#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Suppressor of Cytokine Signaling 4 (SOCS4) Protects against Severe Cytokine Storm and Enhances Viral Clearance during Influenza Infection


The suppressor of cytokine signaling proteins are key regulators of immunity. As yet there is no described biological role for SOCS4, despite its broad expression in cells of the immune system. Given the important role of other SOCS proteins in controlling the immune response, we have generated SOCS4-mutant mice and used a mouse influenza infection model to investigate the biological function of SOCS4. We demonstrate that mice lacking SOCS4 rapidly succumb to infection with a pathogenic H1N1 influenza virus and are hypersusceptible to infection with the less virulent H3N2 strain. This is the first demonstration of a functional phenotype in SOCS4-deficient mice. Our study reveals that in SOCS4-deficient animals, there is a dysregulated pro-inflammatory cytokine and chemokine production in the lungs and delayed viral clearance. This is associated with impaired trafficking of virus-specific CD8 T cells to the site of infection and linked to defects in T cell receptor activation. These results demonstrate that SOCS4 is a critical regulator of anti-viral immunity. Understanding the regulation of the inflammatory response to influenza is particularly relevant given the current climate concerning pandemic influenza outbreaks.


Vyšlo v časopise: Suppressor of Cytokine Signaling 4 (SOCS4) Protects against Severe Cytokine Storm and Enhances Viral Clearance during Influenza Infection. PLoS Pathog 10(5): e32767. doi:10.1371/journal.ppat.1004134
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004134

Souhrn

The suppressor of cytokine signaling proteins are key regulators of immunity. As yet there is no described biological role for SOCS4, despite its broad expression in cells of the immune system. Given the important role of other SOCS proteins in controlling the immune response, we have generated SOCS4-mutant mice and used a mouse influenza infection model to investigate the biological function of SOCS4. We demonstrate that mice lacking SOCS4 rapidly succumb to infection with a pathogenic H1N1 influenza virus and are hypersusceptible to infection with the less virulent H3N2 strain. This is the first demonstration of a functional phenotype in SOCS4-deficient mice. Our study reveals that in SOCS4-deficient animals, there is a dysregulated pro-inflammatory cytokine and chemokine production in the lungs and delayed viral clearance. This is associated with impaired trafficking of virus-specific CD8 T cells to the site of infection and linked to defects in T cell receptor activation. These results demonstrate that SOCS4 is a critical regulator of anti-viral immunity. Understanding the regulation of the inflammatory response to influenza is particularly relevant given the current climate concerning pandemic influenza outbreaks.


Zdroje

1. Newall AT, Scuttham PA, Hodgkinson B (2007) Economic Report into the cost of influenza to the Australian Health System. http://www.influenzaspecialistgrouporgau/images/stories/docs/isg_cost_influenza_report_30_2007pdf.

2. La GrutaNL, KedzierskaK, StambasJ, DohertyPC (2007) A question of self-preservation: immunopathology in influenza virus infection. Immunol Cell Biol 85: 85–92.

3. ArankalleVA, LoleKS, AryaRP, TripathyAS, RamdasiAY, et al. (2010) Role of host immune response and viral load in the differential outcome of pandemic H1N1 (2009) influenza virus infection in Indian patients. PLoS One 5: e13099.

4. ThomasP, KeatingR, Hulse-PostD, DohertyP (2006) Cell-mediated protection in influenza infection. Emerg Infect Dis 12: 48–54.

5. AlexanderWS (2002) Suppressors of cytokine signalling (SOCS) in the immune system. Nat Rev Immunol 2: 410–416.

6. HiltonDJ, RichardsonRT, AlexanderWS, VineyEM, WillsonTA, et al. (1998) Twenty proteins containing a C-terminal SOCS box form five structural classes. Proc Natl Acad Sci U S A 95: 114–119.

7. FengZP, ChandrashekaranIR, LowA, SpeedTP, NicholsonSE, et al. (2012) The N-terminal domains of SOCS proteins: a conserved region in the disordered N-termini of SOCS4 and 5. Proteins 80: 946–957.

8. ZhangJG, FarleyA, NicholsonSE, WillsonTA, ZugaroLM, et al. (1999) The conserved SOCS box motif in suppressors of cytokine signaling binds to elongins B and C and may couple bound proteins to proteasomal degradation. Proc Natl Acad Sci U S A 96: 2071–2076.

9. LinossiEM, NicholsonSE (2012) The SOCS box-adapting proteins for ubiquitination and proteasomal degradation. IUBMB Life 64: 316–323.

10. KershawNJ, MurphyJM, LiauNP, VargheseLN, LaktyushinA, et al. (2013) SOCS3 binds specific receptor-JAK complexes to control cytokine signaling by direct kinase inhibition. Nat Struct Mol Biol 20: 469–476.

11. BabonJJ, KershawNJ, MurphyJM, VargheseLN, LaktyushinA, et al. (2012) Suppression of cytokine signaling by SOCS3: characterization of the mode of inhibition and the basis of its specificity. Immunity 36: 239–250.

12. GreenhalghCJ, Rico-BautistaE, LorentzonM, ThausAL, MorganPO, et al. (2005) SOCS2 negatively regulates growth hormone action in vitro and in vivo. J Clin Invest 115: 397–406.

13. EndoT, SasakiA, MinoguchiM, JooA, YoshimuraA (2003) CIS1 interacts with the Y532 of the prolactin receptor and suppresses prolactin-dependent STAT5 activation. J Biochem 133: 109–113.

14. LavensD, MontoyeT, PiessevauxJ, ZabeauL, VandekerckhoveJ, et al. (2006) A complex interaction pattern of CIS and SOCS2 with the leptin receptor. J Cell Sci 119: 2214–2224.

15. AlexanderWS, StarrR, FennerJE, ScottCL, HandmanE, et al. (1999) SOCS1 is a critical inhibitor of interferon gamma signaling and prevents the potentially fatal neonatal actions of this cytokine. Cell 98: 597–608.

16. MetcalfD, GreenhalghCJ, VineyE, WillsonTA, StarrR, et al. (2000) Gigantism in mice lacking suppressor of cytokine signalling-2. Nature 405: 1069–1073.

17. CrokerBA, MetcalfD, RobbL, WeiW, MifsudS, et al. (2004) SOCS3 is a critical physiological negative regulator of G-CSF signaling and emergency granulopoiesis. Immunity 20: 153–165.

18. CrokerBA, KrebsDL, ZhangJG, WormaldS, WillsonTA, et al. (2003) SOCS3 negatively regulates IL-6 signaling in vivo. Nat Immunol 4: 540–545.

19. KarioE, MarmorMD, AdamskyK, CitriA, AmitI, et al. (2005) Suppressors of cytokine signaling 4 and 5 regulate epidermal growth factor receptor signaling. J Biol Chem 280: 7038–7048.

20. BullockAN, RodriguezMC, DebreczeniJE, SongyangZ, KnappS (2007) Structure of the SOCS4-ElonginB/C complex reveals a distinct SOCS box interface and the molecular basis for SOCS-dependent EGFR degradation. Structure 15: 1493–1504.

21. HuG, ZhouR, LiuJ, GongAY, ChenXM (2010) MicroRNA-98 and let-7 regulate expression of suppressor of cytokine signaling 4 in biliary epithelial cells in response to Cryptosporidium parvum infection. J Infect Dis 202: 125–135.

22. SasiW, JiangWG, SharmaA, MokbelK (2010) Higher expression levels of SOCS 1,3,4,7 are associated with earlier tumour stage and better clinical outcome in human breast cancer. BMC Cancer 10: 178.

23. SutherlandJM, KeightleyRA, NixonB, RomanSD, RobkerRL, et al. (2012) Suppressor of cytokine signaling 4 (SOCS4): moderator of ovarian primordial follicle activation. J Cell Physiol 227: 1188–1198.

24. AugustinM, SedlmeierR, PetersT, HuffstadtU, KochmannE, et al. (2005) Efficient and fast targeted production of murine models based on ENU mutagenesis. Mamm Genome 16: 405–413.

25. BenderBS, CroghanT, ZhangL, SmallPAJr (1992) Transgenic mice lacking class I major histocompatibility complex-restricted T cells have delayed viral clearance and increased mortality after influenza virus challenge. J Exp Med 175: 1143–1145.

26. DentonAE, DohertyPC, TurnerSJ, La GrutaNL (2007) IL-18, but not IL-12, is required for optimal cytokine production by influenza virus-specific CD8+ T cells. Eur J Immunol 37: 368–375.

27. ImaiY, KubaK, NeelyGG, Yaghubian-MalhamiR, PerkmannT, et al. (2008) Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell 133: 235–249.

28. PeirisJS, HuiKP, YenHL (2010) Host response to influenza virus: protection versus immunopathology. Curr Opin Immunol 22: 475–481.

29. LawrenceCW, BracialeTJ (2004) Activation, differentiation, and migration of naive virus-specific CD8+ T cells during pulmonary influenza virus infection. J Immunol 173: 1209–1218.

30. LawrenceCW, ReamRM, BracialeTJ (2005) Frequency, specificity, and sites of expansion of CD8+ T cells during primary pulmonary influenza virus infection. J Immunol 174: 5332–5340.

31. SekiY, HayashiK, MatsumotoA, SekiN, TsukadaJ, et al. (2002) Expression of the suppressor of cytokine signaling-5 (SOCS5) negatively regulates IL-4-dependent STAT6 activation and Th2 differentiation. Proc Natl Acad Sci U S A 99: 13003–13008.

32. PothlichetJ, ChignardM, Si-TaharM (2008) Cutting edge: innate immune response triggered by influenza A virus is negatively regulated by SOCS1 and SOCS3 through a RIG-I/IFNAR1-dependent pathway. J Immunol 180: 2034–2038.

33. WeiH, WangS, ChenQ, ChenY, ChiX, et al. (2014) Suppression of Interferon Lambda Signaling by SOCS-1 Results in Their Excessive Production during Influenza Virus Infection. PLoS Pathog 10: e1003845.

34. Ramirez-MartinezG, Cruz-LagunasA, Jimenez-AlvarezL, EspinosaE, Ortiz-QuinteroB, et al. (2013) Seasonal and pandemic influenza H1N1 viruses induce differential expression of SOCS-1 and RIG-I genes and cytokine/chemokine production in macrophages. Cytokine 62: 151–159.

35. PauliEK, SchmolkeM, WolffT, ViemannD, RothJ, et al. (2008) Influenza A virus inhibits type I IFN signaling via NF-kappaB-dependent induction of SOCS-3 expression. PLoS Pathog 4: e1000196.

36. HuangY, ZaasAK, RaoA, DobigeonN, WoolfPJ, et al. (2011) Temporal dynamics of host molecular responses differentiate symptomatic and asymptomatic influenza A infection. PLoS Genet 7: e1002234.

37. ChanMC, CheungCY, ChuiWH, TsaoSW, NichollsJM, et al. (2005) Proinflammatory cytokine responses induced by influenza A (H5N1) viruses in primary human alveolar and bronchial epithelial cells. Respir Res 6: 135.

38. JulkunenI, SarenevaT, PirhonenJ, RonniT, MelenK, et al. (2001) Molecular pathogenesis of influenza A virus infection and virus-induced regulation of cytokine gene expression. Cytokine Growth Factor Rev 12: 171–180.

39. PaquetteSG, BannerD, ZhaoZ, FangY, HuangSS, et al. (2012) Interleukin-6 is a potential biomarker for severe pandemic H1N1 influenza A infection. PLoS One 7: e38214.

40. JanewayCJr, MedzhitovR (2000) Viral interference with IL-1 and toll signaling. Proc Natl Acad Sci U S A 97: 10682–10683.

41. SchmitzN, KurrerM, BachmannMF, KopfM (2005) Interleukin-1 is responsible for acute lung immunopathology but increases survival of respiratory influenza virus infection. J Virol 79: 6441–6448.

42. LeJM, FredricksonG, ReisLF, DiamantsteinT, HiranoT, et al. (1988) Interleukin 2-dependent and interleukin 2-independent pathways of regulation of thymocyte function by interleukin 6. Proc Natl Acad Sci U S A 85: 8643–8647.

43. CheungCY, PoonLL, LauAS, LukW, LauYL, et al. (2002) Induction of proinflammatory cytokines in human macrophages by influenza A (H5N1) viruses: a mechanism for the unusual severity of human disease? Lancet 360: 1831–1837.

44. MikolsCL, YanL, NorrisJY, RussellTD, KhalifahAP, et al. (2006) IL-12 p80 is an innate epithelial cell effector that mediates chronic allograft dysfunction. Am J Respir Crit Care Med 174: 461–470.

45. CooperAM, KhaderSA (2007) IL-12p40: an inherently agonistic cytokine. Trends Immunol 28: 33–38.

46. EverittAR, ClareS, PertelT, JohnSP, WashRS, et al. (2012) IFITM3 restricts the morbidity and mortality associated with influenza. Nature 484: 519–523.

47. ZhangYH, ZhaoY, LiN, PengYC, GiannoulatouE, et al. (2013) Interferon-induced transmembrane protein-3 genetic variant rs12252-C is associated with severe influenza in Chinese individuals. Nat Commun 4: 1418.

48. Murali-KrishnaK, AltmanJD, SureshM, SourdiveDJ, ZajacAJ, et al. (1998) Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity 8: 177–187.

49. ToughDF, BorrowP, SprentJ (1996) Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science 272: 1947–1950.

50. Di GenovaG, SavelyevaN, SuchackiA, ThirdboroughSM, StevensonFK (2010) Bystander stimulation of activated CD4+ T cells of unrelated specificity following a booster vaccination with tetanus toxoid. Eur J Immunol 40: 976–985.

51. TajimaM, WakitaD, NoguchiD, ChamotoK, YueZ, et al. (2008) IL-6-dependent spontaneous proliferation is required for the induction of colitogenic IL-17-producing CD8+ T cells. J Exp Med 205: 1019–1027.

52. GeginatJ, SallustoF, LanzavecchiaA (2001) Cytokine-driven proliferation and differentiation of human naive, central memory, and effector memory CD4(+) T cells. J Exp Med 194: 1711–1719.

53. UnutmazD, PileriP, AbrignaniS (1994) Antigen-independent activation of naive and memory resting T cells by a cytokine combination. J Exp Med 180: 1159–1164.

54. CoseS, BrammerC, KhannaKM, MasopustD, LefrancoisL (2006) Evidence that a significant number of naive T cells enter non-lymphoid organs as part of a normal migratory pathway. Eur J Immunol 36: 1423–1433.

55. MarzioR, MauelJ, Betz-CorradinS (1999) CD69 and regulation of the immune function. Immunopharmacol Immunotoxicol 21: 565–582.

56. TestiR, D'AmbrosioD, De MariaR, SantoniA (1994) The CD69 receptor: a multipurpose cell-surface trigger for hematopoietic cells. Immunol Today 15: 479–483.

57. Smith-GarvinJE, BurnsJC, GohilM, ZouT, KimJS, et al. (2010) T-cell receptor signals direct the composition and function of the memory CD8+ T-cell pool. Blood 116: 5548–5559.

58. HanJ, ShuiJW, ZhangX, ZhengB, HanS, et al. (2005) HIP-55 is important for T-cell proliferation, cytokine production, and immune responses. Mol Cell Biol 25: 6869–6878.

59. D'SouzaWN, ChangCF, FischerAM, LiM, HedrickSM (2008) The Erk2 MAPK regulates CD8 T cell proliferation and survival. J Immunol 181: 7617–7629.

60. RainerTH (2002) L-selectin in health and disease. Resuscitation 52: 127–141.

61. JenkinsMR, KedzierskaK, DohertyPC, TurnerSJ (2007) Heterogeneity of effector phenotype for acute phase and memory influenza A virus-specific CTL. J Immunol 179: 64–70.

62. BettsMR, BrenchleyJM, PriceDA, De RosaSC, DouekDC, et al. (2003) Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. J Immunol Methods 281: 65–78.

63. LeeC, KolesnikTB, CaminschiI, ChakravortyA, CarterW, et al. (2009) Suppressor of cytokine signalling 1 (SOCS1) is a physiological regulator of the asthma response. Clin Exp Allergy 39: 897–907.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 5
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#