#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Schmallenberg Virus Pathogenesis, Tropism and Interaction with the Innate Immune System of the Host


Schmallenberg virus (SBV) is an emerging orthobunyavirus of ruminants associated with outbreaks of congenital malformations in aborted and stillborn animals. Since its discovery in November 2011, SBV has spread very rapidly to many European countries. Here, we developed molecular and serological tools, and an experimental in vivo model as a platform to study SBV pathogenesis, tropism and virus-host cell interactions. Using a synthetic biology approach, we developed a reverse genetics system for the rapid rescue and genetic manipulation of SBV. We showed that SBV has a wide tropism in cell culture and “synthetic” SBV replicates in vitro as efficiently as wild type virus. We developed an experimental mouse model to study SBV infection and showed that this virus replicates abundantly in neurons where it causes cerebral malacia and vacuolation of the cerebral cortex. These virus-induced acute lesions are useful in understanding the progression from vacuolation to porencephaly and extensive tissue destruction, often observed in aborted lambs and calves in naturally occurring Schmallenberg cases. Indeed, we detected high levels of SBV antigens in the neurons of the gray matter of brain and spinal cord of naturally affected lambs and calves, suggesting that muscular hypoplasia observed in SBV-infected lambs is mostly secondary to central nervous system damage. Finally, we investigated the molecular determinants of SBV virulence. Interestingly, we found a biological SBV clone that after passage in cell culture displays increased virulence in mice. We also found that a SBV deletion mutant of the non-structural NSs protein (SBVΔNSs) is less virulent in mice than wild type SBV. Attenuation of SBV virulence depends on the inability of SBVΔNSs to block IFN synthesis in virus infected cells. In conclusion, this work provides a useful experimental framework to study the biology and pathogenesis of SBV.


Vyšlo v časopise: Schmallenberg Virus Pathogenesis, Tropism and Interaction with the Innate Immune System of the Host. PLoS Pathog 9(1): e32767. doi:10.1371/journal.ppat.1003133
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003133

Souhrn

Schmallenberg virus (SBV) is an emerging orthobunyavirus of ruminants associated with outbreaks of congenital malformations in aborted and stillborn animals. Since its discovery in November 2011, SBV has spread very rapidly to many European countries. Here, we developed molecular and serological tools, and an experimental in vivo model as a platform to study SBV pathogenesis, tropism and virus-host cell interactions. Using a synthetic biology approach, we developed a reverse genetics system for the rapid rescue and genetic manipulation of SBV. We showed that SBV has a wide tropism in cell culture and “synthetic” SBV replicates in vitro as efficiently as wild type virus. We developed an experimental mouse model to study SBV infection and showed that this virus replicates abundantly in neurons where it causes cerebral malacia and vacuolation of the cerebral cortex. These virus-induced acute lesions are useful in understanding the progression from vacuolation to porencephaly and extensive tissue destruction, often observed in aborted lambs and calves in naturally occurring Schmallenberg cases. Indeed, we detected high levels of SBV antigens in the neurons of the gray matter of brain and spinal cord of naturally affected lambs and calves, suggesting that muscular hypoplasia observed in SBV-infected lambs is mostly secondary to central nervous system damage. Finally, we investigated the molecular determinants of SBV virulence. Interestingly, we found a biological SBV clone that after passage in cell culture displays increased virulence in mice. We also found that a SBV deletion mutant of the non-structural NSs protein (SBVΔNSs) is less virulent in mice than wild type SBV. Attenuation of SBV virulence depends on the inability of SBVΔNSs to block IFN synthesis in virus infected cells. In conclusion, this work provides a useful experimental framework to study the biology and pathogenesis of SBV.


Zdroje

1. JonesKE, PatelNG, LevyMA, StoreygardA, BalkD, et al. (2008) Global trends in emerging infectious diseases. Nature 451: 990–993.

2. KupferschmidtK (2012) Infectious disease. Scientists rush to find clues on new animal virus. Science 335: 1028–1029.

3. HoffmannB, ScheuchM, HoperD, JungblutR, HolstegM, et al. (2012) Novel orthobunyavirus in Cattle, Europe, 2011. Emerg Infect Dis 18: 469–472.

4. GariglianyMM, BayrouC, KleijnenD, CassartD, JollyS, et al. (2012) Schmallenberg virus: a new Shamonda/Sathuperi-like virus on the rise in Europe. Antiviral Res 95: 82–87.

5. BeerM, ConrathsFJ, WHVDP (2012) ‘Schmallenberg virus’ - a novel orthobunyavirus emerging in Europe. Epidemiol Infect 1–8.

6. OIE (2012) Schmallenberg virus. OIE technical factsheet on SBV. May 2012.

7. GariglianyMM, BayrouC, KleijnenD, CassartD, DesmechtD (2012) Schmallenberg virus in domestic cattle, Belgium, 2012. Emerg Infect Dis 18: 1512–1514.

8. ElbersAR, LoeffenWL, QuakS, de Boer-LuijtzeE, van der SpekAN, et al. (2012) Seroprevalence of Schmallenberg virus antibodies among dairy cattle, the Netherlands, winter 2011–2012. Emerg Infect Dis 18: 1065–1071.

9. DominguezM, HendrikxP, ZientaraS, CalavasD, JayM, et al. (2012) Preliminary estimate of Schmallenberg virus infection impact in sheep flocks - France. Vet Rec 171: 426.

10. WalterCT, BarrJN (2011) Recent advances in the molecular and cellular biology of bunyaviruses. J Gen Virol 92: 2467–2484.

11. GollerKV, HoperD, SchirrmeierH, MettenleiterTC, BeerM (2012) Schmallenberg virus as possible ancestor of Shamonda virus. Emerg Infect Dis 18: 1644–1646.

12. CybinskiDH, St GeorgeTD, PaullNI (1978) Antibodies to Akabane virus in Australia. Aust Vet J 54: 1–3.

13. Della-PortaAJ, MurrayMD, CybinskiDH (1976) Congenital bovine epizootic arthrogryposis and hydranencephaly in Australia. Distribution of antibodies to Akabane virus in Australian Cattle after the 1974 epizootic. Aust Vet J 52: 496–501.

14. LiaoYK, LuYS, GotoY, InabaY (1996) The isolation of Akabane virus (Iriki strain) from calves in Taiwan. J Basic Microbiol 36: 33–39.

15. ShimshonyS (1980) An epizootic of Akabane disease in bovines, ovines and caprines in Israel, 1969–70: epidemiological assessment. Acta Morphol Acad Sci Hung 28: 197–199.

16. CauseyOR, KempGE, CauseyCE, LeeVH (1972) Isolations of Simbu-group viruses in Ibadan, Nigeria 1964–69, including the new types Sango, Shamonda, Sabo and Shuni. Ann Trop Med Parasitol 66: 357–362.

17. YanaseT, FukutomiT, YoshidaK, KatoT, OhashiS, et al. (2004) The emergence in Japan of Sathuperi virus, a tropical Simbu serogroup virus of the genus Orthobunyavirus. Arch Virol 149: 1007–1013.

18. RasmussenLD, KristensenB, KirkebyC, RasmussenTB, BelshamGJ, et al. (2012) Culicoids as vectors of Schmallenberg virus. Emerg Infect Dis 18: 1204–1206.

19. BridgenA, WeberF, FazakerleyJK, ElliottRM (2001) Bunyamwera bunyavirus nonstructural protein NSs is a nonessential gene product that contributes to viral pathogenesis. Proc Natl Acad Sci U S A 98: 664–669.

20. ThomasD, BlakqoriG, WagnerV, BanholzerM, KesslerN, et al. (2004) Inhibition of RNA polymerase II phosphorylation by a viral interferon antagonist. J Biol Chem 279: 31471–31477.

21. KohlA, ClaytonRF, WeberF, BridgenA, RandallRE, et al. (2003) Bunyamwera virus nonstructural protein NSs counteracts interferon regulatory factor 3-mediated induction of early cell death. J Virol 77: 7999–8008.

22. NeumannG, KawaokaY (2004) Reverse genetics systems for the generation of segmented negative-sense RNA viruses entirely from cloned cDNA. Curr Top Microbiol Immunol 283: 43–60.

23. ShiX, KohlA, LeonardVH, LiP, McLeesA, et al. (2006) Requirement of the N-terminal region of orthobunyavirus nonstructural protein NSm for virus assembly and morphogenesis. J Virol 80: 8089–8099.

24. TerasakiK, MurakamiS, LokugamageKG, MakinoS (2011) Mechanism of tripartite RNA genome packaging in Rift Valley fever virus. Proc Natl Acad Sci U S A 108: 804–809.

25. LowenAC, NoonanC, McLeesA, ElliottRM (2004) Efficient bunyavirus rescue from cloned cDNA. Virology 330: 493–500.

26. OgawaY, SugiuraK, KatoK, TohyaY, AkashiH (2007) Rescue of Akabane virus (family Bunyaviridae) entirely from cloned cDNAs by using RNA polymerase I. J Gen Virol 88: 3385–3390.

27. IkegamiT, WonS, PetersCJ, MakinoS (2006) Rescue of infectious rift valley fever virus entirely from cDNA, analysis of virus lacking the NSs gene, and expression of a foreign gene. J Virol 80: 2933–2940.

28. BlakqoriG, WeberF (2005) Efficient cDNA-based rescue of La Crosse bunyaviruses expressing or lacking the nonstructural protein NSs. J Virol 79: 10420–10428.

29. BridgenA, ElliottRM (1996) Rescue of a segmented negative-strand RNA virus entirely from cloned complementary DNAs. Proc Natl Acad Sci U S A 93: 15400–15404.

30. BuchholzUJ, FinkeS, ConzelmannKK (1999) Generation of bovine respiratory syncytial virus (BRSV) from cDNA: BRSV NS2 is not essential for virus replication in tissue culture, and the human RSV leader region acts as a functional BRSV genome promoter. J Virol 73: 251–259.

31. SouthamCM, MooreAE (1951) West Nile, Ilheus, and Bunyamwera virus infections in man. Am J Trop Med Hyg 31: 724–741.

32. HerderV, WohlseinP, PetersM, HansmannF, BaumgartnerW (2012) Salient lesions in domestic ruminants infected with the emerging so-called Schmallenberg virus in Germany. Vet Pathol 49: 588–591.

33. VialatP, BillecocqA, KohlA, BouloyM (2000) The S segment of rift valley fever phlebovirus (Bunyaviridae) carries determinants for attenuation and virulence in mice. J Virol 74: 1538–1543.

34. MullerR, SaluzzoJF, LopezN, DreierT, TurellM, et al. (1995) Characterization of clone 13, a naturally attenuated avirulent isolate of Rift Valley fever virus, which is altered in the small segment. Am J Trop Med Hyg 53: 405–411.

35. BlakqoriG, DelhayeS, HabjanM, BlairCD, Sanchez-VargasI, et al. (2007) La Crosse bunyavirus nonstructural protein NSs serves to suppress the type I interferon system of mammalian hosts. J Virol 81: 4991–4999.

36. BouloyM, JanzenC, VialatP, KhunH, PavlovicJ, et al. (2001) Genetic evidence for an interferon-antagonistic function of rift valley fever virus nonstructural protein NSs. J Virol 75: 1371–1377.

37. KillipMJ, YoungDF, RossCS, ChenS, GoodbournS, et al. (2011) Failure to activate the IFN-beta promoter by a paramyxovirus lacking an interferon antagonist. Virology 415: 39–46.

38. HotezPJ, SavioliL, FenwickA (2012) Neglected tropical diseases of the Middle East and North Africa: review of their prevalence, distribution, and opportunities for control. PLoS Negl Trop Dis 6: e1475.

39. LeisnhamPT, JulianoSA (2012) Impacts of climate, land use, and biological invasion on the ecology of immature Aedes mosquitoes: implications for La Crosse emergence. Ecohealth 9: 217–228.

40. AzevedoRS, NunesMR, ChiangJO, BensabathG, VasconcelosHB, et al. (2007) Reemergence of Oropouche fever, northern Brazil. Emerg Infect Dis 13: 912–915.

41. XuB, LiuL, HuangX, MaH, ZhangY, et al. (2011) Metagenomic analysis of fever, thrombocytopenia and leukopenia syndrome (FTLS) in Henan Province, China: discovery of a new bunyavirus. PLoS Pathog 7: e1002369.

42. RandolphSE, RogersDJ (2010) The arrival, establishment and spread of exotic diseases: patterns and predictions. Nat Rev Microbiol 8: 361–371.

43. PeperkampK, DijkmanR, van MaanenC, VosJ, WoudaW, et al. (2012) Polioencephalo-myelitis in a calf due to infection with Schmallenberg virus. Vet Rec 170: 570.

44. European Food Safety Authority (2012) Schmallenberg virus: likely epidemiologival scenarios and data needs. Technical report. Supporting publications 2012:EN-360. Parma, Italy.

45. EvansCA, ReynoldsJM, ReynoldsML, SaundersNR, SegalMB (1974) The development of a blood-brain barrier mechanism in foetal sheep. J Physiol 238: 371–386.

46. SaundersNR, LiddelowSA, DziegielewskaKM (2012) Barrier mechanisms in the developing brain. Front Pharmacol 3: 46.

47. WernikeK, BreithauptA, KellerM, HoffmannB, BeerM, et al. (2012) Schmallenberg virus infection of adult type I interferon receptor knock-out mice. PLoS One 7: e40380.

48. ArnaudF, BlackSG, MurphyL, GriffithsDJ, NeilSJ, et al. (2010) Interplay between ovine bone marrow stromal cell antigen 2/tetherin and endogenous retroviruses. J Virol 84: 4415–4425.

49. FarmerJL, BurghardtRC, JousanFD, HansenPJ, BazerFW, et al. (2008) Galectin 15 (LGALS15) functions in trophectoderm migration and attachment. Faseb J 22: 548–560.

50. GillespieJI, GreenwellJR, JohnsonC (1991) Agonist-induced fluctuations in cytoplasmic calcium in primary cultures of bovine endothelial cells. Exp Physiol 76: 667–676.

51. RatinierM, CaporaleM, GolderM, FranzoniG, AllanK, et al. (2011) Identification and characterization of a novel non-structural protein of bluetongue virus. PLoS Pathog 7: e1002477.

52. LeonardVH, KohlA, HartTJ, ElliottRM (2006) Interaction of Bunyamwera Orthobunyavirus NSs protein with mediator protein MED8: a mechanism for inhibiting the interferon response. J Virol 80: 9667–9675.

53. SpiegelM, PichlmairA, Martinez-SobridoL, CrosJ, Garcia-SastreA, et al. (2005) Inhibition of Beta interferon induction by severe acute respiratory syndrome coronavirus suggests a two-step model for activation of interferon regulatory factor 3. J Virol 79: 2079–2086.

54. BarryG, BreakwellL, FragkoudisR, Attarzadeh-YazdiG, Rodriguez-AndresJ, et al. (2009) PKR acts early in infection to suppress Semliki Forest virus production and strongly enhances the type I interferon response. J Gen Virol 90: 1382–1391.

55. PalmariniM, SharpJM, de las HerasM, FanH (1999) Jaagsiekte sheep retrovirus is necessary and sufficient to induce a contagious lung cancer in sheep. J Virol 73: 6964–6972.

56. VarelaM, ChowYH, SturkieC, MurciaP, PalmariniM (2006) Association of RON tyrosine kinase with the Jaagsiekte sheep retrovirus envelope glycoprotein. Virology 350: 347–357.

57. CaporaleM, WashR, PiniA, SaviniG, FranchiP, et al. (2011) Determinants of bluetongue virus virulence in murine models of disease. J Virol 85: 11479–11489.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 1
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#