J. M. Ribeiro, Blood-feeding arthropods: live syringes or invertebrate pharmacologists? Infect Agents Dis, vol.4, pp.143-152, 1995.

W. C. Black, Evolution of arthropod disease vectors, Greenblatt C, Spigelman M, pp.49-63, 2003.

J. M. Ribeiro and I. M. Francischetti, Role of arthropod saliva in blood feeding: sialome and post-sialome perspectives, Annu Rev Entomol, vol.48, pp.73-88, 2003.

B. B. Andrade, C. R. Teixeira, A. Barral, and M. Barral-netto, Haematophagous arthropod saliva and host defense system: a tale of tear and blood, vol.77, pp.665-693, 2005.

I. M. Francischetti, A. Sa-nunes, B. J. Mans, I. M. Santos, and J. M. Ribeiro, The role of saliva in tick feeding, Front Biosci, vol.14, pp.2051-2088, 2009.

T. Reunala, H. Brummer-korvenkontio, and T. Palosuo, Are we really allergic to mosquito bites, Ann Med, vol.26, pp.301-306, 1994.

A. J. Bircher, Systemic immediate allergic reactions to arthropod stings and bites, Dermatology, vol.210, pp.119-127, 2005.

R. Bellini, R. Veronesi, S. Draghetti, and M. Carrieri, Study on the flying height of Aedes caspius and Culex pipiens females in the Po Delta area, Italy, J Am Mosq Control Assoc, vol.13, pp.356-360, 1997.

T. Balenghien, A. Carron, G. Sinegre, and D. J. Bicout, Mosquito density forecast from flooding: population dynamics model for Aedes caspius (Pallas), Bull Entomol Res, vol.100, pp.247-254, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02659425

P. A. Nuttall, G. C. Paesen, C. H. Lawrie, and H. Wang, Vector-host interactions in disease transmission, J Mol Microbiol Biotechnol, vol.2, pp.381-386, 2000.

I. Rohousova and P. Volf, WHO: Deaths by cause, sex and mortality stratum in WHO Regions, estimates for 2002. Geneva: World Health Organization, Folia Parasitol (Praha), vol.53, pp.161-171, 2004.

R. W. Snow, C. A. Guerra, A. M. Noor, H. Y. Myint, and S. I. Hay, The global distribution of clinical episodes of Plasmodium falciparum malaria, Nature, vol.434, pp.214-217, 2005.

P. J. Hotez, A. Fenwick, L. Savioli, and D. H. Molyneux, Rescuing the bottom billion through control of neglected tropical diseases, Lancet, vol.373, pp.1570-1575, 2009.

G. L. Campbell, C. S. Ceianu, and H. M. Savage, Epidemic West Nile encephalitis in Romania: waiting for history to repeat itself, Ann N Y Acad Sci, vol.951, pp.94-101, 2001.

S. Kurkela, T. Manni, J. Myllynen, A. Vaheri, and O. Vapalahti, Clinical and laboratory manifestations of Sindbis virus infection: prospective study, J Infect Dis, vol.191, pp.1820-1829, 2002.

C. Chastel,

, Bull Acad Natl Med, vol.193, pp.485-493, 2009.

A. D. Barrett and D. E. Teuwen, Yellow fever vaccine -how does it work and why do rare cases of serious adverse events take place?, Curr Opin Immunol, vol.21, pp.308-313, 2009.

A. Wilder-smith and S. Halstead, Japanese encephalitis: update on vaccines and vaccine recommendations, Curr Opin Infect Dis, 2010.

T. Ikegami and S. Makino, Rift valley fever vaccines, vol.27, pp.69-72, 2009.

P. Rendi-wagner, Advances in vaccination against tick-borne encephalitis, Expert Rev Vaccines, vol.7, pp.589-596, 2008.

B. Pradines, H. Vial, and P. Olliaro,

, Med Trop (Mars), vol.63, pp.79-98, 2003.

A. Talbert, A. Nyange, and F. Molteni, Spraying tick-infested houses with lambda-cyhalothrin reduces the incidence of tick-borne relapsing fever in children under five years old, Trans R Soc Trop Med Hyg, vol.92, pp.251-253, 1998.

B. Alexander and M. Maroli, Control of phlebotomine sandflies, Med Vet Entomol, vol.17, pp.1-18, 2003.

, WHO: Malaria Vector Control and Personal Protection, WHO Technical Report, p.936, 2006.

. Fontaine, Parasites & Vectors, vol.4, p.187, 2011.

A. Enayati, J. Lines, R. Maharaj, J. Hemingway, R. Feachem et al., Shrinking the Malaria Map: A Prospectus on Malaria Elimination, vol.9, pp.140-154, 2009.

C. G. Nevill, E. S. Some, . Vo, W. Mutemi, L. New et al., Insecticide-treated bednets reduce mortality and severe morbidity from malaria among children on the Kenyan coast, Trop Med Int Health, vol.1, pp.139-146, 1996.

J. Keiser, B. H. Singer, and J. Utzinger, Reducing the burden of malaria in different eco-epidemiological settings with environmental management: a systematic review, Lancet Infect Dis, vol.5, pp.695-708, 2005.

G. F. Killeen, T. A. Smith, H. M. Ferguson, H. Mshinda, S. Abdulla et al., Preventing childhood malaria in Africa by protecting adults from mosquitoes with insecticide-treated nets, PLoS Med, vol.4, p.229, 2007.

R. Patterson and . Dr, William Gorgas and his war with the mosquito, CMAJ, vol.141, pp.596-597, 1989.

G. F. Killeen, U. Fillinger, I. Kiche, L. C. Gouagna, and B. G. Knols, Eradication of Anopheles gambiae from Brazil: lessons for malaria control in Africa?, Lancet Infect Dis, vol.2, pp.618-627, 2002.

C. Pennetier, V. Corbel, P. Boko, A. Odjo, N. 'guessan et al., Synergy between repellents and non-pyrethroid insecticides strongly extends the efficacy of treated nets against Anopheles gambiae, Malar J, vol.6, p.38, 2007.

R. C. Malima, S. M. Magesa, P. K. Tungu, V. Mwingira, F. S. Magogo et al., An experimental hut evaluation of Olyset nets against anopheline mosquitoes after seven years use in Tanzanian villages, Malar J, vol.7, p.38, 2008.

S. Kasili, H. Kutima, C. Mwandawiro, P. M. Ngumbi, C. O. Anjili et al., Laboratory and semi-field evaluation of long-lasting insecticidal nets against leishmaniasis vector, Phlebotomus (Phlebotomus) duboscqi in Kenya, J Vector Borne Dis, vol.47, pp.1-10, 2010.

P. Tungu, S. Magesa, C. Maxwell, R. Malima, D. Masue et al., Evaluation of PermaNet 3.0 a deltamethrin-PBO combination net against Anopheles gambiae and pyrethroid resistant Culex quinquefasciatus mosquitoes: an experimental hut trial in Tanzania, Malar J, vol.9, p.21, 2010.

C. F. Curtis and A. E. Mnzava, Comparison of house spraying and insecticidetreated nets for malaria control, Bull World Health Organ, vol.78, pp.1389-1400, 2000.

C. Lengeler, Insecticide-treated bed nets and curtains for preventing malaria. Cochrane Database Syst Rev, p.363, 2004.

G. F. Killeen, F. E. Mckenzie, B. D. Foy, C. Schieffelin, P. F. Billingsley et al., The potential impact of integrated malaria transmission control on entomologic inoculation rate in highly endemic areas, Am J Trop Med Hyg, vol.62, pp.545-551, 2000.

U. Thavara, A. Tawatsin, J. Chompoosri, W. Suwonkerd, U. R. Chansang et al., Laboratory and field evaluations of the insect repellent 3535 (ethyl butylacetylaminopropionate) and deet against mosquito vectors in Thailand, J Am Mosq Control Assoc, vol.17, pp.190-195, 2001.

E. M. Mathenge, G. O. Misiani, D. O. Oulo, L. W. Irungu, P. N. Ndegwa et al., Comparative performance of the Mbita trap, CDC light trap and the human landing catch in the sampling of Anopheles arabiensis, An. funestus and culicine species in a rice irrigation in western Kenya, Malar J, vol.4, p.7, 2005.

C. M. Mbogo, J. M. Mwangangi, J. Nzovu, W. Gu, G. Yan et al., Spatial and temporal heterogeneity of Anopheles mosquitoes and Plasmodium falciparum transmission along the Kenyan coast, Am J Trop Med Hyg, vol.68, pp.734-742, 2003.

E. Orlandi-pradines, C. Rogier, B. Koffi, F. Jarjaval, M. Bell et al., Pages F: Major variations in malaria exposure of travellers in rural areas: an entomological cohort study in western Cote d'Ivoire, Malar J, vol.8, p.171, 2009.

L. W. Cohnstaedt, J. I. Gillen, and L. E. Munstermann, Light-emitting diode technology improves insect trapping, J Am Mosq Control Assoc, vol.24, pp.331-334, 2008.

J. F. Carroll and E. T. Schmidtmann, Tick sweep: modification of the tick drag-flag method for sampling nymphs of the deer tick (Acari: Ixodidae), J Med Entomol, vol.29, pp.352-355, 1992.

A. Barral, E. Honda, A. Caldas, J. Costa, V. Vinhas et al., Human immune response to sand fly salivary gland antigens: a useful epidemiological marker?, Am J Trop Med Hyg, vol.62, pp.740-745, 2000.

R. J. Nascimento, J. M. Santana, S. P. Lozzi, C. N. Araujo, and A. R. Teixeira, Human IgG1 and IgG4: the main antibodies against Triatoma infestans (Hemiptera: Reduviidae) salivary gland proteins, Am J Trop Med Hyg, vol.65, pp.219-226, 2001.

Z. Peng, A. N. Beckett, R. J. Engler, D. R. Hoffman, N. L. Ott et al., Immune responses to mosquito saliva in 14 individuals with acute systemic allergic reactions to mosquito bites, J Allergy Clin Immunol, vol.114, pp.1189-1194, 2004.

F. Remoue, B. Cisse, F. Ba, C. Sokhna, J. P. Herve et al., Evaluation of the antibody response to Anopheles salivary antigens as a potential marker of risk of malaria, Trans R Soc Trop Med Hyg, vol.100, pp.363-370, 2006.

E. Orlandi-pradines, L. Almeras, D. De-senneville, L. Barbe, S. Remoue et al., Antibody response against saliva antigens of Anopheles gambiae and Aedes aegypti in travellers in tropical, Africa. Microbes Infect, vol.9, pp.1454-1462, 2007.

A. Poinsignon, F. Remoue, M. Rossignol, S. Cornelie, D. Courtin et al., Human IgG antibody response to Glossina saliva: an epidemiologic marker of exposure to Glossina bites, Am J Trop Med Hyg, vol.78, pp.750-753, 2008.

L. Wasinpiyamongkol, S. Patramool, N. Luplertlop, P. Surasombatpattana, S. Doucoure et al., Blood-feeding and immunogenic Aedes aegypti saliva proteins, Proteomics, vol.10, pp.1906-1916, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00820653

F. Ødegaard, How many species of arthropods? Erwin's estimate revised, Biological Journal of the Linnean Society, vol.71, pp.583-597, 2000.

T. S. Adams, Hematophagy and Hormone Release, Annals of the Entomological Society of America, vol.92, pp.1-13, 1999.

Y. S. Balashov, Evolution of Haematophagy in Insects and Ticks. Entomological review, vol.79, pp.943-954, 1999.

W. C. Black and B. C. Kondratieff, Evolution of arthropod disease vectors, Marquardt WC, 2005.

M. J. Lehane, The Biology of Blood-Sucking in insects, 2005.

D. D. Chadee, J. C. Beier, and R. T. Mohammed, Fast and slow blood-feeding durations of Aedes aegypti mosquitoes in Trinidad, J Vector Ecol, vol.27, pp.172-177, 2002.

T. Krober and P. Guerin, The tick blood meal: from a living animal or from a silicone membrane, ALTEX, vol.24, pp.39-41, 2007.

N. J. Besansky, C. A. Hill, and C. Costantini, No accounting for taste: host preference in malaria vectors, Trends Parasitol, vol.20, pp.249-251, 2004.

M. A. Johansson, N. Arana-vizcarrondo, B. J. Biggerstaff, and J. E. Staples, Incubation periods of Yellow fever virus, Am J Trop Med Hyg, vol.83, pp.183-188, 2010.

A. Prakash, D. R. Bhattacharyya, P. K. Mohapatra, and J. Mahanta, Malaria transmission risk by the mosquito Anopheles baimaii (formerly known as An. dirus species D) at different hours of the night in North-east India, Med Vet Entomol, vol.19, pp.423-427, 2005.

C. J. Schofield, Vector population responses to control interventions, Ann Soc Belg Med Trop, vol.71, issue.1, pp.201-217, 1991.

K. K. Wu and P. Thiagarajan, Role of endothelium in thrombosis and hemostasis, Annu Rev Med, vol.47, pp.315-331, 1996.

G. Romney and M. Glick, An updated concept of coagulation with clinical implications, J Am Dent Assoc, vol.140, pp.567-574, 2009.

J. M. Ribeiro and R. H. Nussenzveig, The salivary catechol oxidase/peroxidase activities of the mosquito Anopheles albimanus, J Exp Biol, vol.179, pp.273-287, 1993.

E. A. Lerner, J. M. Ribeiro, R. J. Nelson, and M. R. Lerner, Isolation of maxadilan, a potent vasodilatory peptide from the salivary glands of the sand fly Lutzomyia longipalpis, J Biol Chem, vol.266, pp.11234-11236, 1991.

M. S. Cupp, J. M. Ribeiro, D. E. Champagne, and E. W. Cupp, Analyses of cDNA and recombinant protein for a potent vasoactive protein in saliva of a blood-feeding black fly, Simulium vittatum, J Exp Biol, vol.201, pp.1553-1561, 1998.

J. M. Ribeiro, O. Katz, L. K. Pannell, J. Waitumbi, and A. Warburg, Salivary glands of the sand fly Phlebotomus papatasi contain pharmacologically active amounts of adenosine and 5'-AMP, J Exp Biol, vol.202, pp.1551-1559, 1999.

D. B. Cines, E. S. Pollak, C. A. Buck, J. Loscalzo, G. A. Zimmerman et al., Endothelial cells in physiology and in the pathophysiology of vascular disorders, vol.91, pp.3527-3561, 1998.

. Fontaine, Parasites & Vectors, vol.4, p.187, 2011.

J. M. Ribeiro, J. J. Sarkis, P. A. Rossignol, and A. Spielman, Salivary apyrase of Aedes aegypti: characterization and secretory fate, Comp Biochem Physiol B, vol.79, pp.81-86, 1984.

J. J. Sarkis, J. A. Guimaraes, and J. M. Ribeiro, Salivary apyrase of Rhodnius prolixus. Kinetics and purification, Biochem J, vol.233, pp.885-891, 1986.

J. M. Ribeiro, J. A. Vaughan, and A. F. Azad, Characterization of the salivary apyrase activity of three rodent flea species, Comp Biochem Physiol B, vol.95, pp.215-219, 1990.

D. E. Champagne, C. T. Smartt, J. M. Ribeiro, and A. A. James, The salivary glandspecific apyrase of the mosquito Aedes aegypti is a member of the 5'-nucleotidase family, Proc Natl Acad Sci, vol.92, pp.694-698, 1995.

O. Marinotti, M. De-brito, and C. K. Moreira, Apyrase and alpha-glucosidase in the salivary glands of Aedes albopictus, Comp Biochem Physiol B Biochem Mol Biol, vol.113, pp.675-679, 1996.

B. J. Mans, A. R. Gaspar, A. I. Louw, and A. W. Neitz, Apyrase activity and platelet aggregation inhibitors in the tick Ornithodoros savignyi (Acari: Argasidae), Exp Appl Acarol, vol.22, pp.353-366, 1998.

J. G. Valenzuela, Y. Belkaid, E. Rowton, and J. M. Ribeiro, The salivary apyrase of the blood-sucking sand fly Phlebotomus papatasi belongs to the novel Cimex family of apyrases, J Exp Biol, vol.204, pp.229-237, 2001.

E. Faudry, S. P. Lozzi, J. M. Santana, D. Souza-ault, M. Kieffer et al., Triatoma infestans apyrases belong to the 5'-nucleotidase family, J Biol Chem, vol.279, pp.19607-19613, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02319043

H. E. Reno and R. J. Novak, Characterization of apyrase-like activity in Ochlerotatus triseriatus, Ochlerotatus hendersoni, and Aedes aegypti, Am J Trop Med Hyg, vol.73, pp.541-545, 2005.

R. Hamasaki, H. Kato, Y. Terayama, H. Iwata, and J. G. Valenzuela, Functional characterization of a salivary apyrase from the sand fly, Phlebotomus duboscqi, a vector of Leishmania major, J Insect Physiol, vol.55, pp.1044-1049, 2009.

I. M. Francischetti, J. M. Ribeiro, D. Champagne, and J. Andersen, Purification, cloning, expression, and mechanism of action of a novel platelet aggregation inhibitor from the salivary gland of the blood-sucking bug, Rhodnius prolixus, J Biol Chem, vol.275, pp.12639-12650, 2000.

G. C. Troy, An overview of hemostasis, Vet Clin North Am Small Anim Pract, vol.18, pp.5-20, 1988.

P. Carmeliet and . Biomedicine, Clotting factors build blood vessels, Science, vol.293, pp.1602-1604, 2001.

M. Levi, H. Cate, and T. Van-der-poll, Endothelium: interface between coagulation and inflammation, Crit Care Med, vol.30, pp.220-224, 2002.

H. Isawa, M. Yuda, Y. Orito, and Y. Chinzei, A mosquito salivary protein inhibits activation of the plasma contact system by binding to factor XII and high molecular weight kininogen, J Biol Chem, vol.277, pp.27651-27658, 2002.

H. Isawa, Y. Orito, S. Iwanaga, N. Jingushi, A. Morita et al., Identification and characterization of a new kallikrein-kinin system inhibitor from the salivary glands of the malaria vector mosquito Anopheles stephensi, Insect Biochem Mol Biol, vol.37, pp.466-477, 2007.

C. Canetti, J. S. Silva, S. H. Ferreira, and F. Q. Cunha, Tumour necrosis factor-alpha and leukotriene B(4) mediate the neutrophil migration in immune inflammation, Br J Pharmacol, vol.134, pp.1619-1628, 2001.

C. D. Ramos, C. Canetti, J. T. Souto, J. S. Silva, C. M. Hogaboam et al., MIP-1alpha[CCL3] acting on the CCR1 receptor mediates neutrophil migration in immune inflammation via sequential release of TNF-alpha and LTB4, J Leukoc Biol, vol.78, pp.167-177, 2005.

A. Chakravarti, I. Allaeys, and P. E. Poubelle,

, Med Sci (Paris), vol.23, pp.862-867, 2007.

J. G. Valenzuela, R. Charlab, T. N. Mather, and J. M. Ribeiro, Purification, cloning, and expression of a novel salivary anticomplement protein from the tick, Ixodes scapularis, J Biol Chem, vol.275, pp.18717-18723, 2000.

K. Tyson, C. Elkins, H. Patterson, E. Fikrig, and A. De-silva, Biochemical and functional characterization of Salp20, an Ixodes scapularis tick salivary protein that inhibits the complement pathway, Insect Mol Biol, vol.16, pp.469-479, 2007.

V. Daix, H. Schroeder, N. Praet, J. P. Georgin, I. Chiappino et al., Ixodes ticks belonging to the Ixodes ricinus complex encode a family of anticomplement proteins, Insect Mol Biol, vol.16, pp.155-166, 2007.

P. Gwakisa, K. Yoshihara, L. To, T. Gotoh, H. Amano et al., Salivary gland extract of Rhipicephalus appendiculatus ticks inhibits in vitro transcription and secretion of cytokines and production of nitric oxide by LPS-stimulated JA-4 cells, Vet Parasitol, vol.99, pp.53-61, 2001.

J. Kopecky and M. Kuthejlova, Suppressive effect of Ixodes ricinus salivary gland extract on mechanisms of natural immunity in vitro, Parasite Immunol, vol.20, pp.169-174, 1998.

M. Kubes, P. Kocakova, M. Slovak, M. Slavikova, N. Fuchsberger et al., Heterogeneity in the effect of different ixodid tick species on human natural killer cell activity, Parasite Immunol, vol.24, pp.23-28, 2002.

J. W. Hovius, M. A. De-jong, J. Den-dunnen, M. Litjens, E. Fikrig et al., Salp15 binding to DC-SIGN inhibits cytokine expression by impairing both nucleosome remodeling and mRNA stabilization, PLoS Pathog, vol.4, p.31, 2008.

J. Salat, G. C. Paesen, P. Rezacova, M. Kotsyfakis, Z. Kovarova et al., Crystal structure and functional characterization of an immunomodulatory salivary cystatin from the soft tick Ornithodoros moubata, Biochem J, vol.429, pp.103-112, 2010.

E. Y. Bissonnette, P. A. Rossignol, and A. D. Befus, Extracts of mosquito salivary gland inhibit tumour necrosis factor alpha release from mast cells, Parasite Immunol, vol.15, pp.27-33, 1993.

R. R. Cavalcante, M. H. Pereira, and N. F. Gontijo, Anti-complement activity in the saliva of phlebotomine sand flies and other haematophagous insects, Parasitology, vol.127, pp.87-93, 2003.

J. Sawynok, Adenosine receptor activation and nociception, Eur J Pharmacol, vol.347, pp.1-11, 1998.

R. Charlab, E. D. Rowton, and J. M. Ribeiro, The salivary adenosine deaminase from the sand fly Lutzomyia longipalpis, Exp Parasitol, vol.95, pp.45-53, 2000.

J. M. Ribeiro, R. Charlab, and J. G. Valenzuela, The salivary adenosine deaminase activity of the mosquitoes Culex quinquefasciatus and Aedes aegypti, J Exp Biol, vol.204, 2001.

T. M. Mohamed, Adenosine deaminase from camel tick Hyalomma dromedarii: purification and characterization, Exp Appl Acarol, vol.40, pp.101-111, 2006.

C. M. Theodos and R. G. Titus, Salivary gland material from the sand fly Lutzomyia longipalpis has an inhibitory effect on macrophage function in vitro, Parasite Immunol, vol.15, pp.481-487, 1993.

A. Sa-nunes, A. Bafica, D. A. Lucas, T. P. Conrads, T. D. Veenstra et al., Prostaglandin E2 is a major inhibitor of dendritic cell maturation and function in Ixodes scapularis saliva, J Immunol, vol.179, pp.1497-1505, 2007.

A. Skallova, G. Iezzi, F. Ampenberger, M. Kopf, and J. Kopecky, Tick saliva inhibits dendritic cell migration, maturation, and function while promoting development of Th2 responses, J Immunol, vol.180, pp.6186-6192, 2008.

S. Nithiuthai and J. R. Allen, Langerhans cells present tick antigens to lymph node cells from tick-sensitized guinea-pigs, Immunology, vol.55, pp.157-163, 1985.

D. T. Fearon and R. M. Locksley, The instructive role of innate immunity in the acquired immune response, Science, vol.272, pp.50-53, 1996.

G. W. Litman, J. P. Rast, and S. D. Fugmann, The origins of vertebrate adaptive immunity, Nat Rev Immunol, vol.10, pp.543-553, 2010.

R. N. Ramachandra and S. K. Wikel, Modulation of host-immune responses by ticks (Acari: Ixodidae): effect of salivary gland extracts on host macrophages and lymphocyte cytokine production, J Med Entomol, vol.29, pp.818-826, 1992.

S. Urioste, L. R. Hall, S. R. Telford, and R. G. Titus, Saliva of the Lyme disease vector, Ixodes dammini, blocks cell activation by a nonprostaglandin E2-dependent mechanism, J Exp Med, vol.180, pp.1077-1085, 1994.

M. L. Cross, E. W. Cupp, and F. J. Enriquez, Differential modulation of murine cellular immune responses by salivary gland extract of Aedes aegypti, Am J Trop Med Hyg, vol.51, pp.690-696, 1994.

H. A. Wasserman, S. Singh, and D. E. Champagne, Saliva of the Yellow Fever mosquito, Aedes aegypti, modulates murine lymphocyte function, Parasite Immunol, vol.26, pp.295-306, 2004.

M. Brossard and S. K. Wikel, Immunology of interactions between ticks and hosts, Med Vet Entomol, vol.11, pp.270-276, 1997.

M. L. Mbow, J. A. Bleyenberg, L. R. Hall, and R. G. Titus, Phlebotomus papatasi sand fly salivary gland lysate down-regulates a Th1, but up-regulates a Th2, response in mice infected with Leishmania major, J Immunol, vol.161, pp.5571-5577, 1998.

G. B. Schoeler, S. A. Manweiler, and S. K. Wikel, Ixodes scapularis: effects of repeated infestations with pathogen-free nymphs on macrophage and T lymphocyte cytokine responses of BALB/c and C3H/HeN mice, Exp Parasitol, vol.92, pp.239-248, 1999.

G. Caljon, J. Van-den-abbeele, J. M. Sternberg, M. Coosemans, D. Baetselier et al., Tsetse fly saliva biases the immune response to Th2 and Fontaine et al. Parasites & Vectors, vol.4, p.187, 2011.

, induces anti-vector antibodies that are a useful tool for exposure assessment, Int J Parasitol, vol.36, pp.1025-1035, 2006.

N. Mejri and M. Brossard, Splenic dendritic cells pulsed with Ixodes ricinus tick saliva prime naive CD4+T to induce Th2 cell differentiation in vitro and in vivo, Int Immunol, vol.19, pp.535-543, 2007.

R. G. Titus and J. M. Ribeiro, Salivary gland lysates from the sand fly Lutzomyia longipalpis enhance Leishmania infectivity, Science, vol.239, pp.1306-1308, 1988.

C. M. Theodos, J. M. Ribeiro, and R. G. Titus, Analysis of enhancing effect of sand fly saliva on Leishmania infection in mice, Infect Immun, vol.59, pp.1592-1598, 1991.

H. S. Bezerra and M. J. Teixeira, Effect of Lutzomyia whitmani (Diptera: Psychodidae) salivary gland lysates on Leishmania (Viannia) braziliensis infection in BALB/c mice, Mem Inst Oswaldo Cruz, vol.96, pp.349-351, 2001.

R. D. Mesquita, A. B. Carneiro, A. Bafica, F. Gazos-lopes, C. M. Takiya et al., Trypanosoma cruzi infection is enhanced by vector saliva through immunosuppressant mechanisms mediated by lysophosphatidylcholine, Infect Immun, vol.76, pp.5543-5552, 2008.

L. D. Jones, E. Hodgson, T. Williams, S. Higgs, and P. A. Nuttall, Saliva activated transmission (SAT) of Thogoto virus: relationship with vector potential of different haematophagous arthropods, Med Vet Entomol, vol.6, pp.261-265, 1992.

R. V. Morris, C. B. Shoemaker, J. R. David, G. C. Lanzaro, and R. G. Titus, Sandfly maxadilan exacerbates infection with Leishmania major and vaccinating against it protects against L. major infection, J Immunol, vol.167, pp.5226-5230, 2001.

N. Ramamoorthi, S. Narasimhan, U. Pal, F. Bao, X. F. Yang et al., The Lyme disease agent exploits a tick protein to infect the mammalian host, Nature, vol.436, pp.573-577, 2005.

A. C. Rocha, E. M. Braga, M. S. Araujo, B. S. Franklin, and P. F. Pimenta, Effect of the Aedes fluviatilis saliva on the development of Plasmodium gallinaceum infection in Gallus (gallus) domesticus, vol.99, pp.709-715, 2004.

S. Thangamani, S. Higgs, S. Ziegler, D. Vanlandingham, R. Tesh et al., Host immune response to mosquito-transmitted chikungunya virus differs from that elicited by needle inoculated virus, PLoS One, vol.5, p.12137, 2010.

B. S. Schneider, L. Soong, Y. A. Girard, G. Campbell, P. Mason et al., Potentiation of West Nile encephalitis by mosquito feeding, Viral Immunol, vol.19, pp.74-82, 2006.

L. M. Styer, P. Y. Lim, K. L. Louie, R. G. Albright, L. D. Kramer et al., Mosquito saliva causes enhancement of West Nile virus infection in mice, J Virol, vol.85, pp.1517-1527, 2011.

J. E. Osorio, M. S. Godsey, G. R. Defoliart, and T. M. Yuill, La Crosse viremias in whitetailed deer and chipmunks exposed by injection or mosquito bite, Am J Trop Med Hyg, vol.54, pp.338-342, 1996.

J. F. Edwards, S. Higgs, and B. J. Beaty, Mosquito feeding-induced enhancement of Cache Valley Virus (Bunyaviridae) infection in mice, J Med Entomol, vol.35, pp.261-265, 1998.

J. A. Vaughan, L. F. Scheller, R. A. Wirtz, and A. F. Azad, Infectivity of Plasmodium berghei sporozoites delivered by intravenous inoculation versus mosquito bite: implications for sporozoite vaccine trials, Infect Immun, vol.67, pp.4285-4289, 1999.

K. H. Limesand, S. Higgs, L. D. Pearson, and B. J. Beaty, Potentiation of vesicular stomatitis New Jersey virus infection in mice by mosquito saliva, Parasite Immunol, vol.22, pp.461-467, 2000.

V. Gillan and E. Devaney, Mosquito transmission modulates the immune response in mice infected with the L3 of Brugia pahangi, Parasite Immunol, vol.26, pp.359-363, 2004.

O. Theodor, A study of the reaction to phlebotomus bites with some remarks on "Harara, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol.29, pp.273-284, 1935.

Y. N. Dubin, J. D. Reese, and L. A. Seamans, Attempt to produce protection against mosquitoes by active immunization, J Immunol, vol.58, pp.293-297, 1948.

J. A. Mckiel and A. S. West, Effects of repeated exposures of hypersensitive humans and laboratory rabbits to mosquito antigen, Canadian Journal of Zoology, vol.39, pp.597-603, 1961.

A. B. Wilson and A. N. Clements, The Nature of the Skin Reaction to Mosquito Bites in Laboratory Animals, Int Arch Allergy Appl Immunol, vol.26, pp.294-314, 1965.

J. A. Ellis, S. Z. Shapiro, O. Moi-yoi, O. Moloo, and S. K. , Lesions and saliva-specific antibody responses in rabbits with immediate and delayed hypersensitivity reactions to the bites of Glossina morsitans centralis, Vet Pathol, vol.23, pp.661-667, 1986.

M. D. Chapman, N. A. Marshall, and A. Saxon, Identification and partial purification of species-specific allergens from Triatoma protracta (Heteroptera: Reduviidae), J Allergy Clin Immunol, vol.78, pp.436-442, 1986.

H. D. Shen, C. C. Chen, H. N. Chang, L. Y. Chang, W. C. Tu et al., Human IgE and IgG antibodies to mosquito proteins detected by the immunoblot technique, Ann Allergy, vol.63, pp.143-146, 1989.

M. Gauci, B. F. Stone, and Y. H. Thong, Isolation and immunological characterisation of allergens from salivary glands of the Australian paralysis tick Ixodes holocyclus, Int Arch Allergy Appl Immunol, vol.87, pp.208-212, 1988.

S. Wongkamchai, P. Khongtak, S. Leemingsawat, N. Komalamisra, N. Junsong et al., Comparative identification of protein profiles and major allergens of saliva, salivary gland and whole body extracts of mosquito species in Thailand, Asian Pac J Allergy Immunol, vol.28, pp.162-169, 2010.

L. Almeras, A. Fontaine, M. Belghazi, S. Bourdon, E. Boucomont-chapeaublanc et al., Salivary gland protein repertoire from Aedes aegypti mosquitoes. Vector Borne Zoonotic Dis, vol.10, pp.391-402, 2010.
URL : https://hal.archives-ouvertes.fr/pasteur-00516657

H. Brummer-korvenkontio, P. Lappalainen, T. Reunala, and T. Palosuo, Detection of mosquito saliva-specific IgE and IgG4 antibodies by immunoblotting, J Allergy Clin Immunol, vol.93, pp.551-555, 1994.

H. Brummer-korvenkontio, K. Palosuo, T. Palosuo, M. Brummer-korvenkontio, P. Leinikki et al., Detection of mosquito saliva-specific IgE antibodies by capture ELISA, Allergy, vol.52, pp.342-345, 1997.

Z. Peng and F. E. Simons, Cross-reactivity of skin and serum specific IgE responses and allergen analysis for three mosquito species with worldwide distribution, J Allergy Clin Immunol, vol.100, pp.192-198, 1997.

S. H. Jeon, J. W. Park, and B. H. Lee, Characterization of human IgE and mouse IgG1 responses to allergens in three mosquito species by immunoblotting and ELISA, Int Arch Allergy Immunol, vol.126, pp.206-212, 2001.

S. Cornelie, F. Remoue, S. Doucoure, T. Ndiaye, F. X. Sauvage et al., An insight into immunogenic salivary proteins of Anopheles gambiae in African children, Malar J, vol.6, p.75, 2007.
URL : https://hal.archives-ouvertes.fr/ird-00156556

A. Parmar, A. S. Grewal, and P. Dhillon, Immunological cross-reactivity between salivary gland proteins of Hyalomma anatolicum anatolicum and Boophilus microplus ticks, Vet Immunol Immunopathol, vol.51, pp.345-352, 1996.

N. A. Marshall, M. D. Chapman, and A. Saxon, Species-specific allergens from the salivary glands of Triatominae (Heteroptera:Reduviidae), J Allergy Clin Immunol, vol.78, pp.430-435, 1986.

Z. Peng, W. Xu, A. A. James, H. Lam, D. Sun et al., Expression, purification, characterization and clinical relevance of rAed a 1-a 68-kDa recombinant mosquito Aedes aegypti salivary allergen, Int Immunol, vol.13, pp.1445-1452, 2001.

Z. Peng, W. Xu, H. Lam, L. Cheng, A. A. James et al., A new recombinant mosquito salivary allergen, rAed a 2: allergenicity, clinical relevance, and cross-reactivity, Allergy, vol.61, pp.485-490, 2006.

E. Beaudouin, G. Kanny, J. M. Renaudin, and D. A. Moneret-vautrin, Allergen-specific immunotherapy to mosquitoes, Allergy, vol.56, p.787, 2001.

R. Ariano and R. C. Panzani, Efficacy and safety of specific immunotherapy to mosquito bites, Eur Ann Allergy Clin Immunol, vol.36, pp.131-138, 2004.

Z. Peng and F. E. Simons, Comparison of proteins, IgE, and IgG binding antigens, and skin reactivity in commercial and laboratory-made mosquito extracts, Ann Allergy Asthma Immunol, vol.77, pp.371-376, 1996.

J. F. Bell, S. J. Stewart, and S. K. Wikel, Resistance to tick-borne Francisella tularensis by tick-sensitized rabbits: allergic klendusity, Am J Trop Med Hyg, vol.28, pp.876-880, 1979.

S. K. Wikel, R. N. Ramachandra, D. K. Bergman, T. R. Burkot, and J. Piesman, Infestation with pathogen-free nymphs of the tick Ixodes scapularis induces host resistance to transmission of Borrelia burgdorferi by ticks, Infect Immun, vol.65, pp.335-338, 1997.

Y. Belkaid, S. Kamhawi, G. Modi, J. Valenzuela, N. Noben-trauth et al., Development of a natural model of cutaneous leishmaniasis: powerful effects of vector saliva and saliva preexposure on the long-term outcome of Leishmania major infection in the mouse ear dermis, J Exp Med, vol.188, pp.1941-1953, 1998.

M. J. Donovan, A. S. Messmore, D. A. Scrafford, D. L. Sacks, S. Kamhawi et al., Uninfected mosquito bites confer protection against infection with malaria parasites, Infect Immun, vol.75, pp.2523-2530, 2007.

. Fontaine, Parasites & Vectors, vol.4, p.187, 2011.

C. Kebaier, T. Voza, and J. Vanderberg, Neither mosquito saliva nor immunity to saliva has a detectable effect on the infectivity of Plasmodium sporozoites injected into mice, Infect Immun, vol.78, pp.545-551, 2010.

J. G. Valenzuela, Y. Belkaid, M. K. Garfield, S. Mendez, S. Kamhawi et al., Toward a defined anti-Leishmania vaccine targeting vector antigens: characterization of a protective salivary protein, J Exp Med, vol.194, pp.331-342, 2001.

F. Oliveira, P. G. Lawyer, S. Kamhawi, and J. G. Valenzuela, Immunity to distinct sand fly salivary proteins primes the anti-Leishmania immune response towards protection or exacerbation of disease, PLoS Negl Trop Dis, vol.2, p.226, 2008.

J. Dai, P. Wang, S. Adusumilli, C. J. Booth, S. Narasimhan et al., Antibodies against a tick protein, Salp15, protect mice from the Lyme disease agent, Cell Host Microbe, vol.6, pp.482-492, 2009.

A. C. Steere, V. K. Sikand, F. Meurice, D. L. Parenti, E. Fikrig et al., Vaccination against Lyme disease with recombinant Borrelia burgdorferi outer-surface lipoprotein A with adjuvant. Lyme Disease Vaccine Study Group, N Engl J Med, vol.339, pp.209-215, 1998.

B. S. Schneider, C. Mathieu, R. Peronet, and S. Mecheri, Anopheles stephensi saliva enhances progression of cerebral malaria in a murine model, Vector Borne Zoonotic Dis, vol.11, pp.423-432, 2011.

W. R. Ballou, The development of the RTS, S malaria vaccine candidate: challenges and lessons, Parasite Immunol, vol.31, pp.492-500, 2009.

D. Dodoo, F. Atuguba, S. Bosomprah, N. A. Ansah, P. Ansah et al., Antibody levels to multiple malaria vaccine candidate antigens in relation to clinical malaria episodes in children in the Kasena-Nankana district of Northern Ghana, Malar J, vol.10, p.108, 2011.

A. Mulenga, C. Sugimoto, Y. Sako, K. Ohashi, A. Musoke et al., Molecular characterization of a Haemaphysalis longicornis tick salivary gland-associated 29-kilodalton protein and its effect as a vaccine against tick infestation in rabbits, Infect Immun, vol.67, pp.1652-1658, 1999.

P. P. Prevot, B. Couvreur, V. Denis, M. Brossard, L. Vanhamme et al., Protective immunity against Ixodes ricinus induced by a salivary serpin, Vaccine, vol.25, pp.3284-3292, 2007.

S. Garcia-varas, R. Manzano-roman, P. Fernandez-soto, A. Encinas-grandes, A. Oleaga et al., Purification and characterisation of a Pselectin-binding molecule from the salivary glands of Ornithodoros moubata that induces protective anti-tick immune responses in pigs, Int J Parasitol, vol.40, pp.313-326, 2009.

J. De-la-fuente and K. M. Kocan, Advances in the identification and characterization of protective antigens for recombinant vaccines against tick infestations, Expert Rev Vaccines, vol.2, pp.583-593, 2003.

M. Labuda, A. R. Trimnell, M. Lickova, M. Kazimirova, G. M. Davies et al., An antivector vaccine protects against a lethal vector-borne pathogen, PLoS Pathog, vol.2, p.27, 2006.

J. De-la-fuente, C. Almazan, M. Canales, J. M. Perez-de-la-lastra, K. M. Kocan et al., A ten-year review of commercial vaccine performance for control of tick infestations on cattle, Anim Health Res Rev, vol.8, pp.23-28, 2007.

M. Chugh, T. Adak, N. Sehrawat, and S. K. Gakhar, Effect of anti-mosquito midgut antibodies on development of malaria parasite, Plasmodium vivax and fecundity in vector mosquito Anopheles culicifacies (Diptera: culicidae)

, Indian J Exp Biol, vol.49, pp.245-253, 2011.

C. E. Farrance, A. Rhee, R. M. Jones, K. Musiychuk, M. Shamloul et al., A Plant-Produced Pfs230 Vaccine Candidate Blocks Transmission of Plasmodium falciparum, Clin Vaccine Immunol, 2011.

T. Reunala, H. Brummer-korvenkontio, K. Palosuo, M. Miyanij, R. Ruiz-maldonado et al., Frequent occurrence of IgE and IgG4 antibodies against saliva of Aedes communis and Aedes aegypti mosquitoes in children, Int Arch Allergy Immunol, vol.104, pp.366-371, 1994.

A. D. Wilson, L. J. Harwood, S. Bjornsdottir, E. Marti, and M. J. Day, Detection of IgG and IgE serum antibodies to Culicoides salivary gland antigens in horses with insect dermal hypersensitivity (sweet itch), Equine Vet J, vol.33, pp.707-713, 2001.

Z. Peng, H. Li, and F. E. Simons, Immunoblot analysis of IgE and IgG binding antigens in extracts of mosquitos Aedes vexans, Culex tarsalis and Culiseta inornata, Int Arch Allergy Immunol, vol.110, pp.46-51, 1996.

A. Lotfy, R. A. Al-sherbiny, M. M. , S. Ali, and N. M. , Patients with papular urticaria have IgG antibodies to bedbug (Cimex lectularius) antigens, vol.98, pp.550-556, 2006.

B. S. Schwartz, J. M. Ribeiro, and M. D. Goldstein, Anti-tick antibodies: an epidemiologic tool in Lyme disease research, Am J Epidemiol, vol.132, pp.58-66, 1990.

K. Palosuo, H. Brummer-korvenkontio, J. Mikkola, T. Sahi, and T. Reunala, Seasonal increase in human IgE and IgG4 antisaliva antibodies to Aedes mosquito bites, Int Arch Allergy Immunol, vol.114, pp.367-372, 1997.

Z. Peng, N. Rasic, Y. Liu, and F. E. Simons, Mosquito saliva-specific IgE and IgG antibodies in 1059 blood donors, J Allergy Clin Immunol, vol.110, pp.816-817, 2002.

A. Schwarz, N. Medrano-mercado, P. F. Billingsley, G. A. Schaub, and J. M. Sternberg, IgM-antibody responses of chickens to salivary antigens of Triatoma infestans as early biomarkers for low-level infestation of triatomines, Int J Parasitol, vol.40, pp.1295-1302, 2010.

P. F. Billingsley, J. Baird, J. A. Mitchell, and C. Drakeley, Immune interactions between mosquitoes and their hosts, Parasite Immunol, vol.28, pp.143-153, 2006.

J. I. Shililu, W. A. Maier, H. M. Seitz, and A. S. Orago, Seasonal density, sporozoite rates and entomological inoculation rates of Anopheles gambiae and Anopheles funestus in a high-altitude sugarcane growing zone in Western Kenya, Trop Med Int Health, vol.3, pp.706-710, 1998.

D. L. Doolan, C. Dobano, and J. K. Baird, Acquired immunity to malaria, Clin Microbiol Rev, vol.22, pp.13-36, 2009.

P. M. Drame, A. Poinsignon, P. Besnard, L. Mire, J. et al., Human antibody response to Anopheles gambiae saliva: an immuno-epidemiological biomarker to evaluate the efficacy of insecticide-treated nets in malaria vector control, Am J Trop Med Hyg, vol.83, pp.115-121, 2010.

A. Schwarz, J. A. Juarez, J. Richards, B. Rath, V. Q. Machaca et al., Anti-triatomine saliva immunoassays for the evaluation of impregnated netting trials against Chagas disease transmission, Int J Parasitol, vol.41, pp.591-594, 2011.

T. Balenghien, F. Fouque, P. Sabatier, and D. J. Bicout, Horse-, bird-, and humanseeking behavior and seasonal abundance of mosquitoes in a West Nile virus focus of southern France, J Med Entomol, vol.43, pp.936-946, 2006.
URL : https://hal.archives-ouvertes.fr/pasteur-00308950

S. Toprak and N. Ozer, Distribution of sand fly (Diptera: Psychodidae) species and efficiency of capturing methods in Sanliurfa province, Turkey, J Med Entomol, vol.44, pp.23-28, 2007.

C. M. Wheeler, J. L. Coleman, and J. L. Benach, Salivary gland antigens of Ixodes dammini are glycoproteins that have interspecies cross-reactivity, J Parasitol, vol.77, pp.965-973, 1991.

R. T. Trevejo, W. K. Reisen, G. Yoshimura, and W. C. Reeves, Detection of chicken antibodies to mosquito salivary gland antigens by enzyme immunoassay, J Am Mosq Control Assoc, vol.21, pp.39-48, 2005.

J. L. Pinnas, R. E. Lindberg, T. M. Chen, and G. C. Meinke, Studies of kissing bugsensitive patients: evidence for the lack of cross-reactivity between Triatoma protracta and Triatoma rubida salivary gland extracts, J Allergy Clin Immunol, vol.77, pp.364-370, 1986.

P. Volf and I. Rohousova, Species-specific antigens in salivary glands of phlebotomine sandflies, Parasitology, vol.122, pp.37-41, 2001.

I. Rohousova, S. Ozensoy, Y. Ozbel, and P. Volf, Detection of species-specific antibody response of humans and mice bitten by sand flies, Parasitology, vol.130, pp.493-499, 2005.

P. Arensburger, K. Megy, R. M. Waterhouse, J. Abrudan, P. Amedeo et al., Sequencing of Culex quinquefasciatus establishes a platform for mosquito comparative genomics, Science, vol.330, pp.86-88, 2010.

R. A. Holt, G. M. Subramanian, A. Halpern, G. G. Sutton, R. Charlab et al., The genome sequence of the malaria mosquito Anopheles gambiae, Science, vol.298, pp.129-149, 2002.

E. Mongin, C. Louis, R. A. Holt, E. Birney, and F. H. Collins, The Anopheles gambiae genome: an update, Trends Parasitol, vol.20, pp.49-52, 2004.

C. A. Hill and S. K. Wikel, The Ixodes scapularis Genome Project: an opportunity for advancing tick research, Trends Parasitol, vol.21, pp.151-153, 2005.

V. Nene, J. R. Wortman, D. Lawson, B. Haas, C. Kodira et al., Genome sequence of Aedes aegypti, a major arbovirus vector, Science, vol.316, pp.1718-1723, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00156214

B. Arca, F. Lombardo, J. G. Valenzuela, I. M. Francischetti, O. Marinotti et al., An updated catalogue of salivary gland transcripts in the Fontaine et al. Parasites & Vectors, J Exp Biol, vol.208, pp.3971-3986, 2005.

A. Santos, J. M. Ribeiro, M. J. Lehane, N. F. Gontijo, A. B. Veloso et al., The sialotranscriptome of the blood-sucking bug Triatoma brasiliensis (Hemiptera, Triatominae), Insect Biochem Mol Biol, vol.37, pp.702-712, 2007.

B. Arca, F. Lombardo, I. M. Francischetti, V. M. Pham, M. Mestres-simon et al., An insight into the sialome of the adult female mosquito Aedes albopictus, Insect Biochem Mol Biol, vol.37, pp.107-127, 2007.

E. Calvo, V. M. Pham, O. Marinotti, J. F. Andersen, and J. M. Ribeiro, The salivary gland transcriptome of the neotropical malaria vector Anopheles darlingi reveals accelerated evolution of genes relevant to hematophagy, BMC Genomics, vol.10, p.57, 2009.

J. Ribeiro, B. J. Mans, and B. Arcà, An insight into the sialome of bloodfeeding Nematocera, Insect Biochemistry and Molecular Biology, vol.40, pp.767-784, 2010.

E. Calvo, I. Sanchez-vargas, A. J. Favreau, K. D. Barbian, V. M. Pham et al., An insight into the sialotranscriptome of the West Nile mosquito vector, Culex tarsalis, BMC Genomics, vol.11, p.51, 2010.

E. Anatriello, J. M. Ribeiro, I. K. De-miranda-santos, L. G. Brandao, J. M. Anderson et al., An insight into the sialotranscriptome of the brown dog tick, Rhipicephalus sanguineus, BMC Genomics, vol.11, p.450, 2010.

J. Alves-silva, J. M. Ribeiro, J. Van-den-abbeele, G. Attardo, Z. Hao et al., An insight into the sialome of Glossina morsitans morsitans, BMC Genomics, vol.11, p.213, 2010.

E. Calvo, A. Dao, V. M. Pham, and J. M. Ribeiro, An insight into the sialome of Anopheles funestus reveals an emerging pattern in anopheline salivary protein families, Insect Biochem Mol Biol, vol.37, pp.164-175, 2007.

L. Almeras, E. Orlandi-pradines, A. Fontaine, C. Villard, E. Boucomont et al., Sialome individuality between Aedes aegypti colonies. Vector Borne Zoonotic Dis, vol.9, pp.531-541, 2009.

S. Ben-hadj-ahmed, B. Kaabi, I. Chelbi, S. Cherni, M. Derbali et al., Colonization of Phlebotomus papatasi changes the effect of pre-immunization with saliva from lack of protection towards protection against experimental challenge with Leishmania major and saliva, Parasit Vectors, p.126, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00620944

M. D. Laurenti, V. L. Da-matta, T. Pernichelli, N. F. Secundino, L. C. Pinto et al., Effects of salivary gland homogenate from wild-caught and laboratory-reared Lutzomyia longipalpis on the evolution and immunomodulation of Leishmania (Leishmania) amazonensis infection, Scand J Immunol, vol.70, pp.389-395, 2009.

A. Fontaine, A. Pascual, I. Diouf, N. Bakkali, S. Bourdon et al., Mosquito salivary gland protein preservation in the field for immunological and biochemical analysis, Parasit Vectors, p.33, 2011.

P. Volf, P. Tesarova, and E. N. Nohynkova, Salivary proteins and glycoproteins in phlebotomine sandflies of various species, sex and age, Med Vet Entomol, vol.14, pp.251-256, 2000.

V. Choumet, A. Carmi-leroy, C. Laurent, P. Lenormand, J. C. Rousselle et al., The salivary glands and saliva of Anopheles gambiae as an essential step in the Plasmodium life cycle: a global proteomic study, Proteomics, vol.7, pp.3384-3394, 2007.
URL : https://hal.archives-ouvertes.fr/pasteur-00527461

D. B. Prates, L. D. Santos, J. C. Miranda, A. P. Souza, M. S. Palma et al., Changes in amounts of total salivary gland proteins of Lutzomyia longipallpis (Diptera: Psychodidae) according to age and diet, J Med Entomol, vol.45, pp.409-413, 2008.

F. Lombardo, R. Ronca, C. Rizzo, M. Mestres-simon, A. Lanfrancotti et al., The Anopheles gambiae salivary protein gSG6: an anopheline-specific protein with a blood-feeding role, Insect Biochem Mol Biol, vol.39, pp.457-466, 2009.

A. Poinsignon, S. Cornelie, M. Mestres-simon, A. Lanfrancotti, M. Rossignol et al., Novel peptide marker corresponding to salivary protein gSG6 potentially identifies exposure to Anopheles bites, PLoS One, vol.3, p.2472, 2008.

C. Rizzo, R. Ronca, G. Fiorentino, F. Verra, V. Mangano et al., Humoral response to the Anopheles gambiae salivary protein gSG6: a serological indicator of exposure to Afrotropical malaria vectors, PLoS One, vol.6, p.17980, 2011.

A. Poinsignon, B. Samb, S. Doucoure, P. M. Drame, J. B. Sarr et al., First attempt to validate the gSG6-P1 salivary peptide as an immuno-epidemiological tool for evaluating human exposure to Anopheles funestus bites, Trop Med Int Health, vol.15, pp.1198-1203, 2010.

M. L. Sanders, D. C. Jaworski, J. L. Sanchez, R. F. Defraites, G. E. Glass et al., Antibody to a cDNAderived calreticulin protein from Amblyomma americanum as a biomarker of tick exposure in humans, Am J Trop Med Hyg, vol.59, pp.279-285, 1998.

M. L. Sanders, G. E. Glass, R. B. Nadelman, G. P. Wormser, A. L. Scott et al., Antibody levels to recombinant tick calreticulin increase in humans after exposure to Ixodes scapularis (Say) and are correlated with tick engorgement indices, Am J Epidemiol, vol.149, pp.777-784, 1999.

C. Teixeira, R. Gomes, C. N. Reynoso, D. Jochim, R. Oliveira et al., Discovery of markers of exposure specific to bites of Lutzomyia longipalpis, the vector of Leishmania infantum chagasi in Latin America, PLoS Negl Trop Dis, vol.4, p.638, 2010.

A. P. Souza, B. B. Andrade, D. Aquino, P. Entringer, J. C. Miranda et al., Using recombinant proteins from Lutzomyia longipalpis saliva to estimate human vector exposure in visceral Leishmaniasis endemic areas, PLoS Negl Trop Dis, vol.4, p.649, 2010.

C. D. Paddock, J. H. Mckerrow, E. Hansell, K. W. Foreman, I. Hsieh et al., Identification, cloning, and recombinant expression of procalin, a major triatomine allergen, J Immunol, vol.167, pp.2694-2699, 2001.

J. Horackova, N. Rudenko, M. Golovchenko, and L. Grubhoffer, Der-p2 (Dermatophagoides pteronyssinus) allergen-like protein from the hard tick Ixodes ricinus -a novel member of ML (MD-2-related lipidrecognition) domain protein family, Parasitology, vol.137, pp.1139-1149, 2010.

C. Hilger, J. C. Bessot, N. Hutt, F. Grigioni, D. Blay et al., IgEmediated anaphylaxis caused by bites of the pigeon tick Argas reflexus: cloning and expression of the major allergen Arg r 1, J Allergy Clin Immunol, vol.115, pp.617-622, 2005.

G. Caljon, K. Broos, I. De-goeyse, D. Ridder, K. Sternberg et al., Den Abbeele JV: Identification of a functional Antigen5-related allergen in the saliva of a blood feeding insect, the tsetse fly, Insect Biochem Mol Biol, vol.39, pp.332-341, 2009.

R. Gomes, C. Teixeira, M. J. Teixeira, F. Oliveira, M. J. Menezes et al., Immunity to a salivary protein of a sand fly vector protects against the fatal outcome of visceral leishmaniasis in a hamster model, Proc Natl Acad Sci, vol.105, pp.7845-7850, 2008.

A. Poinsignon, S. Cornelie, F. Ba, D. Boulanger, C. Sow et al., Human IgG response to a salivary peptide, gSG6-P1, as a new immuno-epidemiological tool for evaluating low-level exposure to Anopheles bites, Malar J, vol.8, 0198.

J. W. Pickering, T. B. Martins, M. C. Schroder, and H. R. Hill, Comparison of a multiplex flow cytometric assay with enzyme-linked immunosorbent assay for auantitation of antibodies to tetanus, diphtheria, and Haemophilus influenzae Type b, Clin Diagn Lab Immunol, vol.9, pp.872-876, 2002.

G. Lal, P. Balmer, H. Joseph, M. Dawson, and R. Borrow, Development and evaluation of a tetraplex flow cytometric assay for quantitation of serum antibodies to Neisseria meningitidis serogroups A, C, Y, and W-135, Clin Diagn Lab Immunol, vol.11, pp.272-279, 2004.

E. Ambrosino, C. Dumoulin, E. Orlandi-pradines, F. Remoue, A. Toure-balde et al., A multiplex assay for the simultaneous detection of antibodies against 15 Plasmodium falciparum and Anopheles gambiae saliva antigens, Malar J, vol.9, p.317, 2010.

. Fontaine, Implication of haematophagous arthropod salivary proteins in host-vector interactions, Parasites & Vectors, vol.4, p.187, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02959162

. Fontaine, Parasites & Vectors, 2011.