J. De-la-fuente, A. Estrada-pena, J. M. Venzal, K. M. Kocan, and D. E. Sonenshine, Overview: Ticks as vectors of pathogens that cause disease in humans and animals, Front Biosci, vol.13, pp.6938-6984, 2008.

A. Rizzoli, C. Silaghi, A. Obiegala, R. I. Hubalek, Z. Foldvari et al., Ixodes ricinus and Its Transmitted Pathogens in Urban and Peri-Urban Areas in Europe: New Hazards and Relevance for Public Health. Front Public Health, vol.2, p.4248671, 2014.

L. Simo, M. Kazimirova, J. Richardson, and S. I. Bonnet, The Essential Role of Tick Salivary Glands and Saliva in Tick Feeding and Pathogen Transmission, Front Cell Infect Microbiol, vol.7, p.5479950, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01608039

J. De-la-fuente, S. Antunes, S. Bonnet, A. Cabezas-cruz, A. G. Domingos et al., Tick-Pathogen Interactions and Vector Competence: Identification of Molecular Drivers for Tick-Borne Diseases, Front Cell Infect Microbiol, vol.7, p.5383669, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01607008

X. Y. Liu, J. De-la-fuente, M. Cote, R. C. Galindo, S. Moutailler et al., IrSPI, a tick serine protease inhibitor involved in tick feeding and Bartonella henselae infection, PLoS Negl Trop Dis, vol.8, issue.7, 2014.

V. Cotte, S. Bonnet, L. Rhun, D. , L. Naour et al., Transmission of Bartonella henselae by Ixodes ricinus, Emerg Infect Dis, vol.14, issue.7, 2008.

A. A. Blisnick, T. Foulon, and S. I. Bonnet, Serine Protease Inhibitors in Ticks: An Overview of Their Role in Tick Biology and Tick-Borne Pathogen Transmission, Front Cell Infect Microbiol, vol.7, p.5438962, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01536659

R. E. Paul, M. Cote, L. Naour, E. Bonnet, and S. I. , Environmental factors influencing tick densities over seven years in a French suburban forest, Parasit Vectors, vol.9, issue.1, p.4884405, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01370808

M. Tonk, A. Cabezas-cruz, J. J. Valdes, R. O. Rego, L. Grubhoffer et al., Ixodes ricinus defensins attack distantly-related pathogens, Dev Comp Immunol, vol.53, issue.2, pp.358-65, 2015.

L. Simo, D. Zitnan, and Y. Park, Neural control of salivary glands in ixodid ticks, J Insect Physiol, vol.58, issue.4, pp.459-66, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01601792

J. Kotal, H. Langhansova, J. Lieskovska, J. F. Andersen, I. M. Francischetti et al., Modulation of host immunity by tick saliva, J Proteomics, vol.128, p.4619117, 2015.

I. M. Francischetti, T. N. Mather, and J. M. Ribeiro, Tick saliva is a potent inhibitor of endothelial cell proliferation and angiogenesis, Thromb Haemost, vol.94, issue.1, p.2893037, 2005.

B. J. Mans, A. I. Louw, and A. W. Neitz, Evolution of hematophagy in ticks: common origins for blood coagulation and platelet aggregation inhibitors from soft ticks of the genus Ornithodoros, Mol Biol Evol, vol.19, issue.10, pp.1695-705, 2002.

F. J. Alarcon-chaidez, U. U. Muller-doblies, and S. Wikel, Characterization of a recombinant immunomodulatory protein from the salivary glands of Dermacentor andersoni, Parasite Immunol, vol.25, issue.2, pp.69-77, 2003.

S. Konnai, H. Nishikado, S. Yamada, S. Imamura, T. Ito et al., Molecular identification and expression analysis of lipocalins from blood feeding taiga tick, Ixodes persulcatus Schulze, Exp Parasitol, vol.127, issue.2, pp.467-74, 2011.

R. Garcia-fernandez, M. Perbandt, D. Rehders, P. Ziegelmuller, N. Piganeau et al., Threedimensional Structure of a Kunitz-type Inhibitor in Complex with an Elastase-like Enzyme, J Biol Chem, vol.290, issue.22, pp.14154-65, 2015.

P. Central and P. , , p.4447985

T. A. Wilgus, S. Roy, and J. C. Mcdaniel, Neutrophils and Wound Repair: Positive Actions and Negative Reactions, Adv Wound Care, vol.2, issue.7, p.3763227, 2013.

D. Shah and K. Mital, The Role of Trypsin:Chymotrypsin in Tissue Repair, Adv Ther, vol.35, issue.1, p.5778189, 2018.

J. J. Valdes, A. Schwarz, C. De-vaca, I. Calvo, E. Pedra et al., Tryptogalinin is a tick Kunitz serine protease inhibitor with a unique intrinsic disorder, PLoS One, vol.8, issue.5, p.3643938, 2013.

X. Y. Liu and S. I. Bonnet, Hard tick factors implicated in pathogen transmission, PLoS Negl Trop Dis, vol.8, issue.1, p.3907338, 2014.

M. Kotsyfakis, A. Schwarz, J. Erhart, and J. M. Ribeiro, Tissue-and time-dependent transcription in Ixodes ricinus salivary glands and midguts when blood feeding on the vertebrate host, Sci Rep, vol.5, p.4357865, 2015.

J. Chmelar, J. M. Anderson, J. Mu, R. C. Jochim, J. G. Valenzuela et al., Insight into the sialome of the castor bean tick, Ixodes ricinus, BMC Genomics, vol.9, p.2410133, 2008.

S. X. Dai, A. D. Zhang, and J. F. Huang, Evolution, expansion and expression of the Kunitz/BPTI gene family associated with long-term blood feeding in Ixodes Scapularis, BMC Evol Biol, vol.12, p.3273431, 2012.

M. K. Islam, N. Tsuji, T. Miyoshi, M. A. Alim, X. Huang et al., The Kunitz-like modulatory protein haemangin is vital for hard tick blood-feeding success, PLoS Pathog, vol.5, issue.7, p.2701603, 2009.

T. S. Soares, F. Oliveira, R. J. Torquato, S. D. Sasaki, M. S. Araujo et al., BmTI-A, a Kunitz type inhibitor from Rhipicephalus microplus able to interfere in vessel formation, Vet Parasitol, vol.219, pp.44-52, 2016.

C. A. Lima, R. J. Torquato, S. D. Sasaki, G. Z. Justo, and A. S. Tanaka, Biochemical characterization of a Kunitz type inhibitor similar to dendrotoxins produced by Rhipicephalus (Boophilus) microplus (Acari: Ixodidae) hemocytes, Vet Parasitol, vol.167, issue.2-4, pp.279-87, 2010.

C. C. Drewes, R. Y. Dias, C. B. Hebeda, S. M. Simons, S. A. Barreto et al., Actions of the Kunitz-type serine protease inhibitor Amblyomin-X on VEGF-A-induced angiogenesis, Toxicon, vol.60, issue.3, pp.333-373, 2012.

E. W. Davie, K. Fujikawa, K. Kurachi, and W. Kisiel, The role of serine proteases in the blood coagulation cascade, Adv Enzymol Relat Areas Mol Biol, vol.48, pp.277-318, 1979.

N. Mejri, N. Franscini, B. Rutti, and M. Brossard, Th2 polarization of the immune response of BALB/c mice to Ixodes ricinus instars, importance of several antigens in activation of specific Th2 subpopulations, Parasite Immunol, vol.23, issue.2, pp.61-70, 2001.

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, issue.5, pp.818-844, 1992.

V. Rodrigues, B. Fernandez, A. Vercoutere, L. Chamayou, A. Andersen et al., Immunomodulatory Effects of Amblyomma variegatum Saliva on Bovine Cells: Characterization of Cellular Responses and Identification of Molecular Determinants, Front Cell Infect Microbiol, vol.7, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02073207

P. Central and P. , , p.5759025

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, issue.3, p.2191645, 1994.

H. C. Chang, S. Sehra, R. Goswami, W. Yao, Q. Yu et al., The transcription factor PU.1 is required for the development of IL-9-producing T cells and allergic inflammation, Nat Immunol, vol.11, issue.6, p.3136246, 2010.

J. Anguita, N. Ramamoorthi, J. W. Hovius, S. Das, V. Thomas et al., Salp15, an ixodes scapularis salivary protein, inhibits CD4(+) T cell activation, Immunity, vol.16, issue.6, pp.849-59, 2002.

G. Leboulle, M. Crippa, Y. Decrem, N. Mejri, M. Brossard et al., Characterization of a novel salivary immunosuppressive protein from Ixodes ricinus ticks, J Biol Chem, vol.277, issue.12, pp.10083-10092, 2002.

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, issue.9, pp.6186-92, 2008.

D. D. Taub, S. M. Turcovski-corrales, M. L. Key, D. L. Longo, and W. J. Murphy, Chemokines and T lymphocyte activation: I. Beta chemokines costimulate human T lymphocyte activation in vitro, J Immunol, vol.156, issue.6, pp.2095-103, 1996.

W. Elyaman, E. M. Bradshaw, C. Uyttenhove, V. Dardalhon, A. Awasthi et al., IL-9 induces differentiation of TH17 cells and enhances function of FoxP3+ natural regulatory T cells, Proc Natl Acad Sci U S A, vol.106, issue.31, pp.12885-90, 2009.

R. De-waal-malefyt, J. S. Abrams, S. M. Zurawski, J. C. Lecron, S. Mohan-peterson et al., Differential regulation of IL-13 and IL-4 production by human CD8+ and CD4+ Th0, Th1 and Th2 T cell clones and EBV-transformed B cells, Int Immunol, vol.7, issue.9, pp.1405-1421, 1995.

Z. Attia, J. C. Rowe, E. Kim, S. Varikuti, H. E. Steiner et al., Inhibitors of elastase stimulate murine B lymphocyte differentiation into IgG-and IgA-producing cells, Eur J Immunol, vol.48, issue.8, pp.1295-301, 2018.

P. Schneider, F. Mackay, V. Steiner, K. Hofmann, J. L. Bodmer et al., BAFF, a novel ligand of the tumor necrosis factor family, stimulates B cell growth, J Exp Med, vol.189, issue.11, p.2193079, 1999.

G. Yu, T. Boone, J. Delaney, N. Hawkins, M. Kelley et al., APRIL and TALL-I and receptors BCMA and TACI: system for regulating humoral immunity, Nat Immunol, vol.1, issue.3, pp.252-258, 2000.

L. Kovar, J. Kopecky, and B. Rihova, Salivary gland extract from Ixodes ricinus tick modulates the host immune response towards the Th2 cytokine profile, Parasitol Res, vol.88, issue.12, pp.1066-72, 2002.

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, issue.4, pp.535-578, 2007.

F. Ganapamo, B. Rutti, and M. Brossard, In vitro production of interleukin-4 and interferon-gamma by lymph node cells from BALB/c mice infested with nymphal Ixodes ricinus ticks, Immunology, vol.85, issue.1, pp.120-124, 1995.

N. Zeidner, M. Dreitz, D. Belasco, and D. Fish, Suppression of acute Ixodes scapularis-induced Borrelia burgdorferi infection using tumor necrosis factor-alpha, interleukin-2, and interferon-gamma, J Infect Dis, vol.173, issue.1, pp.187-95, 1996.

M. Thelen and J. V. Stein, How chemokines invite leukocytes to dance, Nat Immunol, vol.9, issue.9, pp.953-962, 2008.

J. Kopecky, M. Kuthejlova, and J. Pechova, Salivary gland extract from Ixodes ricinus ticks inhibits production of interferon-gamma by the upregulation of interleukin-10, Parasite Immunol, vol.21, issue.7, pp.351-357, 1999.

H. Horka, V. Staudt, M. Klein, C. Taube, S. Reuter et al., The tick salivary protein sialostatin L inhibits the Th9-derived production of the asthma-promoting cytokine IL-9 and is effective in the prevention of experimental asthma, J Immunol, vol.188, issue.6, p.3523721, 2012.

S. G. Preston, J. Majtan, C. Kouremenou, O. Rysnik, L. F. Burger et al., Novel immunomodulators from hard ticks selectively reprogramme human dendritic cell responses
URL : https://hal.archives-ouvertes.fr/hal-01870572

, PLoS Pathog, vol.9, issue.6, p.1003450, 2013.

P. Central and P. , , p.3695081

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, issue.1, p.3523712, 2010.

C. Coeshott, C. Ohnemus, A. Pilyavskaya, S. Ross, M. Wieczorek et al., Converting enzymeindependent release of tumor necrosis factor alpha and IL-1beta from a stimulated human monocytic cell line in the presence of activated neutrophils or purified proteinase 3, Proc Natl Acad Sci U S A, vol.96, issue.11, p.26869, 1999.

K. Kwiatkowski and J. Mika, The importance of chemokines in neuropathic pain development and opioid analgesic potency, Pharmacol Rep, vol.70, issue.4, pp.821-851, 2018.

M. D. Turner, B. Nedjai, T. Hurst, and D. J. Pennington, Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease, Biochim Biophys Acta, vol.1843, issue.11, pp.2563-82, 2014.

N. Suffee, H. Hlawaty, A. Meddahi-pelle, L. Maillard, L. Louedec et al., RANTES/CCL5-induced pro-angiogenic effects depend on CCR1, CCR5 and glycosaminoglycans, Angiogenesis, vol.15, issue.4, pp.727-771, 2012.

Y. Adachi, C. Aoki, N. Yoshio-hoshino, K. Takayama, D. T. Curiel et al., Interleukin-6 induces both cell growth and VEGF production in malignant mesotheliomas, Int J Cancer, vol.119, issue.6, pp.1303-1314, 2006.

K. S. Mark, W. J. Trickler, and D. W. Miller, Tumor necrosis factor-alpha induces cyclooxygenase-2 expression and prostaglandin release in brain microvessel endothelial cells, J Pharmacol Exp Ther, vol.297, issue.3, pp.1051-1059, 2001.

R. Salcedo, H. A. Young, M. L. Ponce, J. M. Ward, H. K. Kleinman et al., Eotaxin (CCL11) induces in vivo angiogenic responses by human CCR3+ endothelial cells, J Immunol, vol.166, issue.12, pp.7571-7579, 2001.

B. Arendse, J. Van-snick, and F. Brombacher, IL-9 is a susceptibility factor in Leishmania major infection by promoting detrimental Th2/type 2 responses, J Immunol, vol.174, issue.4, pp.2205-2216, 2005.

L. Barron, H. Dooms, K. K. Hoyer, W. Kuswanto, J. Hofmann et al., Cutting edge: mechanisms of IL-2-dependent maintenance of functional regulatory T cells, J Immunol, vol.144, issue.11, p.3059533, 2010.

T. Chinen, A. K. Kannan, A. G. Levine, X. Fan, U. Klein et al., An essential role for the IL-2 receptor in Treg cell function, Nat Immunol, vol.17, issue.11, p.5071159, 2016.

D. K. Brake and A. A. Perez-de-leon, Immunoregulation of bovine macrophages by factors in the salivary glands of Rhipicephalus microplus, Parasit Vectors, vol.5, p.3320552, 2012.

K. Takatsu, Interleukin-5 and IL-5 receptor in health and diseases, Proc Jpn Acad Ser B Phys Biol Sci, vol.87, issue.8, pp.463-85, 2011.

P. A. Nuttall, A. R. Trimnell, M. Kazimirova, and M. Labuda, Exposed and concealed antigens as vaccine targets for controlling ticks and tick-borne diseases, Parasite Immunol, vol.28, issue.4, pp.155-63, 2006.

T. K. Kim, L. Tirloni, A. F. Pinto, J. Moresco, J. R. Yates et al., Ixodes scapularis Tick Saliva Proteins Sequentially Secreted Every 24 h during Blood Feeding, PLoS Negl Trop Dis, vol.10, issue.1, p.4709002, 2016.

S. Bonnet, M. Jouglin, L. Malandrin, C. Becker, A. Agoulon et al., Transstadial and transovarial persistence of Babesia divergens DNA in Ixodes ricinus ticks fed on infected blood in a new skin-feeding technique, Parasitology, vol.134, issue.2, pp.197-207, 2007.

A. Micsonai, F. Wien, L. Kernya, Y. H. Lee, Y. Goto et al., Accurate secondary structure prediction and fold recognition for circular dichroism spectroscopy, Proc Natl Acad Sci, vol.112, issue.24, p.4475991, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01485547

P. Schuck, Size-distribution analysis of macromolecules by sedimentation velocity ultracentrifugation and lamm equation modeling, Biophys J, vol.78, issue.3, pp.1606-1625, 2000.

C. Reis, M. Cote, L. Rhun, D. Lecuelle, B. Levin et al., Vector competence of the tick Ixodes ricinus for transmission of Bartonella birtlesii, PLoS Negl Trop Dis, vol.5, issue.5, p.3104967, 2011.

M. Bugarel, L. Beutin, F. Scheutz, E. Loukiadis, and P. Fach, Identification of genetic markers for differentiation of Shiga toxin-producing, enteropathogenic, and avirulent strains of Escherichia coli O26, Appl Environ Microbiol, vol.77, issue.7, p.3067419, 2011.

C. Almazan, S. Bonnet, M. Cote, M. Slovak, Y. Park et al., A Versatile Model of Hard Tick Infestation on Laboratory Rabbits, J Vis Exp, issue.140, 2018.

P. Central and P. ,

T. G. Patton, G. Dietrich, K. Brandt, M. C. Dolan, J. Piesman et al., Saliva, salivary gland, and hemolymph collection from Ixodes scapularis ticks, J Vis Exp, issue.60, 2012.

T. D. Schmittgen and K. J. Livak, Analyzing real-time PCR data by the comparative C(T) method, Nat Protoc, vol.3, issue.6, pp.1101-1109, 2008.

J. Koci, L. Simo, and Y. Park, Validation of internal reference genes for real-time quantitative polymerase chain reaction studies in the tick, Ixodes scapularis (Acari: Ixodidae), J Med Entomol, vol.50, issue.1, p.3703510, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01601685

L. Simo, M. Slovak, Y. Park, and D. Zitnan, Identification of a complex peptidergic neuroendocrine network in the hard tick, Rhipicephalus appendiculatus, Cell Tissue Res, vol.335, issue.3, p.3573535, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01601260

G. Jourdi, V. Siguret, A. C. Martin, J. L. Golmard, A. Godier et al., Association rate constants rationalise the pharmacodynamics of apixaban and rivaroxaban, Thromb Haemost, vol.114, issue.1, pp.78-86, 2015.

G. Jourdi, I. Gouin-thibault, V. Siguret, S. Gandrille, P. Gaussem et al., FXa-alpha2-Macroglobulin Complex Neutralizes Direct Oral Anticoagulants Targeting FXa In Vitro and In Vivo, Thromb Haemost, vol.118, issue.9, pp.1535-1579, 2018.

A. Carreau, C. Kieda, and C. Grillon, Nitric oxide modulates the expression of endothelial cell adhesion molecules involved in angiogenesis and leukocyte recruitment, Exp Cell Res, vol.317, issue.1, pp.29-41, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00608218

M. Paprocka, A. Krawczenko, D. Dus, A. Kantor, A. Carreau et al., CD133 positive progenitor endothelial cell lines from human cord blood, Cytometry A, vol.79, issue.8, pp.594-602, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00720684

G. Carpentier, Angiogenesis analyser tool for Image J, 2012.

L. A. Kelley, S. Mezulis, C. M. Yates, M. N. Wass, and M. J. Sternberg, The Phyre2 web portal for protein modeling, prediction and analysis, Nat Protoc, vol.10, issue.6, p.5298202, 2015.

S. Tirard, Haldane and the origin of life, J. B. S, vol.96, pp.735-739, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01653001

Z. Q. Zhang, Animal biodiversity: An outline of higher-level classification and survey of taxonomic richness, Zootaxa, vol.3703, pp.1-82, 2013.

K. A. Lanier, A. S. Petrov, and L. D. Williams, The Central Symbiosis of Molecular Biology: Molecules in Mutualism, J Mol Evol, vol.85, pp.8-13, 2017.

V. Ahmadjian and J. B. Jacobs, Relationship between fungus and alga in the lichen Cladonia cristatella Tuck, Nature, vol.289, p.169, 1981.

F. Backhed, R. E. Ley, J. L. Sonnenburg, D. A. Peterson, and J. I. Gordon, Host-bacterial mutualism in the human intestine, Science, vol.307, pp.1915-1935, 2005.

B. J. Mans, D. De-klerk, R. Pienaar, and A. A. Latif, Nuttalliella namaqua: a living fossil and closest relative to the ancestral tick lineage: implications for the evolution of blood-feeding in ticks, PLoS One, vol.6, p.23675, 2011.

J. De-la-fuente, The fossil record and the origin of ticks (Acari: Parasitiformes: Ixodida), vol.29, pp.331-375, 2003.

E. Penalver, A. Arillo, X. Delclos, D. Peris, D. A. Grimaldi et al., parasitised feathered dinosaurs as revealed by Cretaceous amber assemblages, Nat Commun, vol.8, p.1924, 2017.

F. Van-oefele, Studien Über Die Altägyptische Parasitologie, Arch. Parsitol, vol.04, pp.481-530, 1901.

H. , Bibliography of ticks and tickborne diseases : from Homer (about 800 B.C.) to 31, 1970.

T. Smith and F. L. Kilbourne, Investigations into the nature, causation and prevention of Texas or southern cattle fever, Bur Anim Ind Bull, vol.1, pp.301-324, 1893.

B. Pomerantsev, Basic directions of evolution in the Ixodoidea, Parazitologicheskii Sbornik, vol.10, pp.5-19, 1948.

J. Camicas and P. Morel, Position systématique et classification des tiques (Acarida: Ixodida), Acarologia, vol.18, pp.410-420, 1977.

S. C. Barker and A. Murrell, Systematics and evolution of ticks with a list of valid genus and species names, Parasitology, vol.129, pp.15-36, 2004.

F. Jongejan and G. Uilenberg, The global importance of ticks, Parasitology, vol.129, pp.3-14, 2004.

J. De-la-fuente, A. Estrada-pena, J. M. Venzal, K. M. Kocan, and D. E. Sonenshine, Overview: Ticks as vectors of pathogens that cause disease in humans and animals, Front Biosci, vol.13, pp.6938-6984, 2008.

E. Lindgren, Y. Andersson, J. E. Suk, B. Sudre, and J. C. Semenza, Public health. Monitoring EU emerging infectious disease risk due to climate change, Science, vol.336, pp.418-427, 2012.

A. R. Walker, Amblyomma tick feeding in relation to host health, Trop Anim Health Prod, vol.28, pp.26-28, 1996.

R. Gothe, K. Kunze, and H. Hoogstraal, The mechanisms of pathogenicity in the tick paralyses, J Med Entomol, vol.16, pp.357-69, 1979.

J. C. Bessot, M. C. Kopferschmitt, F. Blay, A. Dietemann, A. Nirrengarten et al., Chocs anaphylactiques après morsure de tiques de pigeon (Argas reflexus). À propos de douze cas, Revue Française d'Allergologie et d'Immunologie Clinique, vol.37, pp.431-437

A. Guillier, A. Fauconneau, F. De, S. Barruel, M. S. Guez et al., Allergic hypersensitivity to red meat induced by tick bites: a French case report, Eur J Dermatol, vol.25, p.277, 0203.

J. W. Steinke, T. A. Platts-mills, and S. P. Commins, The alpha-gal story: Lessons learned from connecting the dots, Journal of Allergy and Clinical Immunology, vol.135, pp.589-596, 2015.

K. E. Jones, N. G. Patel, M. A. Levy, A. Storeygard, D. Balk et al., Global trends in emerging infectious diseases, Nature, vol.451, pp.990-993, 2008.

J. De-la-fuente, S. Antunes, S. Bonnet, A. Cabezas-cruz, A. G. Domingos et al., Tick-Pathogen Interactions and Vector Competence: Identification of Molecular Drivers for Tick-Borne Diseases, Front Cell Infect Microbiol, vol.7, p.114, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01607008

S. Bonnet, K. Huber, G. Joncour, M. René-martellet, F. Stachurski et al., Biologie des tiques, Tiques et Maladies à tiques, : une introduction multidisciplinaire, p.336, 2016.

A. Rizzoli, C. Silaghi, A. Obiegala, I. Rudolf, Z. Hubálek et al., Ixodes ricinus and its transmitted pathogens in urban and peri-urban areas in Europe: new hazards and relevance for public health, Frontiers in Public Health, vol.2, 2014.

R. Farkas, A. Estrada-peña, T. Jaenson, I. Pascucci, and M. Madder, Basic biology and geographical distribution of tick species involved in the transmission of animal pathogens, including zoonoses.," in Ticks tick-borne Dis, Geogr. Distrib. Control Strateg. euro-asia, pp.6-26, 2013.

S. Moutailler, J. George, Y. Hansmann, B. Degeilh, G. Joncour et al., Principales Maladies Transmises par les Tiques: Epidemiologie, Clinique Et Diagnostic " in Tiques et Maladies à tiques, p.336, 2016.

S. Bonnet, M. Jouglin, L. Malandrin, C. Becker, A. Agoulon et al., Transstadial and transovarial persistence of Babesia divergens DNA in Ixodes ricinus ticks fed on infected blood in a new skin-feeding technique, Parasitology, vol.134, pp.197-207, 2007.

K. L. Mansfield, N. Johnson, L. P. Phipps, J. R. Stephenson, A. R. Fooks et al., Tickborne encephalitis virus -a review of an emerging zoonosis, J Gen Virol, vol.90, pp.1781-94, 2009.

V. Cotte, S. Bonnet, D. Le-rhun, E. L. Naour, A. Chauvin et al., Transmission of Bartonella henselae by Ixodes ricinus, Emerg Infect Dis, vol.14, pp.1074-80, 2008.

C. Reis, M. Cote, D. Le-rhun, B. Lecuelle, M. L. Levin et al., Vector competence of the tick Ixodes ricinus for transmission of Bartonella birtlesii, PLoS Negl Trop Dis, vol.5, p.1186, 2011.

J. Ohnishi, J. Piesman, and A. M. Silva, Antigenic and genetic heterogeneity of Borrelia burgdorferi populations transmitted by ticks, Proc Natl Acad Sci U S A, vol.98, pp.670-675, 2001.

L. Eisen, Pathogen transmission in relation to duration of attachment by Ixodes scapularis ticks, Ticks Tick Borne Dis, vol.9, pp.535-542, 2018.

P. A. Dahm, Toxic effects produced in insects by organophosphorus compounds, Bull World Health Organ, vol.44, pp.215-224, 1971.

J. E. George, Present and future technologies for tick control, Ann N Y Acad Sci, vol.916, pp.583-591, 2000.

L. G. Costa, The neurotoxicity of organochlorine and pyrethroid pesticides, Handb Clin Neurol, vol.131, pp.135-183, 2015.

H. Mehlhorn, B. Schumacher, A. Jatzlau, F. Abdel-ghaffar, K. A. Al-rasheid et al., Efficacy of deltamethrin (Butox(R) 7.5 pour on) against nymphs and adults of ticks (Ixodes ricinus, Rhipicephalus sanguineus) in treated hair of cattle and sheep, Parasitol Res, vol.108, pp.963-71, 2011.

J. B. Prullage, H. V. Tran, P. Timmons, J. Harriman, S. T. Chester et al., The combined mode of action of fipronil and amitraz on the motility of Rhipicephalus sanguineus, Vet Parasitol, vol.179, pp.302-312, 2011.

P. R. De-oliveira, I. B. Calligaris, P. H. Nunes, G. H. Bechara, and M. I. Camargo-mathias, Fluazuron-induced morphological changes in Rhipicephalus sanguineus Latreille, 1806 (Acari: Ixodidae) nymphs: An ultra-structural evaluation of the cuticle formation and digestive processes, Acta Trop, vol.133, pp.45-55, 2014.

L. V. Gomes, W. D. Lopes, B. C. Cruz, W. F. Teixeira, G. Felippelli et al., Acaricidal effects of fluazuron (2.5 mg/kg) and a combination of fluazuron (1.6 mg/kg) + ivermectin (0.63 mg/kg), administered at different routes, against Rhipicephalus (Boophilus) microplus parasitizing cattle, Exp Parasitol, vol.153, pp.22-30, 2015.

J. F. Graf, R. Gogolewski, N. Leach-bing, G. A. Sabatini, M. B. Molento et al., Tick control: an industry point of view, Parasitology, vol.129, pp.427-469, 2004.

E. N. Mwangi, S. M. Hassan, G. P. Kaaya, and S. Essuman, The impact of Ixodiphagus hookeri, a tick parasitoid, on Amblyomma variegatum (Acari: Ixodidae) in a field trial in Kenya, Exp Appl Acarol, vol.21, pp.117-143, 1997.

M. Sun, Q. Ren, G. Guan, Y. Li, X. Han et al., Effectiveness of Beauveria bassiana sensu lato strains for biological control against Rhipicephalus (Boophilus) microplus (Acari: Ixodidae) in China, Parasitol Int, vol.62, pp.412-417, 2013.

E. A. Suleiman, M. T. Shigidi, and S. M. Hassan, Metarhizium anisopliae as a biological control agent against Hyalomma anatolicum (Acari: Ixodidae), Pak J Biol Sci, vol.16, pp.1943-1952, 2013.

M. Samish, H. Ginsberg, and I. Glazer, Biological control of ticks, vol.129

D. E. Sonenshine, T. Adams, S. A. Allan, J. Mclaughlin, and F. X. Webster, Chemical composition of some components of the arrestment pheromone of the black-legged tick, Ixodes scapularis (Acari: Ixodidae) and their use in tick control, J Med Entomol, vol.40, pp.849-59, 2003.

Z. Hubalek, J. Halouzka, Z. Juricova, S. Sikutova, and I. Rudolf, Effect of forest clearing on the abundance of Ixodes ricinus ticks and the prevalence of Borrelia burgdorferi s.l, Med Vet Entomol, vol.20, pp.166-72, 2006.

A. Milne, Pasture improvement and the control of sheep tick, Ixodes ricinus L, Ann Appl Biol, vol.35, pp.369-78, 1948.

J. De-la-fuente, C. Almazan, V. Naranjo, E. F. Blouin, J. M. Meyer et al., Autocidal control of ticks by silencing of a single gene by RNA interference, Biochem Biophys Res Commun, vol.344, pp.332-340, 2006.

J. De-la-fuente and O. Merino, Vaccinomics, the new road to tick vaccines, Vaccine, vol.31, pp.5923-5932, 2013.

P. A. Nuttall, A. R. Trimnell, M. Kazimirova, and M. Labuda, Exposed and concealed antigens as vaccine targets for controlling ticks and tick-borne diseases, Parasite Immunol, vol.28, pp.155-63, 2006.

X. Y. Liu and S. I. Bonnet, Hard tick factors implicated in pathogen transmission, PLoS Negl Trop Dis, vol.8, p.2566, 2014.

J. R. Allen and S. J. Humphreys, Immunisation of guinea pigs and cattle against ticks, Nature, vol.280, pp.491-494, 91979-08.

W. Trager, Acquired Immunity to Ticks, The Journal of Parasitology, vol.25, pp.57-81, 1939.

J. A. Roberts, Resistance of cattle to the tick boophilus microplus (canestrini). II. Stages of the life cycle of the parasite against which resistance is manifest, J Parasitol, vol.54, pp.667-73, 1968.

P. Willadsen, R. V. Mckenna, and G. A. Riding, Isolation from the cattle tick, Boophilus microplus, of antigenic material capable of eliciting a protective immunological response in the bovine host, Int J Parasitol, vol.18, pp.183-192, 1988.

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.23-31, 2007.

P. Willadsen, P. Bird, G. S. Cobon, and J. Hungerford, Commercialisation of a recombinant vaccine against Boophilus microplus, Parasitology, vol.110, pp.43-50, 1995.

M. Canales, A. Enriquez, E. Ramos, D. Cabrera, H. Dandie et al., Large-scale production in Pichia pastoris of the recombinant vaccine Gavac against cattle tick, Vaccine, vol.15, pp.414-436, 1997.

R. Andreotti, Performance of two Bm86 antigen vaccin formulation against tick using crossbreed bovines in stall test, Rev Bras Parasitol Vet, vol.15, pp.97-100, 2006.

J. C. Garcia-garcia, C. Montero, M. Redondo, M. Vargas, M. Canales et al., Control of ticks resistant to immunization with Bm86 in cattle vaccinated with the recombinant antigen Bm95 isolated from the cattle tick, Boophilus microplus, Vaccine, vol.18, pp.2275-87, 2000.

S. Ghosh, D. D. Ray, G. Vanlahmuaka, N. K. Das, J. K. Singh et al., Progress in development of vaccine against Hyalomma anatolicum anatolicum-Indian scenario, Vaccine, vol.26, pp.40-47, 2008.

H. Fragoso, P. H. Rad, M. Ortiz, M. Rodriguez, M. Redondo et al., Protection against Boophilus annulatus infestations in cattle vaccinated with the B. microplus Bm86-containing vaccine Gavac. off, Vaccine, vol.16, pp.1990-1992, 1998.

D. Odongo, L. Kamau, R. Skilton, S. Mwaura, C. Nitsch et al., Vaccination of cattle with TickGARD induces cross-reactive antibodies binding to conserved linear peptides of Bm86 homologues in Boophilus decoloratus, Vaccine, vol.25, pp.1287-96, 2007.

M. Canales, C. Almazan, V. Naranjo, F. Jongejan, and J. De-la-fuente, Vaccination with recombinant Boophilus annulatus Bm86 ortholog protein, Ba86, protects cattle against B. annulatus and B. microplus infestations, BMC Biotechnol, vol.9, p.29, 2009.

L. Jeyabal, P. Azhahianambi, K. Susitha, D. D. Ray, P. Chaudhuri et al., Efficacy of rHaa86, an orthologue of Bm86, against challenge infestations of Hyalomma anatolicum anatolicum, Transbound Emerg Dis, vol.57, pp.96-102, 2010.

A. M. Nijhof, J. A. Balk, M. Postigo, A. M. Rhebergen, A. Taoufik et al., Bm86 homologues and novel ATAQ proteins with multiple epidermal growth factor (EGF)-like domains from hard and soft ticks, Int J Parasitol, vol.40, pp.1587-97, 2010.

O. Hajdusek, D. Sojka, P. Kopacek, V. Buresova, Z. Franta et al., Knockdown of proteins involved in iron metabolism limits tick reproduction and development, Proc Natl Acad Sci U S A, vol.106, pp.1033-1041, 2009.

O. Hajdusek, C. Almazan, G. Loosova, M. Villar, M. Canales et al., Characterization of ferritin 2 for the control of tick infestations, Vaccine, vol.28, pp.2993-3001, 2010.

R. C. Galindo, E. Doncel-perez, Z. Zivkovic, V. Naranjo, C. Gortazar et al., Tick subolesin is an ortholog of the akirins described in insects and vertebrates, Dev Comp Immunol, vol.33, pp.612-619, 2009.

J. De-la-fuente, J. A. Moreno-cid, M. Canales, M. Villar, J. M. De-la-lastra et al., Targeting arthropod subolesin/akirin for the development of a universal vaccine for control of vector infestations and pathogen transmission, Vet Parasitol, vol.181, pp.17-22, 2011.

K. M. Kocan, R. Manzano-roman, and J. De-la-fuente, Transovarial silencing of the subolesin gene in three-host ixodid tick species after injection of replete females with subolesin dsRNA, Parasitol Res, vol.100, pp.1411-1416, 2007.

J. De-la-fuente, C. Almazan, E. F. Blouin, V. Naranjo, and K. M. Kocan, Reduction of tick infections with Anaplasma marginale and A. phagocytophilum by targeting the tick protective antigen subolesin, Parasitol Res, vol.100, pp.85-91, 2006.

O. Merino, C. Almazan, M. Canales, M. Villar, J. A. Moreno-cid et al., Targeting the tick protective antigen subolesin reduces vector infestations and pathogen infection by Anaplasma marginale and Babesia bigemina, Vaccine, vol.29, pp.8575-8584, 2011.

M. Bensaci, D. Bhattacharya, R. Clark, and L. T. Hu, Oral vaccination with vaccinia virus expressing the tick antigen subolesin inhibits tick feeding and transmission of Borrelia burgdorferi, Vaccine, vol.30, pp.6040-6046, 2012.

O. Merino, C. Almazan, M. Canales, M. Villar, J. A. Moreno-cid et al., Control of Rhipicephalus (Boophilus) microplus infestations by the combination of subolesin vaccination and tick autocidal control after subolesin gene knockdown in ticks fed on cattle, Vaccine, vol.29, pp.2248-54, 2011.

A. R. Trimnell, R. S. Hails, and P. A. Nuttall, Dual action ectoparasite vaccine targeting 'exposed' and 'concealed' antigens, Vaccine, vol.20, pp.3560-3568, 2002.

A. R. Trimnell, G. M. Davies, O. Lissina, R. S. Hails, and P. A. Nuttall, A cross-reactive tick cement antigen is a candidate broad-spectrum tick vaccine, Vaccine, vol.23, pp.4329-4370, 2005.

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.

A. Mulenga, M. Sugino, M. Nakajima, C. Sugimoto, and M. Onuma, Tick-Encoded serine proteinase inhibitors (serpins); potential target antigens for tick vaccine development, J Vet Med Sci, vol.63, pp.1063-1072, 2001.

S. Imamura, I. Da-silva-vaz-junior, M. Sugino, K. Ohashi, and M. Onuma, A serine protease inhibitor (serpin) from Haemaphysalis longicornis as an anti-tick vaccine, Vaccine, vol.23, pp.1301-1312, 2005.

S. Imamura, B. Namangala, T. Tajima, M. E. Tembo, J. Yasuda et al., Two serine protease inhibitors (serpins) that induce a bovine protective immune response against Rhipicephalus appendiculatus ticks, Vaccine, vol.24, pp.2230-2237, 2006.

S. Imamura, S. Konnai, S. Ida, S. Yamada, C. Nakajima et al., Effects of anti-tick cocktail vaccine against Rhipicephalus appendiculatus, Jpn J Vet Res, vol.56, pp.85-98, 2008.

U. Pal, X. Li, T. Wang, R. R. Montgomery, N. Ramamoorthi et al., TROSPA, an Ixodes scapularis receptor for Borrelia burgdorferi, Cell, vol.119, pp.457-68, 2004.

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.482-92, 2009.

S. Narasimhan, B. Sukumaran, U. Bozdogan, V. Thomas, X. Liang et al., A tick antioxidant facilitates the Lyme disease agent's successful migration from the mammalian host to the arthropod vector, Cell Host Microbe, vol.2, pp.7-18, 2007.

J. Dai, S. Narasimhan, L. Zhang, L. Liu, P. Wang et al., Tick histamine release factor is critical for Ixodes scapularis engorgement and transmission of the lyme disease agent, PLoS Pathog, vol.6, p.1001205, 2010.

T. J. Schuijt, J. Coumou, S. Narasimhan, J. Dai, K. Deponte et al., A tick mannose-binding lectin inhibitor interferes with the vertebrate complement cascade to enhance transmission of the lyme disease agent, Cell Host Microbe, vol.10, pp.136-182, 2011.

T. J. Schuijt, S. Narasimhan, S. Daffre, K. Deponte, J. W. Hovius et al., Identification and characterization of Ixodes scapularis antigens that elicit tick immunity using yeast surface display, PLoS One, vol.6, p.15926, 2011.

S. I. Bonnet, F. Binetruy, A. M. Hernandez-jarguin, and O. Duron, The Tick Microbiome: Why Non-pathogenic Microorganisms Matter in Tick Biology and Pathogen Transmission, Front Cell Infect Microbiol, vol.7, p.236, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02005450

N. M. Abraham, L. Liu, B. L. Jutras, A. K. Yadav, S. Narasimhan et al., Pathogen-mediated manipulation of arthropod microbiota to promote infection, Proc Natl Acad Sci U S A, vol.114, pp.781-790, 2017.

S. Narasimhan and E. Fikrig, Tick microbiome: the force within, Trends Parasitol, vol.31, pp.315-338, 2015.

A. A. Blisnick, T. Foulon, and S. I. Bonnet, Serine Protease Inhibitors in Ticks: An Overview of Their Role in Tick Biology and Tick-Borne Pathogen Transmission, Front Cell Infect Microbiol, vol.7, p.199, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01536659

L. Talleklint and T. G. Jaenson, Increasing geographical distribution and density of Ixodes ricinus (Acari: Ixodidae) in central and northern Sweden, J Med Entomol, vol.35, pp.521-527, 1998.

J. M. Medlock, K. M. Hansford, A. Bormane, M. Derdakova, A. Estrada-pena et al., Driving forces for changes in geographical distribution of Ixodes ricinus ticks in Europe, Parasit Vectors, vol.6, p.1, 2013.

E. Leger, G. Vourc'h, L. Vial, C. Chevillon, and K. D. Mccoy, Changing distributions of ticks: causes and consequences, Exp Appl Acarol, vol.59, pp.219-263, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00965558

A. Estrada-pena, J. M. Venzal, and C. Sanchez-acedo, The tick Ixodes ricinus: distribution and climate preferences in the western Palaearctic, Med Vet Entomol, vol.20, pp.189-97, 2006.

R. Lauterbach, K. Wells, R. B. O'hara, E. K. Kalko, and S. C. Renner, Variable strength of forest stand attributes and weather conditions on the questing activity of Ixodes ricinus ticks over years in managed forests, PLoS One, vol.8, p.55365, 2013.

M. Daniel, M. Maly, V. Danielova, B. Kriz, and P. Nuttall, Abiotic predictors and annual seasonal dynamics of Ixodes ricinus, the major disease vector of Central Europe, Parasit Vectors, vol.8, p.478, 2015.

R. D. Mitchell, J. Zhu, A. L. Carr, A. Dhammi, G. Cave et al., Infrared light detection by the haller's organ of adult american dog ticks, Dermacentor variabilis (Ixodida: Ixodidae), Ticks Tick Borne Dis, vol.8, pp.764-771, 2017.

D. E. Sonenshine and R. M. Roe, Biology of ticks, vol.1, pp.74-96, 2014.

C. Pérez-eid, Les tiques: identification, Biologie, Imortance Médicale et Vétérinaire, 2007.

N. Inoue, K. Hanada, N. Tsuji, I. Igarashi, H. Nagasawa et al., Characterization of phagocytic hemocytes in Ornithodoros moubata (Acari: Ixodidae), J Med Entomol, vol.38, pp.514-523, 2001.

D. Taylor, Innate Immunity in Ticks: A review, vol.15, pp.109-127, 2006.

L. Simo, D. E. Sonenshine, Y. Park, and D. ?it?an, The nervous and sensory systems: structure, function, proteomics and genomics, Biology of Ticks Volume, vol.1, p.560, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01601517

L. Simo, D. Zitnan, and Y. Park, Neural control of salivary glands in ixodid ticks, J Insect Physiol, vol.58, pp.459-66, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01601792

W. J. L'amoreaux, L. Junaid, and S. Trevidi, Morphological evidence that salivary gland degeneration in the American dog tick, Dermacentor variabilis (Say), involves programmed cell death, Tissue Cell, vol.35, pp.95-104, 2003.

L. Simo, M. Kazimirova, J. Richardson, and S. I. Bonnet, The Essential Role of Tick Salivary Glands and Saliva in Tick Feeding and Pathogen Transmission, Front Cell Infect Microbiol, vol.7, p.281, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01608039

G. J. Bonnet, S. Boulanger, and N. , L'interaction tique-hôte et la transmission des pathogènes, Tiques et maladies à tiques, I. editions, p.336, 2016.

D. Sojka, Z. Franta, M. Horn, C. R. Caffrey, M. Mares et al., New insights into the machinery of blood digestion by ticks, Trends Parasitol, vol.29, pp.276-85, 2013.

D. E. Sonenshine, Tick pheromones and their use in tick control, Annu Rev Entomol, vol.51, pp.557-80, 2006.

J. L. Olivier, relationships among feeding, gametogenesis, mating, and syngamiy in ticks, Host regulated developmental mechanisms in vector arthropds, pp.93-99, 1986.

A. D. Lees and J. W. Beament, An egg-waxing organ in ticks, Q J Microsc Sci, vol.89, pp.291-322, 1948.

J. Dmitry, A. Apanaskevich, and J. H. Oliver, Life Cycles and Natural History of Ticks, Biology of Ticks vol, vol.1, 2014.

A. R. Van-oosten, D. J. Heylen, and E. Matthysen, Host specificity of a bird-specialised endophilic ectoparasite, the tree-hole tick Ixodes arboricola, Parasitol Res, vol.113, pp.4397-405, 2014.

A. C. Steere, F. Strle, G. P. Wormser, L. T. Hu, J. A. Branda et al., Lyme borreliosis, Nat Rev Dis Primers, vol.2, p.16090, 2016.

I. Krupka and R. K. Straubinger, Lyme borreliosis in dogs and cats: background, diagnosis, treatment and prevention of infections with Borrelia burgdorferi sensu stricto, Vet Clin North Am Small Anim Pract, vol.40, pp.1103-1122, 2010.

N. M. Allen and H. Jungbluth, Lyme Neuroborreliosis: A Potentially Preventable Cause of Stroke, J Pediatr, vol.170, pp.334-335, 2016.

E. B. Breitschwerdt, Bartonellosis: one health perspectives for an emerging infectious disease, ILAR J, vol.55, pp.46-58, 2014.

U. Okaro, A. Addisu, B. Casanas, and B. Anderson, Bartonella Species, an Emerging Cause of Blood-Culture-Negative Endocarditis, Clin Microbiol Rev, vol.30, pp.709-746, 2017.

S. S. Breitschwerdt, E. B. Hopkins, and S. , Neurological Manifestations of Bartonellosis in Immunocompetent Patients: A Composite of Reports from, 2005.

Z. Woldehiwet, The natural history of Anaplasma phagocytophilum, Vet Parasitol, vol.167, pp.108-130, 2010.

J. S. Dumler, K. S. Choi, J. C. Garcia-garcia, N. S. Barat, D. G. Scorpio et al., Human granulocytic anaplasmosis and Anaplasma phagocytophilum, Emerg Infect Dis, vol.11, pp.1828-1862, 2005.

J. S. Bakken and J. S. Dumler, Human granulocytic anaplasmosis, Infect Dis Clin North Am, vol.29, pp.341-55, 2015.

J. Celli and T. C. Zahrt, Mechanisms of Francisella tularensis intracellular pathogenesis, Cold Spring Harb Perspect Med, vol.3, p.10314, 2013.

S. I. Bonnet, R. E. Paul, E. Bischoff, M. Cote, and E. L. Naour, First identification of Rickettsia helvetica in questing ticks from a French Northern Brittany Forest, PLoS Negl Trop Dis, vol.11, p.5416, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01570199

K. Nilsson, O. Lindquist, and C. Pahlson, Association of Rickettsia helvetica with chronic perimyocarditis in sudden cardiac death, Lancet, vol.354, pp.1169-73, 1999.

E. Ghigo, L. Pretat, B. Desnues, C. Capo, D. Raoult et al., Intracellular life of Coxiella burnetii in macrophages, Ann N Y Acad Sci, vol.1166, pp.55-66, 2009.

D. Raoult, T. Marrie, and J. Mege, Natural history and pathophysiology of Q fever, Lancet Infect Dis, vol.5, pp.219-245, 2005.

A. C. Salamand, F. Collart, T. Caus, J. P. Casalta, A. Mouly-bandini et al., Q fever endocarditis: over 14 years of surgical experience in a referral center for rickettsioses, J Heart Valve Dis, vol.11, pp.84-90, 2002.

A. Chauvin, E. Moreau, S. Bonnet, O. Plantard, and L. Malandrin, Babesia and its hosts: adaptation to long-lasting interactions as a way to achieve efficient transmission, Vet Res, vol.40, p.37, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00903088

J. J. Yusuf, Review on Bovine Babesiosis and its Economical Importance, Journal of Veterinary Medicine and Research, 2017.

M. K. Gorenflot, A. Precigout, E. Carcy, B. Schetters, and T. P. , Human babesiosis, Annals of Tropical Medicine & Parasitology, vol.92, pp.489-501, 1998.

T. S. Gritsun, V. A. Lashkevich, and E. A. Gould, Tick-borne encephalitis, Antiviral Res, vol.57, pp.129-175, 2003.

H. Reid, Louping-ill, In Practice, vol.13, pp.157-160, 1991.

C. L. Jeffries, K. L. Mansfield, L. P. Phipps, P. R. Wakeley, R. Mearns et al., Louping ill virus: an endemic tick-borne disease of Great Britain, J Gen Virol, vol.95, pp.1005-1019, 2014.

J. Suppan, B. Engel, M. Marchetti-deschmann, and S. Nurnberger, Tick attachment cementreviewing the mysteries of a biological skin plug system, Biol Rev Camb Philos Soc, vol.93, pp.1056-1076, 2018.

M. M. Lavoipierre, Feeding mechanism of blood-sucking arthropods, Nature, vol.208, pp.302-305, 1965.

Z. Franta, H. Frantova, J. Konvickova, M. Horn, D. Sojka et al., Dynamics of digestive proteolytic system during blood feeding of the hard tick Ixodes ricinus, vol.3, p.119, 2010.

J. W. Jelinski, Painless Hematophagy: The Functional Role of Novel Tick Metalloproteases in Pain Suppression, 2016.

J. M. Ribeiro and T. N. Mather, Ixodes scapularis: salivary kininase activity is a metallo dipeptidyl carboxypeptidase, Exp Parasitol, vol.89, pp.213-234, 1998.

S. Sangamnatdej, G. C. Paesen, M. Slovak, and P. A. Nuttall, A high affinity serotonin-and histamine-binding lipocalin from tick saliva, Insect Mol Biol, vol.11, pp.79-86, 2002.

G. C. Paesen, P. L. Adams, K. Harlos, P. A. Nuttall, and D. I. Stuart, Tick histamine-binding proteins: isolation, cloning, and three-dimensional structure, Mol Cell, vol.3, pp.661-71, 1999.

R. F. De-jongh, K. C. Vissers, T. F. Meert, L. H. Booij, C. S. De-deyne et al., The role of interleukin-6 in nociception and pain, Anesth Analg, vol.96, pp.1096-103, 2003.

J. Li, Y. P. Zhang, and R. S. Kirsner, Angiogenesis in wound repair: angiogenic growth factors and the extracellular matrix, Microsc Res Tech, vol.60, pp.107-121, 2003.

M. S. Neal, Angiogenesis: is it the key to controlling the healing process?, J Wound Care, vol.10, pp.281-288, 2001.

I. M. Francischetti, T. N. Mather, and J. M. Ribeiro, Tick saliva is a potent inhibitor of endothelial cell proliferation and angiogenesis, Thromb Haemost, vol.94, pp.167-74, 2005.

M. Slovak, I. Stibraniova, V. Hajnicka, and P. A. Nuttall, Antiplatelet-derived growth factor (PDGF) activity in the saliva of ixodid ticks is linked with their long mouthparts, Parasite Immunol, vol.36, pp.32-42, 2014.

V. Hajnicka, I. Vancova-stibraniova, M. Slovak, P. Kocakova, and P. A. Nuttall, Ixodid tick salivary gland products target host wound healing growth factors, Int J Parasitol, vol.41, pp.213-236, 2011.

C. Kramer, Z. Nahmias, D. D. Norman, T. A. Mulvihill, L. B. Coons et al., Dermacentor variabilis: regulation of fibroblast migration by tick salivary gland extract and saliva, Exp Parasitol, vol.119, pp.391-398, 2008.

M. K. Islam, N. Tsuji, T. Miyoshi, M. A. Alim, X. Huang et al., The Kunitz-like modulatory protein haemangin is vital for hard tick blood-feeding success, PLoS Pathog, vol.5, p.1000497, 2009.

S. Fukumoto, T. Sakaguchi, M. You, X. Xuan, and K. Fujisaki, Tick troponin I-like molecule is a potent inhibitor for angiogenesis, Microvasc Res, vol.71, pp.218-239, 2006.

Y. Decrem, J. Beaufays, V. Blasioli, K. Lahaye, M. Brossard et al., A family of putative metalloproteases in the salivary glands of the tick Ixodes ricinus, FEBS J, vol.275, pp.1485-99, 2008.

S. Palta, R. Saroa, and A. Palta, Overview of the coagulation system, Indian J Anaesth, vol.58, pp.515-538, 2014.

I. M. Francischetti, T. N. Mather, and J. M. Ribeiro, Penthalaris, a novel recombinant five-Kunitz tissue factor pathway inhibitor (TFPI) from the salivary gland of the tick vector of Lyme disease, Ixodes scapularis, Thromb Haemost, vol.91, pp.886-98, 2004.

I. M. Francischetti, J. G. Valenzuela, J. F. Andersen, T. N. Mather, and J. M. Ribeiro, Ixolaris, a novel recombinant tissue factor pathway inhibitor (TFPI) from the salivary gland of the tick, Ixodes scapularis: identification of factor X and factor Xa as scaffolds for the inhibition of factor VIIa/tissue factor complex, Blood, vol.99, pp.3602-3614, 2002.

Y. Decrem, G. Rath, V. Blasioli, P. Cauchie, S. Robert et al., Ir-CPI, a coagulation contact phase inhibitor from the tick Ixodes ricinus, inhibits thrombus formation without impairing hemostasis, J Exp Med, vol.206, pp.2381-95, 2009.

P. P. Prevot, B. Adam, K. Z. Boudjeltia, M. Brossard, L. Lins et al., Antihemostatic effects of a serpin from the saliva of the tick Ixodes ricinus, J Biol Chem, vol.281, pp.26361-26370, 2006.

N. Kato, S. Iwanaga, T. Okayama, H. Isawa, M. Yuda et al., Identification and characterization of the plasma kallikrein-kinin system inhibitor, haemaphysalin, from hard tick, Haemaphysalis longicornis, Thromb Haemost, vol.93, pp.359-67, 2005.

T. J. Schuijt, K. Bakhtiari, S. Daffre, K. Deponte, S. J. Wielders et al., Factor Xa activation of factor V is of paramount importance in initiating the coagulation system: lessons from a tick salivary protein, Circulation, vol.128, pp.254-66, 2013.

M. K. Anisuzzaman, M. A. Islam, T. Alim, T. Miyoshi, K. Hatta et al., Longistatin, a plasminogen activator, is key to the availability of blood-meals for ixodid ticks, PLoS Pathog, vol.7, p.1001312, 2011.

I. M. Francischetti, T. N. Mather, and J. M. Ribeiro, Cloning of a salivary gland metalloprotease and characterization of gelatinase and fibrin(ogen)lytic activities in the saliva of the Lyme disease tick vector Ixodes scapularis, Biochem Biophys Res Commun, vol.305, pp.869-75, 2003.

J. Kotal, H. Langhansova, J. Lieskovska, J. F. Andersen, I. M. Francischetti et al., Modulation of host immunity by tick saliva, J Proteomics, vol.128, pp.58-68, 2015.

M. Kazimirova and I. Stibraniova, Tick salivary compounds: their role in modulation of host defences and pathogen transmission, Front Cell Infect Microbiol, vol.3, p.43, 2013.

S. S. Maxwell, T. A. Stoklasek, Y. Dash, K. R. Macaluso, and S. K. Wikel, Tick modulation of the in-vitro expression of adhesion molecules by skin-derived endothelial cells, Ann Trop Med Parasitol, vol.99, pp.661-72, 2005.

I. Vancova, V. Hajnicka, M. Slovak, and P. A. Nuttall, Anti-chemokine activities of ixodid ticks depend on tick species, developmental stage, and duration of feeding, Vet Parasitol, vol.167, pp.274-282, 2010.

I. Vancova, V. Hajnicka, M. Slovak, P. Kocakova, G. C. Paesen et al., Evasin-3-like anti-chemokine activity in salivary gland extracts of ixodid ticks during blood-feeding: a new target for tick control, Parasite Immunol, vol.32, pp.460-463, 2010.

J. Beaufays, B. Adam, C. Menten-dedoyart, L. Fievez, A. Grosjean et al., Ir-LBP, an ixodes ricinus tick salivary LTB4-binding lipocalin, interferes with host neutrophil function, PLoS One, vol.3, p.3987, 2008.

N. B. Wasala and D. C. Jaworski, Dermacentor variabilis: characterization and modeling of macrophage migration inhibitory factor with phylogenetic comparisons to other ticks, insects and parasitic nematodes, Exp Parasitol, vol.130, pp.232-240, 2012.

A. Hidano, S. Konnai, S. Yamada, N. Githaka, M. Isezaki et al., Suppressive effects of neutrophil by Salp16-like salivary gland proteins from Ixodes persulcatus Schulze tick, Insect Mol Biol, vol.23, pp.466-74, 2014.

A. Frauenschuh, C. A. Power, M. Deruaz, B. R. Ferreira, J. S. Silva et al., Molecular cloning and characterization of a highly selective chemokine-binding protein from the tick Rhipicephalus sanguineus, J Biol Chem, vol.282, pp.27250-27258, 2007.

M. Deruaz, A. Frauenschuh, A. L. Alessandri, J. M. Dias, F. M. Coelho et al., Ticks produce highly selective chemokine binding proteins with antiinflammatory activity, J Exp Med, vol.205, pp.2019-2050, 2008.

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.818-844, 1992.

Y. Tian, W. Chen, G. Mo, R. Chen, M. Fang et al., An Immunosuppressant Peptide from the Hard Tick Amblyomma variegatum, Toxins (Basel), vol.8, 2016.

T. Kishimoto, Interleukin-6: discovery of a pleiotropic cytokine, Arthritis Res Ther, vol.8, issue.2, 2006.

J. M. Ribeiro, J. J. Weis, and S. R. Telford, Saliva of the tick Ixodes dammini inhibits neutrophil function, Exp Parasitol, vol.70, pp.382-390, 1990.

C. D. Kramer, N. M. Poole, L. B. Coons, and J. A. Cole, Tick saliva regulates migration, phagocytosis, and gene expression in the macrophage-like cell line, IC-21, Exp Parasitol, vol.127, pp.665-71, 2011.

C. Turni, R. P. Lee, and L. A. Jackson, Effect of salivary gland extracts from the tick, Boophilus microplus, on leucocytes from Brahman and Hereford cattle, Parasite Immunol, vol.24, pp.355-61, 2002.

X. Guo, C. J. Booth, M. A. Paley, X. Wang, K. Deponte et al., Inhibition of neutrophil function by two tick salivary proteins, Infect Immun, vol.77, pp.2320-2329, 2009.

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.169-74, 1998.

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.1077-85, 1994.

B. R. Ferreira and J. S. Silva, Saliva of Rhipicephalus sanguineus tick impairs T cell proliferation and IFN-gamma-induced macrophage microbicidal activity, Vet Immunol Immunopathol, vol.64, pp.279-93, 1998.

M. Kubes, N. Fuchsberger, M. Labuda, E. Zuffova, and P. A. Nuttall, Salivary gland extracts of partially fed Dermacentor reticulatus ticks decrease natural killer cell activity in vitro, Immunology, vol.82, pp.113-119, 1994.

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.23-31, 2002.

C. Menten-dedoyart, C. Faccinetto, M. Golovchenko, I. Dupiereux, P. B. Van-lerberghe et al., Neutrophil extracellular traps entrap and kill Borrelia burgdorferi sensu stricto spirochetes and are not affected by Ixodes ricinus tick saliva, J Immunol, vol.189, pp.5393-401, 2012.

J. Chmelar, C. J. Oliveira, P. Rezacova, I. M. Francischetti, Z. Kovarova et al., A tick salivary protein targets cathepsin G and chymase and inhibits host inflammation and platelet aggregation, Blood, vol.117, pp.736-780, 2011.

G. C. Paesen, C. Siebold, K. Harlos, M. F. Peacey, P. A. Nuttall et al., A tick protein with a modified Kunitz fold inhibits human tryptase, J Mol Biol, vol.368, pp.1172-86, 2007.

D. D. Metcalfe, D. Baram, and Y. A. Mekori, Mast cells, Physiol Rev, vol.77, pp.1033-79, 1997.
URL : https://hal.archives-ouvertes.fr/hal-00282529

V. Hajnicka, I. Vancova, P. Kocakova, M. Slovak, J. Gasperik et al., Manipulation of host cytokine network by ticks: a potential gateway for pathogen transmission, Parasitology, vol.130, pp.333-375, 2005.

I. Vancova, M. Slovak, V. Hajnicka, M. Labuda, L. Simo et al., Differential antichemokine activity of Amblyomma variegatum adult ticks during blood-feeding, Parasite Immunol, vol.29, pp.169-77, 2007.
URL : https://hal.archives-ouvertes.fr/hal-01600968

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.18717-18740, 2000.

H. Schroeder, V. Daix, L. Gillet, J. C. Renauld, and A. Vanderplasschen, The paralogous salivary anti-complement proteins IRAC I and IRAC II encoded by Ixodes ricinus ticks have broad and complementary inhibitory activities against the complement of different host species, Microbes Infect, vol.9, pp.247-50, 2007.

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.469-79, 2007.

B. J. Mans and J. M. Ribeiro, Function, mechanism and evolution of the moubatin-clade of soft tick lipocalins, Insect Biochem Mol Biol, vol.38, pp.841-52, 2008.

M. A. Nunn, A. Sharma, G. C. Paesen, S. Adamson, O. Lissina et al., Complement inhibitor of C5 activation from the soft tick Ornithodoros moubata, J Immunol, vol.174, pp.2084-91, 2005.

R. F. Guo and P. A. Ward, Role of C5a in inflammatory responses, Annu Rev Immunol, vol.23, pp.821-52, 2005.

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.155-66, 2007.

T. M. Carvalho-costa, M. T. Mendes, M. V. Silva, T. A. Da-costa, M. G. Tiburcio et al., Immunosuppressive effects of Amblyomma cajennense tick saliva on murine bone marrow-derived dendritic cells, Parasit Vectors, vol.8, p.22, 2015.

C. J. Oliveira, K. A. Cavassani, D. D. More, G. P. Garlet, J. C. Aliberti et al., Tick saliva inhibits the chemotactic function of MIP-1alpha and selectively impairs chemotaxis of immature dendritic cells by down-regulating cell-surface CCR5, Int J Parasitol, vol.38, pp.705-721, 2008.

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.1497-505, 2007.

M. Slamova, A. Skallova, J. Palenikova, and J. Kopecky, Effect of tick saliva on immune interactions between Borrelia afzelii and murine dendritic cells, Parasite Immunol, vol.33, pp.654-60, 2011.

K. R. Macaluso and S. K. Wikel, Dermacentor andersoni: effects of repeated infestations on lymphocyte proliferation, cytokine production, and adhesion-molecule expression by BALB/c mice, Ann Trop Med Parasitol, vol.95, pp.413-440, 2001.

S. Hannier, J. Liversidge, J. M. Sternberg, and A. S. Bowman, Ixodes ricinus tick salivary gland extract inhibits IL-10 secretion and CD69 expression by mitogen-stimulated murine splenocytes and induces hyporesponsiveness in B lymphocytes, Parasite Immunol, vol.25, pp.27-37, 2003.

J. Anguita, N. Ramamoorthi, J. W. Hovius, S. Das, V. Thomas et al., Salp15, an ixodes scapularis salivary protein, inhibits CD4(+) T cell activation, Immunity, vol.16, pp.849-59, 2002.

N. Mejri, N. Franscini, B. Rutti, and M. Brossard, Th2 polarization of the immune response of BALB/c mice to Ixodes ricinus instars, importance of several antigens in activation of specific Th2 subpopulations, Parasite Immunol, vol.23, pp.61-70, 2001.

T. Rolnikova, M. Kazimirova, and M. Buc, Modulation of human lymphocyte proliferation by salivary gland extracts of ixodid ticks (Acari: Ixodidae): effect of feeding stage and sex, Folia Parasitol (Praha), vol.50, pp.305-317, 2003.

R. D. Gillespie, M. C. Dolan, J. Piesman, and R. G. Titus, Identification of an IL-2 binding protein in the saliva of the Lyme disease vector tick, Ixodes scapularis, J Immunol, vol.166, pp.4319-4345, 2001.

H. Inokuma, T. Aita, K. Ohno, and T. Onishi, Effects of infestation by Rhipicephalus sanguineus on lymphocyte blastogenic responses to mitogens in dogs, J Vet Med Sci, vol.60, pp.1013-1019, 1998.

F. Ganapamo, B. Rutti, and M. Brossard, In vitro production of interleukin-4 and interferongamma by lymph node cells from BALB/c mice infested with nymphal Ixodes ricinus ticks, Immunology, vol.85, pp.120-124, 1995.

S. K. Wikel, Influence of Dermacentor andersoni infestation on lymphocyte responsiveness to mitogens, Ann Trop Med Parasitol, vol.76, pp.627-659, 1982.

L. Kovar, J. Kopecky, and B. Rihova, Salivary gland extract from Ixodes ricinus tick polarizes the cytokine profile toward Th2 and suppresses proliferation of T lymphocytes in human PBMC culture, J Parasitol, vol.87, pp.1342-1350, 2001.

L. Kovar, J. Kopecky, and B. Rihova, Salivary gland extract from Ixodes ricinus tick modulates the host immune response towards the Th2 cytokine profile, Parasitol Res, vol.88, pp.1066-72, 2002.

J. Wu, Y. Wang, H. Liu, H. Yang, D. Ma et al., Two immunoregulatory peptides with antioxidant activity from tick salivary glands, J Biol Chem, vol.285, pp.16606-16619, 2010.

P. Gwakisa, K. Yoshihara, T. L. To, H. Gotoh, F. 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.

D. M. Heinze, S. K. Wikel, S. Thangamani, and F. J. Alarcon-chaidez, Transcriptional profiling of the murine cutaneous response during initial and subsequent infestations with Ixodes scapularis nymphs, Parasit Vectors, vol.5, p.26, 2012.

V. Rodrigues, B. Fernandez, A. Vercoutere, L. Chamayou, A. Andersen et al., Immunomodulatory Effects of Amblyomma variegatum Saliva on Bovine Cells: Characterization of Cellular Responses and Identification of Molecular Determinants, Front Cell Infect Microbiol, vol.7, p.521, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02073207

D. K. Boppana, G. Raj, L. John, S. K. Wikel, B. R. Latha et al., In vivo immunomodulatory effects of ixodid ticks on ovine circulating T-and B-lymphocytes, Parasite Immunol, vol.26, pp.83-93, 2004.

J. M. Ribeiro, G. T. Makoul, J. Levine, D. R. Robinson, and A. Spielman, Antihemostatic, antiinflammatory, and immunosuppressive properties of the saliva of a tick, Ixodes dammini, J Exp Med, vol.161, pp.332-376, 1985.

V. D. Boppana, S. Thangamani, F. J. Alarcon-chaidez, A. J. Adler, and S. K. Wikel, Blood feeding by the Rocky Mountain spotted fever vector, Dermacentor andersoni, induces interleukin-4 expression by cognate antigen responding CD4+ T cells, Parasit Vectors, vol.2, p.47, 2009.

K. M. Bonnet, S. Richardson, J. ?imo, and L. , Tick Saliva and Its Role in Pathogen Transmission, pp.121-192, 2017.

P. A. Nuttall and M. Labuda, Tick-host interactions: saliva-activated transmission, Parasitology, vol.129, pp.177-89, 2004.

L. D. Jones, E. Hodgson, and P. A. Nuttall, Enhancement of virus transmission by tick salivary glands, J Gen Virol, vol.70, issue.7, pp.1895-1903, 1989.

S. Nazario, S. Das, A. M. Silva, K. Deponte, N. Marcantonio et al., Prevention of Borrelia burgdorferi transmission in guinea pigs by tick immunity, Am J Trop Med Hyg, vol.58, pp.780-785, 1998.

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.335-343, 1997.

B. Sukumaran, S. Narasimhan, J. F. Anderson, K. Deponte, N. Marcantonio et al., An Ixodes scapularis protein required for survival of Anaplasma phagocytophilum in tick salivary glands, J Exp Med, vol.203, pp.1507-1524, 2006.

J. Pechova, G. Stepanova, L. Kovar, and J. Kopecky, Tick salivary gland extract-activated transmission of Borrelia afzelii spirochaetes, Folia Parasitol (Praha), vol.49, pp.153-162, 2002.

Z. Krocova, A. Macela, L. Hernychova, M. Kroca, J. Pechova et al., Tick salivary gland extract accelerates proliferation of Francisella tularensis in the host, J Parasitol, vol.89, pp.14-20, 2003.

M. Labuda, L. D. Jones, T. Williams, and P. A. Nuttall, Enhancement of tick-borne encephalitis virus transmission by tick salivary gland extracts, Med Vet Entomol, vol.7, pp.193-199, 1993.

L. M. Porter, Z. M. Radulovic, and A. Mulenga, A repertoire of protease inhibitor families in Amblyomma americanum and other tick species: inter-species comparative analyses, Parasit Vectors, vol.10, p.152, 2017.

L. F. Parizi, A. Ali, L. Tirloni, D. P. Oldiges, G. A. Sabadin et al., Peptidase inhibitors in tick physiology, Med Vet Entomol, vol.32, pp.129-144, 2018.

X. Y. Liu, J. De-la-fuente, M. Cote, R. C. Galindo, S. Moutailler et al., IrSPI, a tick serine protease inhibitor involved in tick feeding and Bartonella henselae infection, PLoS Negl Trop Dis, vol.8, p.2993, 2014.

X. Y. Liu, M. Cote, R. E. Paul, and S. I. Bonnet, Impact of feeding system and infection status of the blood meal on Ixodes ricinus feeding, Ticks Tick Borne Dis, vol.5, pp.323-331, 2014.

S. Bonnet and X. Y. Liu, Laboratory artificial infection of hard ticks: a tool for the analysis of tick-borne pathogen transmission, Acarologia, vol.52, pp.453-464, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01301271

C. B. Burge and S. Karlin, Finding the genes in genomic DNA, Curr Opin Struct Biol, vol.8, pp.346-54, 1998.

C. B. Burge, Modeling dependencies in pre-mRNA splicing signals, Computational Methods in Molecular Biology, S. Salzberg, pp.127-163, 1998.

R. E. Paul, M. Cote, E. L. Naour, and S. I. Bonnet, Environmental factors influencing tick densities over seven years in a French suburban forest, Parasit Vectors, vol.9, p.309, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01370808

B. S. Almazán, C. Cote, M. Slovák, M. Park, Y. ?imo et al., A Versatile Model of Hard Tick Infestation on Laboratory Rabbits, Jove video journal, 2018.

M. Gulia-nuss, A. B. Nuss, J. M. Meyer, D. E. Sonenshine, R. M. Roe et al., Genomic insights into the Ixodes scapularis tick vector of Lyme disease, Nature Communications, vol.7, p.10507, 2016.

A. Schwarz, B. M. Von-reumont, J. Erhart, A. C. Chagas, J. M. Ribeiro et al., De novo Ixodes ricinus salivary gland transcriptome analysis using two next-generation sequencing methodologies, FASEB J, vol.27, pp.4745-56, 2013.

M. Kotsyfakis, A. Schwarz, J. Erhart, and J. M. Ribeiro, Tissue-and time-dependent transcription in Ixodes ricinus salivary glands and midguts when blood feeding on the vertebrate host, Sci Rep, vol.5, p.9103, 2015.

J. G. Valenzuela, I. M. Francischetti, V. M. Pham, M. K. Garfield, T. N. Mather et al., Exploring the sialome of the tick Ixodes scapularis, J Exp Biol, vol.205, pp.2843-64, 2002.

J. M. Ribeiro, F. Alarcon-chaidez, I. M. Francischetti, B. J. Mans, T. N. Mather et al., An annotated catalog of salivary gland transcripts from Ixodes scapularis ticks, Insect Biochem Mol Biol, vol.36, pp.111-140, 2006.

S. X. Dai, A. D. Zhang, and J. F. Huang, Evolution, expansion and expression of the Kunitz/BPTI gene family associated with long-term blood feeding in Ixodes Scapularis, BMC Evol Biol, vol.12, p.4, 2012.

G. C. Paesen, C. Siebold, M. L. Dallas, C. Peers, K. Harlos et al., An ion-channel modulator from the saliva of the brown ear tick has a highly modified Kunitz/BPTI structure, J Mol Biol, vol.389, pp.734-781, 2009.

A. Micsonai, F. Wien, L. Kernya, Y. H. Lee, Y. Goto et al., Accurate secondary structure prediction and fold recognition for circular dichroism spectroscopy, Proc Natl Acad Sci U S A, vol.112, pp.3095-103, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01485547

J. Koci, L. Simo, and Y. Park, Validation of internal reference genes for real-time quantitative polymerase chain reaction studies in the tick, Ixodes scapularis (Acari: Ixodidae), J Med Entomol, vol.50, pp.79-84, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01601685

J. M. Ribeiro, F. Alarcon-chaidez, B. Mans, I. M. Francischetti, T. Mather et al.,

T. K. Kim, L. Tirloni, A. F. Pinto, J. Moresco, J. R. Yates et al., Ixodes scapularis Tick Saliva Proteins Sequentially Secreted Every 24 h during Blood Feeding, PLoS Negl Trop Dis, vol.10, p.4323, 2016.

T. Katoh and M. Tiemeyer, The N's and O's of Drosophila glycoprotein glycobiology, Glycoconj J, vol.30, pp.57-66, 2013.

D. J. Harvey, D. R. Wing, B. Kuster, and I. B. Wilson, Composition of N-linked carbohydrates from ovalbumin and co-purified glycoproteins, J Am Soc Mass Spectrom, vol.11, pp.564-71, 2000.

B. Raynal, P. Lenormand, B. Baron, S. Hoos, and P. England, Quality assessment and optimization of purified protein samples: why and how?, Microb Cell Fact, vol.13, p.180, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01108005

S. Ranasinghe and D. P. Mcmanus, Structure and function of invertebrate Kunitz serine protease inhibitors, Dev Comp Immunol, vol.39, pp.219-246, 2013.

M. L. Circu and T. Y. Aw, Glutathione and apoptosis, Free radical research, vol.42, pp.689-706, 2008.

O. Zitka, S. Skalickova, J. Gumulec, M. Masarik, V. Adam et al., Redox status expressed as GSH:GSSG ratio as a marker for oxidative stress in paediatric tumour patients, Oncology letters, vol.4, pp.1247-1253, 2012.

J. Mai, A. Virtue, J. Shen, H. Wang, and X. Yang, An evolving new paradigm: endothelial cells -conditional innate immune cells, Journal of Hematology & Oncology, vol.6, p.61, 2013.

G. Caignard, Cartographie des intéractions entre protéines virales codées par le gène P des Paramyxoviridae et protéines cellulaires, 2010.

S. Fields and O. Song, A novel genetic system to detect protein-protein interactions, Nature, vol.340, pp.245-251, 1989.

P. O. Vidalain, Y. Jacob, M. C. Hagemeijer, L. M. Jones, G. Neveu et al., A field-proven yeast two-hybrid protocol used to identify coronavirus-host protein-protein interactions, Methods Mol Biol, vol.1282, pp.213-242, 2015.

F. Katzen, Gateway((R)) recombinational cloning: a biological operating system, Expert Opin Drug Discov, vol.2, pp.571-89, 2007.

M. Villar, N. Ayllon, P. Alberdi, A. Moreno, M. Moreno et al., Integrated Metabolomics, Transcriptomics and Proteomics Identifies Metabolic Pathways Affected by Anaplasma phagocytophilum Infection in Tick Cells, Mol Cell Proteomics, vol.14, pp.3154-72, 2015.

H. S. Lee, Y. Qi, and W. Im, Effects of N-glycosylation on protein conformation and dynamics: Protein Data Bank analysis and molecular dynamics simulation study, Sci Rep, vol.5, p.8926, 2015.

H. Nishi, K. Hashimoto, and A. R. Panchenko, Phosphorylation in protein-protein binding: effect on stability and function, Structure, vol.19, pp.1807-1822, 2011.

B. J. Arey, The Role of Glycosylation in Receptor Signaling, pp.273-286, 2012.

S. Goda, K. Takano, Y. Yamagata, Y. Katakura, and K. Yutani, Effect of extra N-terminal residues on the stability and folding of human lysozyme expressed in Pichia pastoris, Protein Eng, vol.13, pp.299-307, 2000.

J. Perner, S. Kropackova, P. Kopacek, and J. M. Ribeiro, Sialome diversity of ticks revealed by RNAseq of single tick salivary glands, PLoS Negl Trop Dis, vol.12, p.6410, 2018.

A. L. Cortajarena and L. Regan, Ligand binding by TPR domains, Protein Sci, vol.15, pp.1193-1201, 2006.

J. W. Chartron, D. G. Vandervelde, W. M. Clemons, and J. , Structures of the Sgt2/SGTA dimerization domain with the Get5/UBL4A UBL domain reveal an interaction that forms a conserved dynamic interface, Cell Rep, vol.2, pp.1620-1652, 2012.

L. J. Worrall, M. A. Wear, A. P. Page, and M. D. Walkinshaw, Cloning, purification and characterization of the Caenorhabditis elegans small glutamine-rich tetratricopeptide repeatcontaining protein, Biochim Biophys Acta, vol.1784, pp.496-503, 2008.

S. T. Liou and C. Wang, Small glutamine-rich tetratricopeptide repeat-containing protein is composed of three structural units with distinct functions, Arch Biochem Biophys, vol.435, pp.253-63, 2005.

M. Kaneko and Y. Nomura, ER signaling in unfolded protein response, Life Sci, vol.74, pp.199-205, 2003.

T. Spies, M. Bresnahan, and J. L. Strominger, Human major histocompatibility complex contains a minimum of 19 genes between the complement cluster and HLA-B, Proc Natl Acad Sci U S A, vol.86, pp.8955-8963, 1989.

J. Binici and J. Koch, BAG-6, a jack of all trades in health and disease, Cell Mol Life Sci, vol.71, pp.1829-1866, 2014.

H. Kawahara, R. Minami, and N. Yokota, BAG6/BAT3: emerging roles in quality control for nascent polypeptides, J Biochem, vol.153, pp.147-60, 2013.

J. G. Lee and Y. Ye, Bag6/Bat3/Scythe: a novel chaperone activity with diverse regulatory functions in protein biogenesis and degradation, Bioessays, vol.35, pp.377-85, 2013.

M. Rangachari, C. Zhu, K. Sakuishi, S. Xiao, J. Karman et al., Bat3 promotes T cell responses and autoimmunity by repressing Tim-3-mediated cell death and exhaustion, Nat Med, vol.18, pp.1394-400, 2012.

R. K. Pai, D. Askew, W. H. Boom, and C. V. Harding, Regulation of class II MHC expression in APCs: roles of types I, III, and IV class II transactivator, J Immunol, vol.169, pp.1326-1359, 2002.

V. R. Simhadri, K. S. Reiners, H. P. Hansen, D. Topolar, V. L. Simhadri et al., Dendritic cells release HLA-B-associated transcript-3 positive exosomes to regulate natural killer function, PLoS One, vol.3, p.3377, 2008.

E. Pogge-von-strandmann, V. R. Simhadri, B. Tresckow, S. Sasse, K. S. Reiners et al., Human leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on natural killer cells, Immunity, vol.27, pp.965-74, 2007.

K. L. Rock and A. L. Goldberg, Degradation of cell proteins and the generation of MHC class Ipresented peptides, Annu Rev Immunol, vol.17, pp.739-79, 1999.

Y. H. Wu, S. F. Shih, and J. Y. Lin, Ricin triggers apoptotic morphological changes through caspase-3 cleavage of BAT3, J Biol Chem, vol.279, pp.19264-75, 2004.

I. Marin, The ubiquilin gene family: evolutionary patterns and functional insights, BMC Evol Biol, vol.14, p.63, 2014.

D. Y. Lee, D. Arnott, and E. J. Brown, Ubiquilin4 is an adaptor protein that recruits Ubiquilin1 to the autophagy machinery, EMBO Rep, vol.14, pp.373-81, 2013.

E. N. N'diaye, J. Debnath, and E. J. Brown, Ubiquilins accelerate autophagosome maturation and promote cell survival during nutrient starvation, Autophagy, vol.5, pp.573-578, 2009.

C. Zhang and A. J. Saunders, An emerging role for Ubiquilin 1 in regulating protein quality control system and in disease pathogenesis, Discov Med, vol.8, pp.18-22, 2009.

S. Wu, A. Mikhailov, H. Kallo-hosein, K. Hara, K. Yonezawa et al., Characterization of ubiquilin 1, an mTOR-interacting protein, Biochim Biophys Acta, vol.1542, pp.41-56, 2002.

A. M. Whiteley, M. A. Prado, I. Peng, A. R. Abbas, B. Haley et al., Ubiquilin1 promotes antigen-receptor mediated proliferation by eliminating mislocalized mitochondrial proteins, Elife, vol.6, 2017.

J. Camps, M. R. Erdos, and T. Ried, The role of lamin B1 for the maintenance of nuclear structure and function, Nucleus, vol.6, pp.8-14, 2015.

T. Shimi, V. Butin-israeli, S. A. Adam, R. B. Hamanaka, A. E. Goldman et al., The role of nuclear lamin B1 in cell proliferation and senescence, Genes Dev, vol.25, pp.2579-93, 2011.

J. W. Shin, K. R. Spinler, J. Swift, J. A. Chasis, N. Mohandas et al., Lamins regulate cell trafficking and lineage maturation of adult human hematopoietic cells, Proc Natl Acad Sci U S A, vol.110, pp.18892-18899, 2013.

M. Christin-piché, Étude du rôle régulateur de la lamine B1 dans l'activation plaquettaire : base moléculaire de la thromboprotection chez les patients porteurs d'anticoagulant lupique et d'anti-lamine B1, 2008.

J. H. Peters, G. E. Chen, and R. O. Hynes, Fibronectin isoform distribution in the mouse. II. Differential distribution of the alternatively spliced EIIIB, EIIIA, and V segments in the adult mouse, Cell Adhes Commun, vol.4, pp.127-175, 1996.

Z. A. Khan, B. M. Chan, S. Uniyal, Y. P. Barbin, H. Farhangkhoee et al., EDB fibronectin and angiogenesis --a novel mechanistic pathway, Angiogenesis, vol.8, pp.183-96, 2005.

J. De-la-fuente, C. Maritz-olivier, V. Naranjo, P. Ayoubi, A. M. Nijhof et al., Evidence of the role of tick subolesin in gene expression, BMC Genomics, vol.9, p.372, 2008.

A. Rizzoli, C. Silaghi, A. Obiegala, I. Rudolf, Z. Hubalek et al., Ixodes ricinus and Its Transmitted Pathogens in Urban and Peri-Urban Areas in Europe: New Hazards and Relevance for Public Health, Front Public Health, vol.2, p.251, 2014.

J. S. Gray, H. Dautel, A. Estrada-pena, O. Kahl, and E. Lindgren, Effects of climate change on ticks and tick-borne diseases in europe, Interdiscip Perspect Infect Dis, p.593232, 2009.

E. Lindgren, L. Talleklint, and T. Polfeldt, Impact of climatic change on the northern latitude limit and population density of the disease-transmitting European tick Ixodes ricinus, Environ Health Perspect, vol.108, pp.119-142, 2000.

M. Vayssier-taussat, S. Moutailler, L. Michelet, E. Devillers, S. Bonnet et al., Next generation sequencing uncovers unexpected bacterial pathogens in ticks in western Europe, PLoS One, vol.8, p.81439, 2013.

S. Bonnet, L. Michelet, S. Moutailler, J. Cheval, C. Hebert et al., Identification of parasitic communities within European ticks using next-generation sequencing, PLoS Negl Trop Dis, vol.8, p.2753, 2014.

S. Moutailler, I. Popovici, E. Devillers, M. Vayssier-taussat, and M. Eloit, Diversity of viruses in Ixodes ricinus, and characterization of a neurotropic strain of Eyach virus, New Microbes New Infect, vol.11, pp.71-81, 2016.

T. Miyoshi, N. Tsuji, M. K. Islam, M. A. Alim, T. Hatta et al., A Kunitz-type proteinase inhibitor from the midgut of the ixodid tick, Haemaphysalis longicornis, and its endogenous target serine proteinase, Mol Biochem Parasitol, vol.170, pp.112-117, 2010.

S. Narasimhan, O. Perez, S. Mootien, K. Deponte, R. A. Koski et al., Characterization of Ixophilin, a thrombin inhibitor from the gut of Ixodes scapularis, PLoS One, vol.8, p.68012, 2013.

A. S. Tanaka, R. Andreotti, A. Gomes, R. J. Torquato, M. U. Sampaio et al., A double headed serine proteinase inhibitor--human plasma kallikrein and elastase inhibitor--from Boophilus microplus larvae, Immunopharmacology, vol.45, pp.171-178, 1999.

R. Andreotti, K. C. Malavazi-piza, S. D. Sasaki, R. J. Torquato, A. Gomes et al., Serine proteinase inhibitors from eggs and larvae of tick Boophilus microplus: purification and biochemical characterization, J Protein Chem, vol.20, pp.337-380, 2001.

S. M. Ceraul, S. M. Dreher-lesnick, A. Mulenga, M. S. Rahman, and A. F. Azad, Functional characterization and novel rickettsiostatic effects of a Kunitz-type serine protease inhibitor from the tick Dermacentor variabilis, Infect Immun, vol.76, pp.5429-5464, 2008.

C. A. Lima, R. J. Torquato, S. D. Sasaki, G. Z. Justo, and A. S. Tanaka, Biochemical characterization of a Kunitz type inhibitor similar to dendrotoxins produced by Rhipicephalus (Boophilus) microplus (Acari: Ixodidae) hemocytes, Vet Parasitol, vol.167, pp.279-87, 2010.

M. Kotsyfakis, A. Schwarz, J. Erhart, and J. M. Ribeiro, Tissue-and time-dependent transcription in Ixodes ricinus salivary glands and midguts when blood feeding on the vertebrate host, Scientific Reports, vol.5, p.9103, 2015.

G. Leboulle, M. Crippa, Y. Decrem, N. Mejri, M. Brossard et al., Characterization of a novel salivary immunosuppressive protein from Ixodes ricinus ticks, J Biol Chem, vol.277, pp.10083-10092, 2002.

J. J. Valdes, A. Schwarz, I. Cabeza-de-vaca, E. Calvo, J. H. Pedra et al., Tryptogalinin is a tick Kunitz serine protease inhibitor with a unique intrinsic disorder, PLoS One, vol.8, p.62562, 2013.

K. Toyomane, S. Konnai, A. Niwa, N. Githaka, M. Isezaki et al., Identification and the preliminary in vitro characterization of IRIS homologue from salivary glands of Ixodes persulcatus Schulze, Ticks Tick Borne Dis, vol.7, pp.119-125, 2016.

, Ce plasmide possède le gène de résistance à l'ampicilline (violet) et une origine de réplication, Figure, vol.39