal outbreaks. Given the limitation of existing anti-influenza therapeutic drugs, development of new drugs is urgently required. Flavonoids extracted from Artemisia rupestris L. have an inhibitory effect on virus infections. Despite this fact, the antiviral properties of 6-demethoxy-4'-O-methylcapillarisin (DMO-CAP), one of such flavonoids, against the influenza virus have not been reported. Thus, the aim of this study is to investigate the anti-IAV virus efficacy and antiviral mechanism of DMO-CAP.

Methods: The inhibitory activity of DMO-CAP against IAV was detected in vitro using viral titers by Western blot analysis, qRT-PCR, and immunofluorescence assays. The mechanism of DMO-CAP against influenza virus was analyzed by Western blot analysis, qRT-PCR, and luciferase assay.

Results: DMO-CAP exhibits broad spectrum of antiviral activities against IAV in vitro. Mechanistically, DMO-CAP treatment induced the phosphorylation of p38 mitogen-activated protein kinase (MAPK), JNK MAPK, and ERK MAPK, which led to the activation of Nrf2/heme oxygenase-1 (HO-1) pathway. Then, the up-regulation of HO-1 expression activated the IFN response and induced the expression of IFN-stimulated genes, thereby leading to efficient anti-IAV effects.

Conclusions: DMO-CAP inhibited IAV replication by activating HO-1-mediated IFN response. DMO-CAP may be a potential agent or supplement against IAV infection.

Conflict of interest statement

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors have declared that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
The structure and cytotoxicity of compound DMO-CAP. a The structure of DMO-CAP. b The viabilities of DMO-CAP on MDCK and RAW264.7 cells was measured by CCK assay
Fig. 2
Fig. 2
Antiviral activity of DMO-CAP against IAV. a Antiviral activity of DMO-CAP in RAW264.7 cells against A/Fort Monmouth/1/1947 (H1N1) and IAV A/Wuhan/359/1995 (H3N2) was tested by viral titers assay. b and c DMO-CAP reduced the expression of M2 RNA and protein in MDCK cells by one-step qRT-PCR assay and Western blot assay. d DMO-CAP reduced the expression of M2 protein by immunofluorescence. e DMO-CAP reduced the expression of M2 protein of influenza resistant strains by Western blot assay. Mock: normal cells without treatment; H1N1: cells were infected with IAV A/FM1/1947 at 0.01MOI; H3N2: cells were infected with IAV A/Wuhan/359/1995 (H3N2) at 0.01MOI. Con: cells were infected pathognomonic viral strains and treated with equal amounts of DMSO or DMO-CAP. The experiments were performed in triplicate and the data represents mean ± SD. ***P < 0.001, **P < 0.01, *P < 0.05 versus Con
Fig. 3
Fig. 3
DMO-CAP inhibited IAV replication through up-regulating HO-1 expression. a HEK293T-17 cells were infected with IAV A/FM1/1947 (0.2 MOI) and then treated with DMO-CAP (50 μM)、CoPP (2 μM) together with DMO-CAP (50 μM) and CoPP (2 μM) for 24 h. b HEK293T-17 cells were transfected with HO-1 siRNA or SCR siRNA for 24 h and then infected with IAV A/FM1/1947 (0.2 MOI) in the absence or presence of DMO-CAP for 24 h. The expression of M2 and HO-1 proteins were analyzed by Western blot
Fig. 4
Fig. 4
DMO-CAP activates the interferon response by stimulating the Nrf2/ARE pathway to up-regulate the expression of HO-1. a Nrf2 DNA binding activity was analyzed in HEK293T-17 cells co-transfected with pGL4.37 [luc2P/ARE/Hygro]/pAP-1-Luc/pNF-κB-Luc and pRL-SV40 vector, the results were presented as Nrf2 DNA binding activity relative to its basal levels in mock 293 T. **P < 0.01 versus Mock. b DMO-CAP promoted Nrf2 nuclear transcription. RAW264.7 cells were infected with IAV A/FM1/1947 (0.2 MOI) for 2 h and treated with 50 μM DMO-CAP for another 3 h. The total amount of cellular, cytoplasmic and nuclear Nrf2 protein were analyzed by Western blot. c DMO-CAP activated the phosphorylation of p38 MAPK, JNK MAPK and ERK MAPK. RAW264.7 cells were infected with IAV A/FM1/1947 (0.2 MOI) and treated with 50μΜ DMO-CAP for 15 min and then phospho-p38、phospho-JNK and phospho-ERK proteins were valued by Western blot. d RAW264.7 cells were infected with IAV A/FM1/1947 (0.2 MOI) for 2 h, followed by treating with or without DMO-CAP (50μΜ or 25μΜ). The mRNA level of IFN-α and IFN-β were detected by qRT-PCR assay. **P < 0.01 versus Mock. e RAW264.7 cells were infected with IAV A/FM1/1947 (0.2 MOI) for 2 h and the protein levels of ISGs were measured by Western blot after treatment with DMO-CAP for 24 h
Fig. 5
Fig. 5
Schematic showing that DMO-CAP inhibits influenza virus replication. DMO-CAP activated the MAPK pathways, thereby leading to Nrf2 expression and subsequent activation of HO-1 gene expression, as well as the up-regulation HO-1 activities host cellular type I IFN response with induction of ISGs expression, which finally leads to inhibition of influenza virus replication

References

    1. Lansbury LE, Brown CS, Nguyen JS. Influenza in long-term care facilities. Influenza Other Respir Viruses. 2017;11:356–366. doi: 10.1111/irv.12464.
    1. World Health Organization. Influenza (Seasonal) [Internet; cited 2018 Oct]. []. Accessed 6 Nov 2018.
    1. Vanderven HA, Liu L, Fernanda ASB, Nguyen TH, Wan Y, Wines B, Hogarth PM, Tilmanis D, Reynaldi A, Parsons MS, Hurt AC, Davenport MP, Kotsimbos T, Cheng AC, Kedzierska K, Zhang XY, Xu JQ, Stephen JK. Functional antibodies in humans with severe H7N9 and seasonal influenza. JCI insight. 2017;2:e92750. doi: 10.1172/jci.insight.92750.
    1. Wong SS, Webby RJ. Traditional and new influenza vaccines. Clin Microbiol Rev. 2013;26:476–492. doi: 10.1128/CMR.00097-12.
    1. Erhard MS, Pieter F, Charles B, Albert O. Influenza virus resistance to antiviral therapy. Adv Pharmacol. 2013;67:217–246. doi: 10.1016/B978-0-12-405880-4.00006-8.
    1. Koszalka P, Tilmanis D, Hurt AC. Influenza antivirals currently in late-phase clinical trial. Influenza Other Respir Viruses. 2017;11:240–246. doi: 10.1111/irv.12446.
    1. Hayden F. Baloxavir Marboxil for uncomplicated influenza in adults and adolescents. N Engl J Med. 2018;379:913–923. doi: 10.1056/NEJMoa1716197.
    1. Ison M, CAPSTONE-2 trial. Infectious Disease Week; 2018 Oct 3-7; San Francisco. CA, USA. []. Accessed 3-7 Oct 2018.
    1. Clark AM, DeDiego ML, Anderson CS, Wang J, Yang H, Nogales A, Martinez-Sobrido L, Zand MS, Sangster MY, Topham DJ. Antigenicity of the 2015-2016 seasonal H1N1 human influenza virus HA and NA proteins. PLoS One. 2017;12:e0188267. doi: 10.1371/journal.pone.0188267.
    1. Baillie JK, Paul D. Influenza-time to target the host? N Engl J Med. 2013;369:191–193. doi: 10.1056/NEJMcibr1304414.
    1. Mandal P, Roychowdhury S, Park PH, Pratt BT, Roger T, Nagy LE. Adiponectin and heme oxygenase-1 suppress TLR4/MyD88-independent signaling in rat Kupffer cells and in mice after chronic ethanol exposure. J Immunol. 2010;185:4928–4937. doi: 10.4049/jimmunol.1002060.
    1. Ma LL, Zhang P, Wang HQ, Li YF, Hu J, Jiang JD, Li YH. heme oxygenase-1 agonist CoPP suppresses influenza virus replication through IRF3-mediated generation of IFN-α/β. Virol. 2019;528:80-88.
    1. Hill BL, Halfmann P, Neumann G, Kawaoka Y. The cytoprotective enzyme heme oxygenase-1 suppresses Ebola virus replication. J Virol. 2013;87:13795–13802. doi: 10.1128/JVI.02422-13.
    1. Bunse CE, Fortmeier V, Tischer S. Modulation of heme oxygenase-1 by metalloporphyrins increases anti-viral T cell responses. Clin Exp Immunol. 2015;179:265–276. doi: 10.1111/cei.12451.
    1. Hossain MK, Saha SK, Abdal Dayem A. Bax Inhibitor-1 acts as an anti-influenza factor by inhibiting ROS mediated cell death and augmenting Heme-oxygenase 1 expression in influenza virus infected cells. Int J Mol Sci. 2018;19:712. doi: 10.3390/ijms19030712.
    1. Ma LL, Wang HQ, Wu P, Hu J, Yin JQ, Wu S, Ge M, Sun WF, Zhao JY, Aisa HA, Li YH, Jiang JD. Rupestonic acid derivative YZH-106 suppresses influenza virus replication by activation of heme oxygenase-1-mediated interferon response. Free Radic Biol Med. 2016;96:347–361. doi: 10.1016/j.freeradbiomed.2016.04.021.
    1. Cummins NW, Weaver EA, May SM. Heme oxygenase-1 regulates the immune response to influenza virus infection and vaccination in aged mice. FASEB J. 2012;26:2911–2918. doi: 10.1096/fj.11-190017.
    1. Yang L, Zhang SW, Liu ZQ, He JH, Rong XJ, Gu ZY XinJiang Medi University 2016; 5: 579.
    1. Kumar S, Pandey AK. Chemistry and biological activities of flavonoids: an overview. Sci World J. 2013;2013:162750.
    1. Liu AL, Liu B, Qin HL, Lee SM, Wang YT, Du GH. Anti-influenza virus activities of flavonoids from the medicinal plant Elsholtzia rugulosa. Planta Med. 2008;74:847–851. doi: 10.1055/s-2008-1074558.
    1. Serkedjieva J, Ivancheva S. Antiherpes virus activity of extracts from the medicinal plant Geranium sanguineum L. J Ethnopharmacol. 1998;64:59–68. doi: 10.1016/S0378-8741(98)00095-6.
    1. Hashiba T, Suzuki M, Nagashima Y, Suzuki S, Inoue S, Tsuburai T, Matsuse T, Ishigatubo Y. Adenovirus-mediated transfer of heme oxygenase-1 cDNA attenuates severe lung injury induced by the influenza virus in mice. Gene Ther. 2001;8:1499–150720. doi: 10.1038/sj.gt.3301540.
    1. Choi AM, Knobil K, Otterbein SL, Eastman DA, Jacoby DB. Oxidant stress responses in influenza virus pneumonia: gene expression and transcription factor activation. Am J Phys. 1996;271:L383–L39121.
    1. Maines MD. Heme oxygenase: function, multiplicity, regulatory mechanisms, and clinical applications. FASEB J. 1988;2:2557–256823. doi: 10.1096/fasebj.2.10.3290025.
    1. Applegate LA, Luscher P, Tyrrell RM. Induction of heme oxygenase: a general response to oxidant stress in cultured mammalian cells. Cancer Res. 1991;51:974–978.
    1. Jawed A, Daniel S, Cheri T, Sujji B, Augustine MK, Choi JL. Cook. Nrf2, a Cap’n’Collar transcription factor, regulates induction of the Heme Oxygenase-1 gene cell Microbiol. J Biol Chem. 1999;274:26071–26078. doi: 10.1074/jbc.274.37.26071.
    1. Maruyama A, Mimura J, Harada N, Itoh K. Nrf2 activation is associated with Z-DNA formation in the human HO-1 promoter. Nucleic Acids Res. 2013;41:5223–5234. doi: 10.1093/nar/gkt243.
    1. Wang B, Pakpour N, Napoli E. Anopheles stephensi p38 MAPK signaling regulates innate immunity and bioenergetics during plasmodium falciparum infection. Parasit Vectors. 2015;8:424. doi: 10.1186/s13071-015-1016-x.
    1. Taruna A, Georgia FS, Mark JE, Diane FJ. Dissociation between IFN-α-induced anti-viral and growth signaling pathways. J Immunol. 1999;162:3289–3297.
    1. Zhao M, Guo H, Chen J. 5-aminolevulinic acid combined with sodium ferrous citrate ameliorates H2O2-induced cardiomyocyte hypertrophy via activation of the MAPK/Nrf2/HO-1 pathway. ajpcell. 2015;31:00369.
    1. Tzima S, Victoratos P, Kranidioti K, Alexiou M, Kollias G. Myeloid heme oxygenase-1 regulates innate immunity and autoimmunity by modulating IFN-beta production. J Exp Med. 2009;206:1167–1179. doi: 10.1084/jem.20081582.
    1. Tseng CK, Lin CK, Wu YH. Human heme oxygenase 1 is a potential host cell factor against dengue virus replication. Sci Rep. 2016;6:32176. doi: 10.1038/srep32176.
    1. Hull TD, Agarwal A, George JF. The mononuclear phagocyte system in homeostasis and disease: a role for heme oxygenase-1. Antioxid Redox Signal. 2014;20:1770–1788. doi: 10.1089/ars.2013.5673.
    1. Kovacsics CE, Gill AJ, Ambegaokar SS, Gelman BB, Kolson DL. Degradation of heme oxygenase-1 by the immunoproteasome in astrocytes: a potential interferon-γ-dependent mechanism contributing to HIV neuropathogenesis. Glia. 2017;65:1264–1277. doi: 10.1002/glia.23160.
    1. Kesic MJ, Simmons SO, Bauer R, Jaspers I. Nrf2 expression modifies influenza a entry and replication in nasal epithelial cells. Free Radic Biol Med. 2011;51:444–453. doi: 10.1016/j.freeradbiomed.2011.04.027.
    1. Ibáñez FJ, Farías MA, Retamal-Díaz A, Espinoza JA, Kalergis AM, González PA. Pharmacological induction of Heme Oxygenase-1 impairs nuclear accumulation of herpes simplex virus capsids upon infection. Front Microbiol. 2017;8:2108. doi: 10.3389/fmicb.2017.02108.
    1. Anthony JS, Bryan RGW. Interferon-inducible antiviral effectors. Nat Rev Immunol. 2008;8:559–568. doi: 10.1038/nri2314.
    1. Traboulsi H, Cloutier A, Boyapelly K. The flavonoid Isoliquiritigenin reduces lung inflammation and mouse morbidity during influenza virus infection. Antimicrob Agents Chemother. 2015;59:6317–6327. doi: 10.1128/AAC.01098-15.
    1. Zhong M, Wang HQ, Yan HY, Wu S, Gu ZY, Li YH. Santin inhibits influenza a virus replication through regulating MAPKs and NF-κB pathways. J Asian Nat Prod Res. 2018;10:1–10. doi: 10.1080/10286020.2018.1520221.

Source: PubMed

3
구독하다