#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Cross-Serotype Immunity Induced by Immunization with a Conserved Rhinovirus Capsid Protein


Human rhinovirus (RV) infections are the principle cause of common colds and precipitate asthma and COPD exacerbations. There is currently no RV vaccine, largely due to the existence of ∼150 strains. We aimed to define highly conserved areas of the RV proteome and test their usefulness as candidate antigens for a broadly cross-reactive vaccine, using a mouse infection model. Regions of the VP0 (VP4+VP2) capsid protein were identified as having high homology across RVs. Immunization with a recombinant VP0 combined with a Th1 promoting adjuvant induced systemic, antigen specific, cross-serotype, cellular and humoral immune responses. Similar cross-reactive responses were observed in the lungs of immunized mice after infection with heterologous RV strains. Immunization enhanced the generation of heterosubtypic neutralizing antibodies and lung memory T cells, and caused more rapid virus clearance. Conserved domains of the RV capsid therefore induce cross-reactive immune responses and represent candidates for a subunit RV vaccine.


Vyšlo v časopise: Cross-Serotype Immunity Induced by Immunization with a Conserved Rhinovirus Capsid Protein. PLoS Pathog 9(9): e32767. doi:10.1371/journal.ppat.1003669
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003669

Souhrn

Human rhinovirus (RV) infections are the principle cause of common colds and precipitate asthma and COPD exacerbations. There is currently no RV vaccine, largely due to the existence of ∼150 strains. We aimed to define highly conserved areas of the RV proteome and test their usefulness as candidate antigens for a broadly cross-reactive vaccine, using a mouse infection model. Regions of the VP0 (VP4+VP2) capsid protein were identified as having high homology across RVs. Immunization with a recombinant VP0 combined with a Th1 promoting adjuvant induced systemic, antigen specific, cross-serotype, cellular and humoral immune responses. Similar cross-reactive responses were observed in the lungs of immunized mice after infection with heterologous RV strains. Immunization enhanced the generation of heterosubtypic neutralizing antibodies and lung memory T cells, and caused more rapid virus clearance. Conserved domains of the RV capsid therefore induce cross-reactive immune responses and represent candidates for a subunit RV vaccine.


Zdroje

1. MakelaMJ, PuhakkaT, RuuskanenO, LeinonenM, SaikkuP, et al. (1998) Viruses and bacteria in the etiology of the common cold. J Clin Microbiol 36: 539–542.

2. JohnstonSL, PattemorePK, SandersonG, SmithS, LampeF, et al. (1995) Community study of role of viral infections in exacerbations of asthma in 9–11 year old children. Bmj 310: 1225–1229.

3. NicholsonKG, KentJ, IrelandDC (1993) Respiratory viruses and exacerbations of asthma in adults. Bmj 307: 982–986.

4. PapiA, BellettatoCM, BraccioniF, RomagnoliM, CasolariP, et al. (2006) Infections and airway inflammation in chronic obstructive pulmonary disease severe exacerbations. Am J Respir Crit Care Med 173: 1114–1121.

5. AlperCM, DoyleWJ, SkonerDP, BuchmanCA, SerokyJT, et al. (1996) Prechallenge antibodies: moderators of infection rate, signs, and symptoms in adults experimentally challenged with rhinovirus type 39. Laryngoscope 106: 1298–1305.

6. AlperCM, DoyleWJ, SkonerDP, BuchmanCA, CohenS, et al. (1998) Prechallenge antibodies moderate disease expression in adults experimentally exposed to rhinovirus strain hanks. Clin Infect Dis 27: 119–128.

7. BarclayWS, al-NakibW, HigginsPG, TyrrellDA (1989) The time course of the humoral immune response to rhinovirus infection. Epidemiol Infect 103: 659–669.

8. PerkinsJC, TuckerDN, KnopeHL, WenzelRP, HornickRB, et al. (1969) Evidence for protective effect of an inactivated rhinovirus vaccine administered by the nasal route. Am J Epidemiol 90: 319–326.

9. MitchisonDA (1965) Prevention of Colds by Vaccination against a Rhinovirus: A Report by the Scientific Committee on Common Cold Vaccines. Br Med J 1: 1344–1349.

10. HamparianVV, ColonnoRJ, CooneyMK, DickEC, GwaltneyJMJr, et al. (1987) A collaborative report: rhinoviruses–extension of the numbering system from 89 to 100. Virology 159: 191–192.

11. PalmenbergAC, SpiroD, KuzmickasR, WangS, DjikengA, et al. (2009) Sequencing and analyses of all known human rhinovirus genomes reveal structure and evolution. Science 324: 55–59.

12. UncapherCR, DeWittCM, ColonnoRJ (1991) The major and minor group receptor families contain all but one human rhinovirus serotype. Virology 180: 814–817.

13. BochkovYA, PalmenbergAC, LeeWM, RatheJA, AminevaSP, et al. (2011) Molecular modeling, organ culture and reverse genetics for a newly identified human rhinovirus C. Nat Med 17: 627–632.

14. LauSK, YipCC, TsoiHW, LeeRA, SoLY, et al. (2007) Clinical features and complete genome characterization of a distinct human rhinovirus (HRV) genetic cluster, probably representing a previously undetected HRV species, HRV-C, associated with acute respiratory illness in children. J Clin Microbiol 45: 3655–3664.

15. GilbertSC (2012) T-cell-inducing vaccines - what's the future. Immunology 135: 19–26.

16. HillaireML, OsterhausAD, RimmelzwaanGF (2011) Induction of virus-specific cytotoxic T lymphocytes as a basis for the development of broadly protective influenza vaccines. J Biomed Biotechnol 2011: 939860.

17. LeeLY, Ha doLA, SimmonsC, de JongMD, ChauNV, et al. (2008) Memory T cells established by seasonal human influenza A infection cross-react with avian influenza A (H5N1) in healthy individuals. J Clin Invest 118: 3478–3490.

18. RichardsKA, TophamD, ChavesFA, SantAJ (2010) Cutting edge: CD4 T cells generated from encounter with seasonal influenza viruses and vaccines have broad protein specificity and can directly recognize naturally generated epitopes derived from the live pandemic H1N1 virus. J Immunol 185: 4998–5002.

19. GernJE, DickEC, KellyEA, VrtisR, KleinB (1997) Rhinovirus-specific T cells recognize both shared and serotype-restricted viral epitopes. J Infect Dis 175: 1108–1114.

20. WimalasunderaSS, KatzDR, ChainBM (1997) Characterization of the T cell response to human rhinovirus in children: implications for understanding the immunopathology of the common cold. J Infect Dis 176: 755–759.

21. HastingsGZ, FrancisMJ, RowlandsDJ, ChainBM (1993) Epitope analysis of the T cell response to a complex antigen: proliferative responses to human rhinovirus capsids. Eur J Immunol 23: 2300–2305.

22. ParryDE, BusseWW, SukowKA, DickCR, SwensonC, et al. (2000) Rhinovirus-induced PBMC responses and outcome of experimental infection in allergic subjects. J Allergy Clin Immunol 105: 692–698.

23. BartlettNW, WaltonRP, EdwardsMR, AniscenkoJ, CaramoriG, et al. (2008) Mouse models of rhinovirus-induced disease and exacerbation of allergic airway inflammation. Nat Med 14: 199–204.

24. EichelbergerM, AllanW, ZijlstraM, JaenischR, DohertyPC (1991) Clearance of influenza virus respiratory infection in mice lacking class I major histocompatibility complex-restricted CD8+ T cells. J Exp Med 174: 875–880.

25. HussellT, OpenshawPJ (1998) Intracellular IFN-gamma expression in natural killer cells precedes lung CD8+ T cell recruitment during respiratory syncytial virus infection. J Gen Virol 79 (Pt 11) 2593–2601.

26. HouS, DohertyPC, ZijlstraM, JaenischR, KatzJM (1992) Delayed clearance of Sendai virus in mice lacking class I MHC-restricted CD8+ T cells. J Immunol 149: 1319–1325.

27. HeidemaJ, RossenJW, LukensMV, KetelMS, ScheltensE, et al. (2008) Dynamics of human respiratory virus-specific CD8+ T cell responses in blood and airways during episodes of common cold. J Immunol 181: 5551–5559.

28. BrienJD, UhrlaubJL, Nikolich-ZugichJ (2008) West Nile virus-specific CD4 T cells exhibit direct antiviral cytokine secretion and cytotoxicity and are sufficient for antiviral protection. J Immunol 181: 8568–8575.

29. BrownDM, DilzerAM, MeentsDL, SwainSL (2006) CD4 T cell-mediated protection from lethal influenza: perforin and antibody-mediated mechanisms give a one-two punch. J Immunol 177: 2888–2898.

30. WilkinsonTM, LiCK, ChuiCS, HuangAK, PerkinsM, et al. (2012) Preexisting influenza-specific CD4(+) T cells correlate with disease protection against influenza challenge in humans. Nat Med 18: 274–280.

31. McKinstryKK, StruttTM, KuangY, BrownDM, SellS, et al. (2012) Memory CD4+ T cells protect against influenza through multiple synergizing mechanisms. J Clin Invest 122: 2847–2856.

32. StruttTM, McKinstryKK, DibbleJP, WinchellC, KuangY, et al. (2010) Memory CD4+ T cells induce innate responses independently of pathogen. Nat Med 16: 558–564 551p following 564.

33. MessageSD, Laza-StancaV, MalliaP, ParkerHL, ZhuJ, et al. (2008) Rhinovirus-induced lower respiratory illness is increased in asthma and related to virus load and Th1/2 cytokine and IL-10 production. Proc Natl Acad Sci U S A 105: 13562–13567.

34. O'HaganDT, RappuoliR, De GregorioE, TsaiT Del Giudice G MF59 adjuvant: the best insurance against influenza strain diversity. Expert Rev Vaccines 10: 447–462.

35. NazarethI, TavaresF, RosillonD, HaguinetF, BauchauV (2013) Safety of AS03-adjuvanted split-virion H1N1 (2009) pandemic influenza vaccine: a prospective cohort study. BMJ Open 3 (2) pii: e001912.

36. McLeanGR, WaltonRP, ShettyS, PaktiawalN, KebadzeT, et al. (2012) Rhinovirus infections and immunisation induce cross-serotype reactive antibodies to VP1. Antiviral Res 95: 193–201.

37. MalliaP, MessageSD, GielenV, ContoliM, GrayK, et al. (2011) Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med 183: 734–742.

38. ChapmanTJ, LambertK, TophamDJ (2011) Rapid reactivation of extralymphoid CD4 T cells during secondary infection. PLoS One 6: e20493.

39. TeijaroJR, TurnerD, PhamQ, WherryEJ, LefrancoisL, et al. (2011) Cutting edge: Tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J Immunol 187: 5510–5514.

40. TurnerRB (1997) Epidemiology, pathogenesis, and treatment of the common cold. Ann Allergy Asthma Immunol 78: 531–539 quiz 539-540.

41. EdgarRC (2004) MUSCLE: multiple sequence alignment with high accuracy and high throughput. Nucleic Acids Res 32: 1792–1797.

42. GaltierN, GouyM, GautierC (1996) SEAVIEW and PHYLO_WIN: two graphic tools for sequence alignment and molecular phylogeny. Comput Appl Biosci 12: 543–548.

43. ClampM, CuffJ, SearleSM, BartonGJ (2004) The Jalview Java alignment editor. Bioinformatics 20: 426–427.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#