#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Infection Induces Expression of a Mosquito Salivary Protein (Agaphelin) That Targets Neutrophil Function and Inhibits Thrombosis without Impairing Hemostasis


Malaria is transmitted by Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Salivary gland contributes to the development of the parasite by creating a favorable environment for the infection and facilitating blood feeding and reproduction of the vector. However, the molecular mechanism by which the vector salivary gland modulates parasite/host interactions is not understood. We discovered that infection of the mosquito salivary gland upregulates several genes; among them, one codes for a protease inhibitor named Agaphelin. Notably, Agaphelin was found to exhibit multiple antihemostatic functions by targeting elastase. As a result, it inhibits platelet function which is required for blood to clot, and it prevents cleavage of TFPI, an anticoagulant that has recently been found to play a crucial role in thrombus formation in vivo. Agaphelin also attenuates neutrophils chemotaxis and the release of Neutrophil Extracellular Traps. These results provide evidence that neutrophils serve as a link between coagulation and the innate immune response. Agaphelin also exhibits anti-inflammatory and antithrombotic effects in vivo. Furthermore, Agaphelin did not promote bleeding, suggesting that targeting neutrophil exhibits potential therapeutic value. Altogether, these results highlight that the interplay between parasite, vector and host is a dynamic process that contributes and sustains the interface among Plasmodium, Anopheles and humans.


Vyšlo v časopise: Infection Induces Expression of a Mosquito Salivary Protein (Agaphelin) That Targets Neutrophil Function and Inhibits Thrombosis without Impairing Hemostasis. PLoS Pathog 10(9): e32767. doi:10.1371/journal.ppat.1004338
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004338

Souhrn

Malaria is transmitted by Plasmodium falciparum-infected Anopheles gambiae mosquitoes. Salivary gland contributes to the development of the parasite by creating a favorable environment for the infection and facilitating blood feeding and reproduction of the vector. However, the molecular mechanism by which the vector salivary gland modulates parasite/host interactions is not understood. We discovered that infection of the mosquito salivary gland upregulates several genes; among them, one codes for a protease inhibitor named Agaphelin. Notably, Agaphelin was found to exhibit multiple antihemostatic functions by targeting elastase. As a result, it inhibits platelet function which is required for blood to clot, and it prevents cleavage of TFPI, an anticoagulant that has recently been found to play a crucial role in thrombus formation in vivo. Agaphelin also attenuates neutrophils chemotaxis and the release of Neutrophil Extracellular Traps. These results provide evidence that neutrophils serve as a link between coagulation and the innate immune response. Agaphelin also exhibits anti-inflammatory and antithrombotic effects in vivo. Furthermore, Agaphelin did not promote bleeding, suggesting that targeting neutrophil exhibits potential therapeutic value. Altogether, these results highlight that the interplay between parasite, vector and host is a dynamic process that contributes and sustains the interface among Plasmodium, Anopheles and humans.


Zdroje

1. ChoumetV, AttoutT, ChartierL, KhunH, SautereauJ, et al. (2012) Visualizing non infectious and infectious Anopheles gambiae blood feedings in naive and saliva-immunized mice. PLoS One 7: e50464.

2. RibeiroJM, FrancischettiIM (2003) Role of arthropod saliva in blood feeding: sialome and post-sialome perspectives. Annu Rev Entomol 48: 73–88.

3. FrancischettiIM (2010) Platelet aggregation inhibitors from hematophagous animals. Toxicon 56: 1130–1144.

4. PhillipsonM, KubesP (2011) The neutrophil in vascular inflammation. Nat Med 17: 1381–1390.

5. SchulzC, EngelmannB, MassbergS (2013) Crossroads of coagulation and innate immunity: the case of deep vein thrombosis. J Thromb Haemost 11 Suppl 1: 233–241.

6. CarboneF, NencioniA, MachF, VuilleumierN, MontecuccoF (2013) Pathophysiological role of neutrophils in acute myocardial infarction. Thromb Haemost 110: 501–514.

7. MartinodK, WagnerDD (2014) Thrombosis: tangled up in NETs. Blood 123: 2768–2776.

8. YippBG, KubesP (2013) NETosis: how vital is it? Blood 122: 2784–2794.

9. AbeH, OkajimaK, OkabeH, TakatsukiK, BinderBR (1994) Granulocyte proteases and hydrogen peroxide synergistically inactivate thrombomodulin of endothelial cells in vitro. J Lab Clin Med 123: 874–881.

10. HiguchiDA, WunTC, LikertKM, BrozeGJJr (1992) The effect of leukocyte elastase on tissue factor pathway inhibitor. Blood 79: 1712–1719.

11. BrozeGJJr, GirardTJ (2012) Tissue factor pathway inhibitor: structure-function. Front Biosci 17: 262–280.

12. MassbergS, GrahlL, von BruehlML, ManukyanD, PfeilerS, et al. (2010) Reciprocal coupling of coagulation and innate immunity via neutrophil serine proteases. Nat Med 16: 887–896.

13. RufW, RuggeriZM (2010) Neutrophils release brakes of coagulation. Nat Med 16: 851–852.

14. AlamSR, NewbyDE, HenriksenPA (2012) Role of the endogenous elastase inhibitor, elafin, in cardiovascular injury: from epithelium to endothelium. Biochem Pharmacol 83: 695–704.

15. PhamCT (2006) Neutrophil serine proteases: specific regulators of inflammation. Nat Rev Immunol 6: 541–550.

16. HirahashiJ, MekalaD, Van ZiffleJ, XiaoL, SaffaripourS, et al. (2006) Mac-1 signaling via Src-family and Syk kinases results in elastase-dependent thrombohemorrhagic vasculopathy. Immunity 25: 271–283.

17. SchofieldZV, WoodruffTM, HalaiR, WuMC, CooperMA (2013) Neutrophils-a key component of ischemia-reperfusion injury. Shock 40: 463–470.

18. von BruhlML, StarkK, SteinhartA, ChandraratneS, KonradI, et al. (2012) Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med 209: 819–835.

19. FuchsTA, BrillA, DuerschmiedD, SchatzbergD, MonestierM, et al. (2010) Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci U S A 107: 15880–15885.

20. JinR, YangG, LiG (2010) Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol 87: 779–789.

21. SunZ, YangP (2004) Role of imbalance between neutrophil elastase and alpha 1-antitrypsin in cancer development and progression. Lancet Oncol 5: 182–190.

22. HenriksenPA (2014) The potential of neutrophil elastase inhibitors as anti-inflammatory therapies. Curr Opin Hematol 21: 23–28.

23. LovatoDV, Nicolau de CamposIT, AminoR, TanakaAS (2006) The full-length cDNA of anticoagulant protein infestin revealed a novel releasable Kazal domain, a neutrophil elastase inhibitor lacking anticoagulant activity. Biochimie 88: 673–681.

24. WikelS (2013) Ticks and tick-borne pathogens at the cutaneous interface: host defenses, tick countermeasures, and a suitable environment for pathogen establishment. Front Microbiol 4: 337.

25. NuttallPA, LabudaM (2004) Tick-host interactions: saliva-activated transmission. Parasitology 129 Suppl: S177–189.

26. ChampagneDE (2005) Antihemostatic molecules from saliva of blood-feeding arthropods. Pathophysiol Haemost Thromb 34: 221–227.

27. Sa-Nunes A, Oliveira CJ (2010) Sialogenins and Immunomodulators Derived from Blood Feeding Parasites. Toxins and Hemostasis From bench to bedside Editors R M Kini, K J Clemetson, FS Markland, MA McLane, T Morita Springer, NY (2010): : 131–152.

28. MillerLH, AckermanHC, SuXZ, WellemsTE (2013) Malaria biology and disease pathogenesis: insights for new treatments. Nat Med 19: 156–167.

29. AveryJW, SmithGM, OwinoSO, SarrD, NagyT, et al. (2012) Maternal malaria induces a procoagulant and antifibrinolytic state that is embryotoxic but responsive to anticoagulant therapy. PLoS One 7: e31090.

30. BakerDA, NolanT, FischerB, PinderA, CrisantiA, et al. (2011) A comprehensive gene expression atlas of sex- and tissue-specificity in the malaria vector, Anopheles gambiae. BMC Genomics 12: 296.

31. LuSM, LuW, QasimMA, AndersonS, ApostolI, et al. (2001) Predicting the reactivity of proteins from their sequence alone: Kazal family of protein inhibitors of serine proteinases. Proc Natl Acad Sci U S A 98: 1410–1415.

32. TakacP, NunnMA, MeszarosJ, PechanovaO, VrbjarN, et al. (2006) Vasotab, a vasoactive peptide from horse fly Hybomitra bimaculata (Diptera, Tabanidae) salivary glands. J Exp Biol 209: 343–352.

33. KrowarschD, CierpickiT, JelenF, OtlewskiJ (2003) Canonical protein inhibitors of serine proteases. Cell Mol Life Sci 60: 2427–2444.

34. RimphanitchayakitV, TassanakajonA (2010) Structure and function of invertebrate Kazal-type serine proteinase inhibitors. Dev Comp Immunol 34: 377–386.

35. CerRZ, MudunuriU, StephensR, LebedaFJ (2009) IC50-to-Ki: a web-based tool for converting IC50 to Ki values for inhibitors of enzyme activity and ligand binding. Nucleic Acids Res 37: W441–445.

36. KanegasakiS, NomuraY, NittaN, AkiyamaS, TamataniT, et al. (2003) A novel optical assay system for the quantitative measurement of chemotaxis. J Immunol Methods 282: 1–11.

37. Si-TaharM, PidardD, BalloyV, MoniatteM, KiefferN, et al. (1997) Human neutrophil elastase proteolytically activates the platelet integrin alphaIIbbeta3 through cleavage of the carboxyl terminus of the alphaIIb subunit heavy chain. Involvement in the potentiation of platelet aggregation. J Biol Chem 272: 11636–11647.

38. PapayannopoulosV, MetzlerKD, HakkimA, ZychlinskyA (2010) Neutrophil elastase and myeloperoxidase regulate the formation of neutrophil extracellular traps. J Cell Biol 191: 677–691.

39. MaD, MizuriniDM, AssumpcaoTC, LiY, QiY, et al. (2013) Desmolaris, a novel factor XIa anticoagulant from the salivary gland of the vampire bat (Desmodus rotundus) inhibits inflammation and thrombosis in vivo. Blood 122: 4094–4106.

40. OwensAP3rd, LuY, WhinnaHC, GachetC, FayWP, et al. (2011) Towards a standardization of the murine ferric chloride-induced carotid arterial thrombosis model. J Thromb Haemost 9: 1862–1863.

41. EcklyA, HechlerB, FreundM, ZerrM, CazenaveJP, et al. (2011) Mechanisms underlying FeCl3-induced arterial thrombosis. J Thromb Haemost 9: 779–789.

42. MikolajczakSA, Silva-RiveraH, PengX, TarunAS, CamargoN, et al. (2008) Distinct malaria parasite sporozoites reveal transcriptional changes that cause differential tissue infection competence in the mosquito vector and mammalian host. Mol Cell Biol 28: 6196–6207.

43. Rosinski-ChupinI, ChertempsT, BoissonB, PerrotS, BischoffE, et al. (2007) Serial Analysis of Gene Expression in Plasmodium berghei salivary gland sporozoites. BMC Genomics 8: 466.

44. KaiserK, MatuschewskiK, CamargoN, RossJ, KappeSH (2004) Differential transcriptome profiling identifies Plasmodium genes encoding pre-erythrocytic stage-specific proteins. Mol Microbiol 51: 1221–1232.

45. van HoefV, BreugelmansB, SpitJ, SimonetG, ZelsS, et al. (2013) Phylogenetic distribution of protease inhibitors of the Kazal-family within the Arthropoda. Peptides 41: 59–65.

46. LiXC, WangXW, WangZH, ZhaoXF, WangJX (2009) A three-domain Kazal-type serine proteinase inhibitor exhibiting domain inhibitory and bacteriostatic activities from freshwater crayfish Procambarus clarkii. Dev Comp Immunol 33: 1229–1238.

47. OwenCA, CampbellMA, SannesPL, BoukedesSS, CampbellEJ (1995) Cell surface-bound elastase and cathepsin G on human neutrophils: a novel, non-oxidative mechanism by which neutrophils focus and preserve catalytic activity of serine proteinases. J Cell Biol 131: 775–789.

48. YoungRE, ThompsonRD, LarbiKY, LaM, RobertsCE, et al. (2004) Neutrophil elastase (NE)-deficient mice demonstrate a nonredundant role for NE in neutrophil migration, generation of proinflammatory mediators, and phagocytosis in response to zymosan particles in vivo. J Immunol 172: 4493–4502.

49. AoshibaK, NagaiA, TakizawaT (1991) Effects of proteinase inhibitors on polymorphonuclear neutrophil polarization. Tohoku J Exp Med 165: 165–170.

50. WoodmanRC, ReinhardtPH, KanwarS, JohnstonFL, KubesP (1993) Effects of human neutrophil elastase (HNE) on neutrophil function in vitro and in inflamed microvessels. Blood 82: 2188–2195.

51. ReevesEP, BanvilleN, RyanDM, O'ReillyN, BerginDA, et al. (2013) Intracellular secretory leukoprotease inhibitor modulates inositol 1,4,5-triphosphate generation and exerts an anti-inflammatory effect on neutrophils of individuals with cystic fibrosis and chronic obstructive pulmonary disease. Biomed Res Int 2013: 560141.

52. AdkisonAM, RaptisSZ, KelleyDG, PhamCT (2002) Dipeptidyl peptidase I activates neutrophil-derived serine proteases and regulates the development of acute experimental arthritis. J Clin Invest 109: 363–371.

53. JenneCN, UrrutiaR, KubesP (2013) Platelets: bridging hemostasis, inflammation, and immunity. Int J Lab Hematol 35: 254–261.

54. PilsczekFH, SalinaD, PoonKK, FaheyC, YippBG, et al. (2010) A novel mechanism of rapid nuclear neutrophil extracellular trap formation in response to Staphylococcus aureus. J Immunol 185: 7413–7425.

55. BraianC, HogeaV, StendahlO (2013) Mycobacterium tuberculosis- induced neutrophil extracellular traps activate human macrophages. J Innate Immun 5: 591–602.

56. FarleyK, StolleyJM, ZhaoP, CooleyJ, Remold-O'DonnellE (2012) A serpinB1 regulatory mechanism is essential for restricting neutrophil extracellular trap generation. J Immunol 189: 4574–4581.

57. TaggartCC, CryanSA, WeldonS, GibbonsA, GreeneCM, et al. (2005) Secretory leucoprotease inhibitor binds to NF-kappaB binding sites in monocytes and inhibits p65 binding. J Exp Med 202: 1659–1668.

58. CollinN, AssumpcaoTC, MizuriniDM, GilmoreDC, Dutra-OliveiraA, et al. (2012) Lufaxin, a novel factor Xa inhibitor from the salivary gland of the sand fly Lutzomyia longipalpis blocks protease-activated receptor 2 activation and inhibits inflammation and thrombosis in vivo. Arterioscler Thromb Vasc Biol 32: 2185–2198.

59. MizuriniDM, FrancischettiIM, AndersenJF, MonteiroRQ (2010) Nitrophorin 2, a factor IX(a)-directed anticoagulant, inhibits arterial thrombosis without impairing haemostasis. Thromb Haemost 104: 1116–1123.

60. NazarethRA, TomazLS, Ortiz-CostaS, AtellaGC, RibeiroJM, et al. (2006) Antithrombotic properties of Ixolaris, a potent inhibitor of the extrinsic pathway of the coagulation cascade. Thromb Haemost 96: 7–13.

61. FurieB, FurieBC (2008) Mechanisms of thrombus formation. N Engl J Med 359: 938–949.

62. GleissnerCA, von HundelshausenP, LeyK (2008) Platelet chemokines in vascular disease. Arterioscler Thromb Vasc Biol 28: 1920–1927.

63. WatsonSP (2009) Platelet activation by extracellular matrix proteins in haemostasis and thrombosis. Curr Pharm Des 15: 1358–1372.

64. RenneT, SchmaierAH, NickelKF, BlombackM, MaasC (2012) In vivo roles of factor XII. Blood 120: 4296–4303.

65. FrancischettiIM, ValenzuelaJG, RibeiroJM (1999) Anophelin: kinetics and mechanism of thrombin inhibition. Biochemistry 38: 16678–16685.

66. AssumpcaoTC, MaD, SchwarzA, ReiterK, SantanaJM, et al. (2013) Salivary Antigen-5/CAP Family Members Are Cu2+-dependent Antioxidant Enzymes That Scavenge OFormula and Inhibit Collagen-induced Platelet Aggregation and Neutrophil Oxidative Burst. J Biol Chem 288: 14341–14361.

67. RibeiroJM, ValenzuelaJG (1999) Purification and cloning of the salivary peroxidase/catechol oxidase of the mosquito Anopheles albimanus. J Exp Biol 202: 809–816.

68. KappeSH, KaiserK, MatuschewskiK (2003) The Plasmodium sporozoite journey: a rite of passage. Trends Parasitol 19: 135–143.

69. JanoffA, RothWJ, SinhaS, BarnwellJW (1988) Degradation of plasmodial antigens by human neutrophil elastase. J Immunol 141: 1332–1340.

70. IfedibaT, VanderbergJP (1981) Complete in vitro maturation of Plasmodium falciparum gametocytes. Nature 294: 364–366.

71. PonnuduraiT, LensenAH, Van GemertGJ, BensinkMP, BolmerM, et al. (1989) Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes. Parasitology 98 Pt 2: 165–173.

72. KabiruEW, MbogoCM, MuiruriSK, OumaJH, GithureJI, et al. (1997) Sporozoite loads of naturally infected Anopheles in Kilifi District, Kenya. J Am Mosq Control Assoc 13: 259–262.

73. EdgarR, DomrachevM, LashAE (2002) Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic acids research 30: 207–210.

74. ThompsonJD, GibsonTJ, PlewniakF, JeanmouginF, HigginsDG (1997) The CLUSTAL_X windows interface: flexible strategies for multiple sequence alignment aided by quality analysis tools. Nucleic Acids Res 25: 4876–4882.

75. KumarS, TamuraK, NeiM (2004) MEGA3: Integrated software for Molecular Evolutionary Genetics Analysis and sequence alignment. Brief Bioinform 5: 150–163.

76. MegyK, EmrichSJ, LawsonD, CampbellD, DialynasE, et al. (2012) VectorBase: improvements to a bioinformatics resource for invertebrate vector genomics. Nucleic Acids Res 40: D729–734.

77. PetersenTN, BrunakS, von HeijneG, NielsenH (2011) SignalP 4.0: discriminating signal peptides from transmembrane regions. Nat Methods 8: 785–786.

78. BordoliL, SchwedeT (2012) Automated protein structure modeling with SWISS-MODEL Workspace and the Protein Model Portal. Methods Mol Biol 857: 107–136.

79. KelleyLA, SternbergMJ (2009) Protein structure prediction on the Web: a case study using the Phyre server. Nat Protoc 4: 363–371.

80. ZhangY (2008) I-TASSER server for protein 3D structure prediction. BMC Bioinformatics 9: 40.

81. RoyA, KucukuralA, ZhangY (2010) I-TASSER: a unified platform for automated protein structure and function prediction. Nat Protoc 5: 725–738.

82. IlinkinI, YeJ, JanardanR (2010) Multiple structure alignment and consensus identification for proteins. BMC Bioinformatics 11: 71.

83. KabschW, SanderC (1983) Dictionary of protein secondary structure: pattern recognition of hydrogen-bonded and geometrical features. Biopolymers 22: 2577–2637.

84. WhitmoreL, WallaceBA (2008) Protein secondary structure analyses from circular dichroism spectroscopy: methods and reference databases. Biopolymers 89: 392–400.

85. ChmelarJ, OliveiraCJ, RezacovaP, FrancischettiIM, KovarovaZ, et al. (2011) A tick salivary protein targets cathepsin G and chymase and inhibits host inflammation and platelet aggregation. Blood 117: 736–744.

86. MaD, AssumpcaoTC, LiY, AndersenJF, RibeiroJ, et al. (2012) Triplatin, a platelet aggregation inhibitor from the salivary gland of the triatomine vector of Chagas disease, binds to TXA(2) but does not interact with glycoprotein PVI. Thromb Haemost 107: 111–123.

87. FrancischettiIM, GhazalehFA, ReisRA, CarliniCR, GuimaraesJA (1998) Convulxin induces platelet activation by a tyrosine-kinase-dependent pathway and stimulates tyrosine phosphorylation of platelet proteins, including PLC gamma 2, independently of integrin alpha IIb beta 3. Arch Biochem Biophys 353: 239–250.

88. AssumpcaoTC, AlvarengaPH, RibeiroJM, AndersenJF, FrancischettiIM (2010) Dipetalodipin, a novel multifunctional salivary lipocalin that inhibits platelet aggregation, vasoconstriction, and angiogenesis through unique binding specificity for TXA2, PGF2alpha, and 15(S)-HETE. J Biol Chem 285: 39001–39012.

89. WeichselA, AndersenJF, ChampagneDE, WalkerFA, MontfortWR (1998) Crystal structures of a nitric oxide transport protein from a blood-sucking insect. Nat Struct Biol 5: 304–309.

90. GarletGP, CardosoCR, CampanelliAP, FerreiraBR, Avila-CamposMJ, et al. (2007) The dual role of p55 tumour necrosis factor-alpha receptor in Actinobacillus actinomycetemcomitans-induced experimental periodontitis: host protection and tissue destruction. Clin Exp Immunol 147: 128–138.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 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#