#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Transmission-Blocking Antibodies against Mosquito C-Type Lectins for Dengue Prevention


C-type lectins are a family of proteins with carbohydrate-binding activity. Several C-type lectins in mammals or arthropods are employed as receptors or attachment factors to facilitate flavivirus invasion. We previously identified a C-type lectin in Aedes aegypti, designated as mosquito galactose specific C-type lectin-1 (mosGCTL-1), facilitating the attachment of West Nile virus (WNV) on the cell membrane. Here, we first identified that 9 A. aegypti mosGCTL genes were key susceptibility factors facilitating DENV-2 infection, of which mosGCTL-3 exhibited the most significant effect. We found that mosGCTL-3 was induced in mosquito tissues with DENV-2 infection, and that the protein interacted with DENV-2 surface envelop (E) protein and virions in vitro and in vivo. In addition, the other identified mosGCTLs interacted with the DENV-2 E protein, indicating that DENV may employ multiple mosGCTLs as ligands to promote the infection of vectors. The vectorial susceptibility factors that facilitate pathogen invasion may potentially be explored as a target to disrupt the acquisition of microbes from the vertebrate host. Indeed, membrane blood feeding of antisera against mosGCTLs dramatically reduced mosquito infective ratio. Hence, the immunization against mosGCTLs is a feasible approach for preventing dengue infection. Our study provides a future avenue for developing a transmission-blocking vaccine that interrupts the life cycle of dengue virus and reduces disease burden.


Vyšlo v časopise: Transmission-Blocking Antibodies against Mosquito C-Type Lectins for Dengue Prevention. PLoS Pathog 10(2): e32767. doi:10.1371/journal.ppat.1003931
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003931

Souhrn

C-type lectins are a family of proteins with carbohydrate-binding activity. Several C-type lectins in mammals or arthropods are employed as receptors or attachment factors to facilitate flavivirus invasion. We previously identified a C-type lectin in Aedes aegypti, designated as mosquito galactose specific C-type lectin-1 (mosGCTL-1), facilitating the attachment of West Nile virus (WNV) on the cell membrane. Here, we first identified that 9 A. aegypti mosGCTL genes were key susceptibility factors facilitating DENV-2 infection, of which mosGCTL-3 exhibited the most significant effect. We found that mosGCTL-3 was induced in mosquito tissues with DENV-2 infection, and that the protein interacted with DENV-2 surface envelop (E) protein and virions in vitro and in vivo. In addition, the other identified mosGCTLs interacted with the DENV-2 E protein, indicating that DENV may employ multiple mosGCTLs as ligands to promote the infection of vectors. The vectorial susceptibility factors that facilitate pathogen invasion may potentially be explored as a target to disrupt the acquisition of microbes from the vertebrate host. Indeed, membrane blood feeding of antisera against mosGCTLs dramatically reduced mosquito infective ratio. Hence, the immunization against mosGCTLs is a feasible approach for preventing dengue infection. Our study provides a future avenue for developing a transmission-blocking vaccine that interrupts the life cycle of dengue virus and reduces disease burden.


Zdroje

1. Rigau-PerezJG, ClarkGG, GublerDJ, ReiterP, SandersEJ, et al. (1998) Dengue and dengue haemorrhagic fever. The Lancet 352: 971–977.

2. WuJY, LunZR, JamesAA, ChenXG (2010) Dengue Fever in mainland China. Am J Trop Med Hyg 83: 664–671.

3. World Health Organization (2009) Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control.

4. RanjitS, KissoonN (2011) Dengue hemorrhagic fever and shock syndromes. Pediatr Crit Care Med 12: 90–100.

5. GouldEA, SolomonT (2008) Pathogenic flaviviruses. The Lancet 371: 500–509.

6. GublerDJ (1998) Dengue and dengue hemorrhagic fever. Clin Microbiol Rev 11: 480–496.

7. NeneV, WortmanJR, LawsonD, HaasB, KodiraC, et al. (2007) Genome sequence of Aedes aegypti, a major arbovirus vector. Science 316: 1718–1723.

8. HalsteadSB (2008) Dengue virus-mosquito interactions. Annu Rev Entomol 53: 273–291.

9. ChengG, CoxJ, WangP, KrishnanMN, DaiJ, et al. (2010) A C-type lectin collaborates with a CD45 phosphatase homolog to facilitate West Nile virus infection of mosquitoes. Cell 142: 714–725.

10. SessionsOM, BarrowsNJ, Souza-NetoJA, RobinsonTJ, HersheyCL, et al. (2009) Discovery of insect and human dengue virus host factors. Nature 458: 1047–1050.

11. ChengG, LiuL, WangP, ZhangY, ZhaoYO, et al. (2011) An in vivo transfection approach elucidates a role for Aedes aegypti thioester-containing proteins in flaviviral infection. PloS ONE 6: e22786.

12. ColpittsTM, CoxJ, VanlandinghamDL, FeitosaFM, ChengG, et al. (2011) Alterations in the Aedes aegypti transcriptome during infection with West Nile, dengue and yellow fever viruses. PLoS Pathogens 7: e1002189.

13. PepinKM, HanleyKA (2008) Density-dependent competitive suppression of sylvatic dengue virus by endemic dengue virus in cultured mosquito cells. Vector Borne Zoonotic Dis 8: 821–828.

14. RossTM (2010) Dengue virus. Clin Lab Med 30: 149–160.

15. RobinsonMJ, SanchoD, SlackEC, LeibundGut-LandmannS, Reis e SousaC (2006) Myeloid C-type lectins in innate immunity. Nature Immunology 7: 1258–1265.

16. FuchsA, LinTY, BeasleyDW, StoverCM, SchwaebleWJ, et al. (2010) Direct complement restriction of flavivirus infection requires glycan recognition by mannose-binding lectin. Cell Host & Microbe 8: 186–195.

17. AvirutnanP, HauhartRE, MarovichMA, GarredP, AtkinsonJP, et al. (2011) Complement-mediated neutralization of dengue virus requires mannose-binding lectin. mBio 2: e00276–11.

18. GeijtenbeekTB, KrooshoopDJ, BleijsDA, van VlietSJ, van DuijnhovenGC, et al. (2000) DC-SIGN-ICAM-2 interaction mediates dentritic cell trafficking. Nature Immunology 1: 353–357.

19. Navarro-SanchezE, AltmeyerR, AmaraA, SchwartzO, FieschiF, et al. (2003) Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Reports 4: 723–728.

20. TassaneetrithepB, BurgessTH, Granelli-PipernoA, TrumpfhellerC, FinkeJ, et al. (2003) DC-SIGN (CD209) mediates dengue virus infection of human dendritic cells. J Exp Med 197: 823–829.

21. ChaturvediU, NagarR, ShrivastavaR (2006) Dengue and dengue haemorrhagic fever: implications of host genetics. FEMS Immunol Med Mic 47: 155–166.

22. MillerJL, de WetBJ, Martinez-PomaresL, RadcliffeCM, DwekRA, et al. (2008) The mannose receptor mediates dengue virus infection of macrophages. PLoS Pathogens 4: e17.

23. ChenST, LinYL, HuangMT, WuMF, ChengSC, et al. (2008) CLEC5A is critical for dengue-virus-induced lethal disease. Nature 453: 672–676.

24. PalU, LiX, WangT, MontgomeryRR, RamamoorthiN, et al. (2004) TROSPA, an Ixodes scapularis receptor for Borrelia burgdorferi. Cell 119: 457–468.

25. JiangT, YuXD, HongWX, ZhouWZ, YuM, et al. (2009) Co-circulation of two genotypes of dengue virus serotype 3 in Guangzhou, China. Virol J 9: 125.

26. KlimstraWB, NangleEM, SmithMS, YurochkoAD, RymanKD (2003) DC-SIGN and L-SIGN can act as attachment receptors for alphaviruses and distinguish between mosquito cell- and mammalian cell-derived viruses. J Virol 77: 12022–12032.

27. ChenST, LiuRS, WuMF, LinYL, ChenSY, et al. (2012) CLEC5A regulates Japanese encephalitis virus-induced neuroinflammation and lethality. PLoS Pathogens 8: e1002655.

28. GunnBM, MorrisonTE, WhitmoreAC, BlevinsLK, HuestonL, et al. (2012) Mannose binding lectin is required for alphavirus-induced arthritis/myositis. PLoS Pathogens 8: e1002586.

29. HofmannH, LiX, ZhangX, LiuW, KühlA, et al. (2013) Severe fever with thrombocytopenia virus glycoproteins are targeted by neutralizing antibodies and can use DC-SIGN as a receptor for pH-dependent entry into human and animal cell lines. J Virol 87: 4384–4394.

30. LongKM, WhitmoreAC, FerrisMT, SempowskiGD, McGeeC, et al. (2013) Dendritic cell immune receptor (DCIR) regulates Chikungunya virus pathogenesis in mice. J Virol 87: 5697–5706.

31. PokidyshevaE, ZhangY, BattistiAJ, Bator-KellyCM, ChipmanPR, et al. (2006) Cryo-EM reconstruction of dengue virus in complex with the carbohydrate recognition domain of DC-SIGN. Cell 124: 485–493.

32. Navarro-SanchezE, AltmeyerR, AmaraA, SchwartzO, FieschiF, et al. (2003) Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Rep 4: 723–728.

33. ThomasML (1989) The leukocyte common antigen family. Annu Rev Immunol 7: 339–369.

34. TrowbridgeIS, ThomasML (1994) CD45: an emerging role as a protein tyrosine phosphatase required for lymphocyte activation and development. Annu Rev Immunol 12: 85–116.

35. BythKF, ConroyLA, HowlettS, SmithAJ, MayJ, et al. (1996) CD45-null transgenic mice reveal a positive regulatory role for CD45 in early thymocyte development, in the selection of CD4+ CD8+ thymocytes, and B cell maturation. J Exp Med 183: 1707–1718.

36. BaldwinTA, OstergaardHL (2001) Developmentally regulated changes in glucosidase II association with, and carbohydrate content of, the protein tyrosine phosphatase CD45. J Immunol 167: 3829–3835.

37. MiuraK, KeisterDB, MuratovaOV, SattabongkotJ, LongCA, et al. (2007) Transmission-blocking activity nduced by malaria vaccine candidates Pfs25/Pvs25 is a direct and predictable function of antibody titer. Malaria Journal 6: 107.

38. ChowdhuryDR, AngovE, KariukiT, KumarN (2009) A potent malaria transmission blocking vaccine based on codon harmonized full length Pfs48/45 expressed in Escherichia coli. PLoS ONE 4: e6352.

39. DinglasanRR, KalumeDE, KanzokSM, GhoshAK, MuratovaO, et al. (2007) Disruption of Plasmodium falciparum development by antibodies against a conservedmosquito midgut antigen. Proc Natl Acad Sci U S A 104: 13461–13466.

40. MathiasDK, PlieskattJL, ArmisteadJS, BethonyJM, Abdul-MajidKB, et al. (2012) Expression, immunogenicity, histopathology, and potency of a mosquito-based malariatransmission-blocking recombinant vaccine. Infect Immun 80: 1606–1614.

41. SabchareonA, WallaceD, SirivichayakulC, LimkittikulK, ChanthavanichP, et al. (2012) Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial. The Lancet 380: 1559–1567.

42. MackenzieJS, GublerDJ, PetersenLR (2004) Emerging flaviviruses: the spread and resurgence of Japanese encephalitis, West Nile and dengue viruses. Nature Medicine 10: S98–109.

43. RobinsonAS, FranzG, AtkinsonPW (2004) Insect transgenesis and its potential role in agriculture and human health. Insect Biochem Molec 34: 113–120.

44. PatesH, CurtisC (2005) Mosquito behavior and vector control. Annu Rev Entomol 50: 53–70.

45. WilkeAB, MarrelliMT (2012) Genetic Control of Mosquitoes: population suppression strategies. Rev Inst Med Trop Sao Paulo 54: 287–292.

46. ZompiS, HarrisE (2012) Animal models of dengue virus infection. Viruses 4: 62–82.

47. LiYP, LiangZL, XiaJL, WuJY, WangL, et al. (2013) Immunogenicity, Safety, and Immune Persistence of a Novel Inactivated Human Enterovirus 71 Vaccine: A Phase II, Randomized, Double-Blind, Placebo-Controlled Trial. J Infect Dis 209 (1) 46–55.

48. BuschMP, KleinmanSH, ToblerLH, KamelHT, NorrisPJ, et al. (2008) Virus and antibody dynamics in acute west nile virus infection. J Infect Dis 198: 984–993.

49. DengYQ, DaiJX, JiGH, JiangT, WangHJ, et al. (2011) A Broadly Flavivirus Cross-Neutralizing Monoclonal Antibody that Recognizes a Novel Epitope within the Fusion Loop of E Protein . PLoS One 6: e16059.

50. OlsthoornRC, BolJF (2001) Sequence comparison and secondary structure analysis of the 3′ noncoding region of flavivirus genomes reveals multiple pseudoknots. RNA 7: 1370–1377.

51. ZhaoH, DengYQ, HongWX, YuXD, JiangT, et al. (2012) Complete Genome Sequence of Dengue Virus Serotype 2 Cosmopolitan Genotype Strain in Guangdong, China. J Virol 86: 13808–13809.

52. ZhaoH, YuXD, ZhangXY, JiangT, HongWX, et al. (2012) Complete Genome Sequence of a Dengue Virus Serotype 4 Strain Isolated in Guangdong, China. J Virol 86: 7021–7022.

53. PizziM (1950) Sampling variation of the fifty per cent end-point, determined by the ReedMuench (Behrens) method. Human Biology 22: 151–190.

54. BaiF, TownT, PradhanD, CoxJ, Ashish, et al. (2007) Antiviral peptide stargeting the west nile virus envelope protein. J Virol 81: 2047–2055.

55. ColemanJ, JuhnJ, JamesAA (2007) Dissection of midgut and salivary glands from Ae. aegypti mosquitoes. J Vis Exp 5: 228.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 2
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#