Clin Exp Vaccine Res.  2014 Jan;3(1):5-11. 10.7774/cevr.2014.3.1.5.

Inflammasomes in antiviral immunity: clues for influenza vaccine development

Affiliations
  • 1Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan. ichinohe@ims.u-tokyo.ac.jp

Abstract

Inflammasomes are cytosolic multiprotein complexes that sense microbial motifs or cellular stress and stimulate caspase-1-dependent cytokine secretion and cell death. Recently, it has become increasingly evident that both DNA and RNA viruses activate inflammasomes, which control innate and adaptive immune responses against viral infections. In addition, recent studies suggest that certain microbiota induce inflammasomes-dependent adaptive immunity against influenza virus infections. Here, we review recent advances in research into the role of inflammasomes in antiviral immunity.

Keyword

Orthomyxoviridae; Human NLRP3 protein; Metagenome; Dendritic cells

MeSH Terms

Adaptive Immunity
Cell Death
Cytosol
Dendritic Cells
DNA
Inflammasomes*
Influenza Vaccines*
Influenza, Human*
Metagenome
Microbiota
Multiprotein Complexes
Orthomyxoviridae
RNA Viruses
DNA
Inflammasomes
Influenza Vaccines
Multiprotein Complexes

Figure

  • Fig. 1 Recognition of RNA viruses by nucleotide-binding domain and leucine-rich-repeat-containing protein 3 (NLRP3) inflammasome. The NLRP3 recognizes the disturbances in intracellular ionic concentrations induced by viroporins from respiratory syncytial virus (RSV), encephalomyocarditis virus (EMCV), poliovirus, enterovirus 71 (EV71), human rhinovirus (HRV), or influenza virus. Measles virus V protein inhibits activation of NLRP3 inflammasome by interacting with the NLRP3. The RNA helicase DHX33 binds to cytosolic double-stranded RNAs (dsRNAs) to trigger NLRP3 inflammasome activation. After activation of the NLRP3, it recruits apoptosis-associated speck-like protein containing a CARD (ASC) that, in turn, recruits pro-caspase-1, which is activated by autocatalytic cleavage. Cleaved caspase-1 catalyses proteolytic processing of pro-interleukin (IL)-1β and pro-IL-18 into the active forms and stimulates their secretion. ER, endoplasmic reticulum; SH, small hydrophobic.

  • Fig. 2 Proposed model of inflammasome-dependent induction of adaptive immunity against influenza virus infection. Activation of caspase-1 in influenza virus-infected respiratory dendritic cells (DCs) stimulates secretion of interleukin-1 beta (IL-1β) and triggers a form of cells death, known as pyroptosis. Bystander DCs activated by inflammatory signals capture viral antigens (vAg) and migrate from the lung to the mediastinal lymph nodes (mLNs) to prime naïve CD8 T cells. Gut-resident microbiota provides signals leading to the expression of mRNA for pro-IL-1β, pro-IL-18, and NLRP3 at steady state. NLRP3, nucleotide-binding domain and leucine-rich-repeat-containing protein 3.


Reference

1. Gao R, Cao B, Hu Y, et al. Human infection with a novel avian-origin influenza A (H7N9) virus. N Engl J Med. 2013; 368:1888–1897.
Article
2. Neumann G, Noda T, Kawaoka Y. Emergence and pandemic potential of swine-origin H1N1 influenza virus. Nature. 2009; 459:931–939.
Article
3. Iwasaki A, Medzhitov R. Regulation of adaptive immunity by the innate immune system. Science. 2010; 327:291–295.
Article
4. Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol. 2010; 11:373–384.
Article
5. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science. 2004; 303:1529–1531.
Article
6. Lund JM, Alexopoulou L, Sato A, et al. Recognition of single-stranded RNA viruses by Toll-like receptor 7. Proc Natl Acad Sci U S A. 2004; 101:5598–5603.
Article
7. Kato H, Sato S, Yoneyama M, et al. Cell type-specific involvement of RIG-I in antiviral response. Immunity. 2005; 23:19–28.
Article
8. Rehwinkel J, Tan CP, Goubau D, et al. RIG-I detects viral genomic RNA during negative-strand RNA virus infection. Cell. 2010; 140:397–408.
Article
9. Bracci L, Canini I, Puzelli S, et al. Type I IFN is a powerful mucosal adjuvant for a selective intranasal vaccination against influenza virus in mice and affects antigen capture at mucosal level. Vaccine. 2005; 23:2994–3004.
Article
10. Ichinohe T, Iwasaki A, Hasegawa H. Innate sensors of influenza virus: clues to developing better intranasal vaccines. Expert Rev Vaccines. 2008; 7:1435–1445.
Article
11. Takada A, Matsushita S, Ninomiya A, Kawaoka Y, Kida H. Intranasal immunization with formalin-inactivated virus vaccine induces a broad spectrum of heterosubtypic immunity against influenza A virus infection in mice. Vaccine. 2003; 21:3212–3218.
Article
12. Tumpey TM, Renshaw M, Clements JD, Katz JM. Mucosal delivery of inactivated influenza vaccine induces B-cell-dependent heterosubtypic cross-protection against lethal influenza A H5N1 virus infection. J Virol. 2001; 75:5141–5150.
Article
13. Koyama S, Aoshi T, Tanimoto T, et al. Plasmacytoid dendritic cells delineate immunogenicity of influenza vaccine subtypes. Sci Transl Med. 2010; 2:25ra24.
Article
14. Ichinohe T, Watanabe I, Ito S, et al. Synthetic double-stranded RNA poly(I:C) combined with mucosal vaccine protects against influenza virus infection. J Virol. 2005; 79:2910–2919.
Article
15. Martinon F, Mayor A, Tschopp J. The inflammasomes: guardians of the body. Annu Rev Immunol. 2009; 27:229–265.
Article
16. Tschopp J, Schroder K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat Rev Immunol. 2010; 10:210–215.
Article
17. Bauernfeind F, Ablasser A, Bartok E, et al. Inflammasomes: current understanding and open questions. Cell Mol Life Sci. 2011; 68:765–783.
Article
18. Poeck H, Bscheider M, Gross O, et al. Recognition of RNA virus by RIG-I results in activation of CARD9 and inflammasome signaling for interleukin 1 beta production. Nat Immunol. 2010; 11:63–69.
Article
19. Burckstummer T, Baumann C, Bluml S, et al. An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol. 2009; 10:266–272.
Article
20. Fernandes-Alnemri T, Yu JW, Datta P, Wu J, Alnemri ES. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 2009; 458:509–513.
Article
21. Hornung V, Ablasser A, Charrel-Dennis M, et al. AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 2009; 458:514–518.
Article
22. Roberts TL, Idris A, Dunn JA, et al. HIN-200 proteins regulate caspase activation in response to foreign cytoplasmic DNA. Science. 2009; 323:1057–1060.
Article
23. Rathinam VA, Jiang Z, Waggoner SN, et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat Immunol. 2010; 11:395–402.
Article
24. Ansari MA, Singh VV, Dutta S, et al. Constitutive interferon-inducible protein 16-inflammasome activation during Epstein-Barr virus latency I, II, and III in B and epithelial cells. J Virol. 2013; 87:8606–8623.
Article
25. Kerur N, Veettil MV, Sharma-Walia N, et al. IFI16 acts as a nuclear pathogen sensor to induce the inflammasome in response to Kaposi Sarcoma-associated herpesvirus infection. Cell Host Microbe. 2011; 9:363–375.
Article
26. Johnson KE, Chikoti L, Chandran B. Herpes simplex virus 1 infection induces activation and subsequent inhibition of the IFI16 and NLRP3 inflammasomes. J Virol. 2013; 87:5005–5018.
Article
27. Ichinohe T, Pang IK, Iwasaki A. Influenza virus activates inflammasomes via its intracellular M2 ion channel. Nat Immunol. 2010; 11:404–410.
Article
28. Ito M, Yanagi Y, Ichinohe T. Encephalomyocarditis virus viroporin 2B activates NLRP3 inflammasome. PLoS Pathog. 2012; 8:e1002857.
Article
29. Triantafilou K, Kar S, van Kuppeveld FJ, Triantafilou M. Rhinovirus-induced calcium flux triggers NLRP3 and NLRC5 activation in bronchial cells. Am J Respir Cell Mol Biol. 2013; 49:923–934.
Article
30. Carter SD, Dent KC, Atkins E, et al. Direct visualization of the small hydrophobic protein of human respiratory syncytial virus reveals the structural basis for membrane permeability. FEBS Lett. 2010; 584:2786–2790.
Article
31. Triantafilou K, Kar S, Vakakis E, Kotecha S, Triantafilou M. Human respiratory syncytial virus viroporin SH: a viral recognition pathway used by the host to signal inflammasome activation. Thorax. 2013; 68:66–75.
Article
32. Mitoma H, Hanabuchi S, Kim T, et al. The DHX33 RNA helicase senses cytosolic RNA and activates the NLRP3 inflammasome. Immunity. 2013; 39:123–135.
Article
33. Netea MG, Nold-Petry CA, Nold MF, et al. Differential requirement for the activation of the inflammasome for processing and release of IL-1beta in monocytes and macrophages. Blood. 2009; 113:2324–2335.
Article
34. Wang K, Xie S, Sun B. Viral proteins function as ion channels. Biochim Biophys Acta. 2011; 1808:510–515.
Article
35. Pang IK, Iwasaki A. Control of antiviral immunity by pattern recognition and the microbiome. Immunol Rev. 2012; 245:209–226.
36. Allen IC, Scull MA, Moore CB, et al. The NLRP3 inflammasome mediates in vivo innate immunity to influenza A virus through recognition of viral RNA. Immunity. 2009; 30:556–565.
Article
37. Thomas PG, Dash P, Aldridge JR Jr, et al. The intracellular sensor NLRP3 mediates key innate and healing responses to influenza A virus via the regulation of caspase-1. Immunity. 2009; 30:566–575.
Article
38. Ichinohe T, Lee HK, Ogura Y, Flavell R, Iwasaki A. Inflammasome recognition of influenza virus is essential for adaptive immune responses. J Exp Med. 2009; 206:79–87.
Article
39. Wang L, Manji GA, Grenier JM, et al. PYPAF7, a novel PYRIN-containing Apaf1-like protein that regulates activation of NF-kappa B and caspase-1-dependent cytokine processing. J Biol Chem. 2002; 277:29874–29880.
Article
40. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002; 10:417–426.
41. Pang IK, Ichinohe T, Iwasaki A. IL-1R signaling in dendritic cells replaces pattern-recognition receptors in promoting CD8(+) T cell responses to influenza A virus. Nat Immunol. 2013; 14:246–253.
Article
42. Fernandez-Sesma A, Marukian S, Ebersole BJ, et al. Influenza virus evades innate and adaptive immunity via the NS1 protein. J Virol. 2006; 80:6295–6304.
Article
43. Smed-Sorensen A, Chalouni C, Chatterjee B, et al. Influenza A virus infection of human primary dendritic cells impairs their ability to cross-present antigen to CD8 T cells. PLoS Pathog. 2012; 8:e1002572.
Article
44. Ross TM, Xu Y, Bright RA, Robinson HL. C3d enhancement of antibodies to hemagglutinin accelerates protection against influenza virus challenge. Nat Immunol. 2000; 1:127–131.
Article
45. Liniger M, Summerfield A, Ruggli N. MDA5 can be exploited as efficacious genetic adjuvant for DNA vaccination against lethal H5N1 influenza virus infection in chickens. PLoS One. 2012; 7:e49952.
Article
46. McMahon JM, Signori E, Wells KE, Fazio VM, Wells DJ. Optimisation of electrotransfer of plasmid into skeletal muscle by pretreatment with hyaluronidase: increased expression with reduced muscle damage. Gene Ther. 2001; 8:1264–1270.
Article
47. Sasaki S, Amara RR, Oran AE, Smith JM, Robinson HL. Apoptosis-mediated enhancement of DNA-raised immune responses by mutant caspases. Nat Biotechnol. 2001; 19:543–547.
Article
48. Okuda K, Kawamoto S, Fukushima J. Cytokine and costimulatory factor-encoding plasmids as adjuvants for DNA vaccination. Methods Mol Med. 2000; 29:197–204.
Article
49. Yang ZY, Kong WP, Huang Y, et al. A DNA vaccine induces SARS coronavirus neutralization and protective immunity in mice. Nature. 2004; 428:561–564.
Article
50. Chen Z, Yoshikawa T, Kadowaki S, et al. Protection and antibody responses in different strains of mouse immunized with plasmid DNAs encoding influenza virus haemagglutinin, neuraminidase and nucleoprotein. J Gen Virol. 1999; 80(Pt 10):2559–2564.
Article
51. Nchinda G, Amadu D, Trumpfheller C, Mizenina O, Uberla K, Steinman RM. Dendritic cell targeted HIV gag protein vaccine provides help to a DNA vaccine including mobilization of protective CD8+ T cells. Proc Natl Acad Sci U S A. 2010; 107:4281–4286.
Article
52. Luckay A, Sidhu MK, Kjeken R, et al. Effect of plasmid DNA vaccine design and in vivo electroporation on the resulting vaccine-specific immune responses in rhesus macaques. J Virol. 2007; 81:5257–5269.
Article
53. Babiuk S, van Drunen Littel-van den Hurk S, Babiuk LA. Delivery of DNA vaccines using electroporation. Methods Mol Med. 2006; 127:73–82.
Article
54. Khurana S, Wu J, Dimitrova M, et al. DNA priming prior to inactivated influenza A(H5N1) vaccination expands the antibody epitope repertoire and increases affinity maturation in a boost-interval-dependent manner in adults. J Infect Dis. 2013; 208:413–417.
Article
55. Balazs AB, Chen J, Hong CM, Rao DS, Yang L, Baltimore D. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature. 2012; 481:81–84.
Article
56. Yamazaki T, Nagashima M, Ninomiya D, et al. Passive immune-prophylaxis against influenza virus infection by the expression of neutralizing anti-hemagglutinin monoclonal antibodies from plasmids. Jpn J Infect Dis. 2011; 64:40–49.
57. Balazs AB, Bloom JD, Hong CM, Rao DS, Baltimore D. Broad protection against influenza infection by vectored immunoprophylaxis in mice. Nat Biotechnol. 2013; 31:647–652.
Article
58. Limberis MP, Adam VS, Wong G, et al. Intranasal antibody gene transfer in mice and ferrets elicits broad protection against pandemic influenza. Sci Transl Med. 2013; 5:187ra72.
Article
59. Schroder K, Tschopp J. The inflammasomes. Cell. 2010; 140:821–832.
Article
60. Mann CJ, Anguela XM, Montane J, et al. Molecular signature of the immune and tissue response to non-coding plasmid DNA in skeletal muscle after electrotransfer. Gene Ther. 2012; 19:1177–1186.
Article
61. Suschak J, Wang S, Remington K, Fitzgerald K, Lu S. Involvement of the Aim2 inflammasome pathway in generating antibody responses elicited by DNA vaccination. J Immunol. 2013; 190:123.20.
62. Kamada N, Seo SU, Chen GY, Nunez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013; 13:321–335.
Article
63. Ichinohe T, Pang IK, Kumamoto Y, et al. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc Natl Acad Sci U S A. 2011; 108:5354–5359.
Article
64. Abt MC, Osborne LC, Monticelli LA, et al. Commensal bacteria calibrate the activation threshold of innate antiviral immunity. Immunity. 2012; 37:158–170.
Article
65. Ganal SC, Sanos SL, Kallfass C, et al. Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity. 2012; 37:171–186.
Article
Full Text Links
  • CEVR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr