, The relative expressions of the opn gene References

P. Mansueto, G. Vitale, D. Lorenzo, G. Rini, G. B. Mansueto et al., Immunopathology of leishmaniasis: an update, Int J Immunopathol Pharmacol, vol.20, issue.3, pp.435-480, 2007.

R. Lainson, L. Ryan, and J. J. Shaw, Infective stages of Leishmania in the sandfly vector and some observations on the mechanism of transmission, Memorias do Instituto Oswaldo Cruz, vol.82, issue.3, pp.421-425, 1987.

P. A. Bates, Transmission of Leishmania metacyclic promastigotes by phlebotomine sand flies, Int J Parasitol, vol.37, issue.10, pp.1097-106, 2007.

S. M. Gossage, M. E. Rogers, and P. A. Bates, Two separate growth phases during the development of Leishmania in sand flies: implications for understanding the life cycle, Int J Parasitol, vol.33, issue.10, pp.1027-1061, 2003.

M. E. Rogers, M. L. Chance, and P. A. Bates, The role of promastigote secretory gel in the origin and transmission of the infective stage of Leishmania mexicana by the sandfly Lutzomyia longipalpis, Parasitology, vol.124, pp.495-507, 2002.

T. Lestinova, I. Rohousova, M. Sima, C. I. Oliveira, and P. Volf, Insights into the sand fly saliva: blood-feeding and immune interactions between sand flies, hosts, and Leishmania, PLoS Negl Trop Dis, vol.11, issue.7, p.5600, 2017.

E. Handman and D. V. Bullen, Interaction of Leishmania with the host macrophage, Trends Parasitol, vol.18, issue.8, pp.332-336, 2002.

D. Liu and J. E. Uzonna, The early interaction of Leishmania with macrophages and dendritic cells and its influence on the host immune response, Front Cell Infect Microbiol, vol.2, p.83, 2012.

F. De-oliveira-cardoso, S. De-souza-cda, V. G. Mendes, A. L. Abreu-silva, G. Da-costa et al., Immunopathological studies of Leishmania amazonensis infection in resistant and in susceptible mice, J Infect Dis, vol.201, issue.12, pp.1933-1940, 2010.

D. Mcmahon-pratt and A. J. , Does the Leishmania major paradigm of pathogenesis and protection hold for New World cutaneous leishmaniases or the visceral disease?, Immunol Rev, vol.201, pp.206-230, 2004.

E. Giraud, H. Lecoeur, G. Soubigou, J. Y. Coppee, G. Milon et al., Distinct transcriptional signatures of bone marrow-derived C57BL/6 and DBA/2 dendritic leucocytes hosting live Leishmania amazonensis amastigotes, PLoS Negl Trop Dis, vol.6, issue.12, p.1980, 2012.

J. Sodek, B. Ganss, M. D. Mckee, and . Osteopontin, Crit Rev Oral Biol Med, vol.11, issue.3, pp.279-303, 2000.

T. Uede, Y. Katagiri, J. Iizuka, and M. Murakami, Osteopontin, a coordinator of host defense system: a cytokine or an extracellular adhesive protein?, Microbiol Immunol, vol.41, issue.9, pp.641-649, 1997.

O. 'regan, A. W. Nau, G. J. Chupp, G. L. Berman, and J. S. , Osteopontin (Eta-1) in cell-mediated immunity: teaching an old dog new tricks, Immunol Today, vol.21, issue.10, pp.475-483, 2000.

G. F. Weber and H. Cantor, The immunology of Eta-1/osteopontin, Cytokine Growth Factor Rev, vol.7, issue.3, pp.241-249, 1996.

R. Patarca, G. J. Freeman, R. P. Singh, F. Y. Wei, T. Durfee et al., Structural and functional studies of the early T lymphocyte activation 1 (Eta-1) gene. Definition of a novel T celldependent response associated with genetic resistance to bacterial infection, J Exp Med, vol.170, issue.1, pp.145-61, 1989.

S. Ashkar, G. F. Weber, V. Panoutsakopoulou, M. E. Sanchirico, M. Jansson et al., Eta-1 (osteopontin): an early component of type-1 (cell-mediated) immunity, Science, vol.287, issue.5454, pp.860-864, 2000.

L. Steinman and S. Zamvil, Transcriptional analysis of targets in multiple sclerosis, Nat Rev Immunol, vol.3, issue.6, pp.483-92, 2003.

B. Regnault, Y. Osorio, D. Miao, G. Eisenbarth, and E. Melanitou, Early over expression of messenger RNA for multiple genes, including insulin, in the pancreatic lymph nodes of NOD mice is associated with islet autoimmunity, BMC Med Genet, vol.2, p.63, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-01643691

C. Gao, H. Guo, Z. Mi, M. J. Grusby, and P. C. Kuo, Osteopontin induces ubiquitindependent degradation of STAT1 in RAW264.7 murine macrophages, J Immunol, vol.178, issue.3, pp.1870-81, 2007.

Q. Tang and J. A. Bluestone, Plasmacytoid DCs and T (reg) cells: casual acquaintance or monogamous relationship?, Nat Immunol, vol.7, issue.6, pp.551-554, 2006.

R. Patarca, R. A. Saavedra, and H. Cantor, Molecular and cellular basis of genetic resistance to bacterial infection: the role of the early T-lymphocyte activation-1/osteopontin gene, Crit Rev Immunol, vol.13, issue.3-4, pp.225-271, 1993.

E. M. Hur, S. Youssef, M. E. Haws, S. Y. Zhang, R. A. Sobel et al., Osteopontininduced relapse and progression of autoimmune brain disease through enhanced survival of activated T cells, Nat Immunol, vol.8, issue.1, pp.74-83, 2007.

G. Xu, H. Nie, N. Li, W. Zheng, D. Zhang et al., Role of osteopontin in amplification and perpetuation of rheumatoid synovitis, J Clin Invest, vol.115, issue.4, pp.1060-1067, 2005.

N. Gassler, F. Autschbach, S. Gauer, J. Bohn, B. Sido et al., Expression of osteopontin (Eta-1) in Crohn disease of the terminal ileum, Scand J Gastroenterol, vol.37, issue.11, pp.1286-95, 2002.

E. Melanitou, The autoimmune contrivance: genetics in the mouse model, Clin Immunol, vol.117, issue.3, pp.195-206, 2005.
URL : https://hal.archives-ouvertes.fr/pasteur-01643714

B. Zhu, K. Suzuki, H. A. Goldberg, S. R. Rittling, D. T. Denhardt et al., Osteopontin modulates CD44-dependent chemotaxis of peritoneal macrophages through G-protein-coupled receptors: evidence of a role for an intracellular form of osteopontin, J Cell Physiol, vol.198, issue.1, pp.155-67, 2004.

M. L. Shinohara, H. J. Kim, J. H. Kim, V. A. Garcia, and H. Cantor, Alternative translation of osteopontin generates intracellular and secreted isoforms that mediate distinct biological activities in dendritic cells, Proc Natl Acad Sci, vol.105, issue.20, pp.7235-7244, 2008.

E. Barak, S. Amin-spector, E. Gerliak, S. Goyard, N. Holland et al., Differentiation of Leishmania donovani in host-free system: analysis of signal perception and response, Mol Biochem Parasitol, vol.141, issue.1, pp.99-108, 2005.

K. Kawamura, K. Iyonaga, H. Ichiyasu, J. Nagano, M. Suga et al., Differentiation, maturation, and survival of dendritic cells by osteopontin regulation, Clin Diagn Lab Immunol, vol.12, issue.1, pp.206-218, 2005.

A. D. Howard, M. J. Kostura, N. Thornberry, G. J. Ding, G. Limjuco et al., IL-1-converting enzyme requires aspartic acid residues for processing of the IL-1 beta precursor at two distinct sites and does not cleave 31-kDa IL-1 alpha, J Immunol, vol.147, issue.9, pp.2964-2973, 1991.

T. Ghayur, S. Banerjee, M. Hugunin, D. Butler, L. Herzog et al., Caspase-1 processes IFN-gammainducing factor and regulates LPS-induced IFN-gamma production, Nature, vol.386, issue.6625, pp.619-642, 1997.

H. M. Hoffman and A. A. Wanderer, Inflammasome and IL-1beta-mediated disorders, Curr Allergy Asthma Rep, vol.10, issue.4, pp.229-264, 2010.

A. G. Porter and R. U. Janicke, Emerging roles of caspase-3 in apoptosis, Cell Death Differ, vol.6, issue.2, pp.99-104, 1999.

M. R. De-zoete, N. W. Palm, S. Zhu, R. A. Flavell, and . Inflammasomes, Cold Spring Harb Perspect Biol, vol.6, issue.12, p.16287, 2014.

K. Schroder and J. Tschopp, The inflammasomes, Cell, vol.140, issue.6, pp.821-853, 2010.

N. Inohara and G. Nunez, NODs: intracellular proteins involved in inflammation and apoptosis, Nat Rev Immunol, vol.3, issue.5, pp.371-82, 2003.

J. P. Ting, R. C. Lovering, E. S. Alnemri, J. Bertin, J. M. Boss et al., The NLR gene family: a standard nomenclature, Immunity, vol.28, issue.3, pp.285-292, 2008.

Y. Zhao, J. Yang, J. Shi, Y. N. Gong, Q. Lu et al., The NLRC4 inflammasome receptors for bacterial flagellin and type III secretion apparatus, Nature, vol.477, issue.7366, pp.596-600, 2011.

L. A. Sacramento, J. L. Costa, M. H. De-lima, P. A. Sampaio, R. P. Almeida et al., Toll-like receptor 2 is required for inflammatory process development during Leishmania infantum infection, Front Microbiol, vol.8, p.262, 2017.

R. C. Hespanhol, C. De-nazare, M. B. Meuser, S. De-nazareth, and S. Corte-real, The expression of mannose receptors in skin fibroblast and their involvement in Leishmania (L.) amazonensis invasion, J Histochem Cytochem, vol.53, issue.1, pp.35-44, 2005.

B. M. Scorza, M. A. Wacker, K. Messingham, P. Kim, A. Klingelhutz et al., Differential activation of human keratinocytes by Leishmania spp. causing localized or disseminated disease, J Invest Dermatol, vol.137, issue.10, pp.2149-56, 2017.

S. G. Reed, Z. A. Andrade, S. B. Roters, J. A. Inverso, and M. Sadigursky, Leishmania mexicana amazonensis infections in 'resistant' inbred mice following removal of the draining lymph node, Clin Exp Immunol, vol.64, issue.1, pp.8-13, 1986.

T. Uede and . Osteopontin, intrinsic tissue regulator of intractable inflammatory diseases, Pathol Int, vol.61, issue.5, pp.265-80, 2011.

G. J. Nau, G. L. Chupp, J. F. Emile, E. Jouanguy, J. S. Berman et al., Osteopontin expression correlates with clinical outcome in patients with mycobacterial infection, Am J Pathol, vol.157, issue.1, pp.37-42, 2000.

D. E. Jones, L. U. Buxbaum, and P. Scott, IL-4-independent inhibition of IL-12 responsiveness during Leishmania amazonensis infection, J Immunol, vol.165, issue.1, pp.364-72, 2000.

J. Ji, J. Sun, H. Qi, and L. Soong, Analysis of T helper cell responses during infection with Leishmania amazonensis, Am J Trop Med Hyg, vol.66, issue.4, pp.338-383, 2002.

D. S. Lima-junior, D. L. Costa, V. Carregaro, L. D. Cunha, A. L. Silva et al., Inflammasome-derived IL-1beta production induces nitric oxide-mediated resistance to Leishmania, Nat Med, vol.19, issue.7, pp.909-924, 2013.

R. Zhou, A. S. Yazdi, P. Menu, and J. Tschopp, A role for mitochondria in NLRP3 inflammasome activation, Nature, vol.469, issue.7329, pp.221-226, 2011.

L. Soong, J. C. Xu, I. S. Grewal, P. Kima, J. Sun et al., Disruption of CD40-CD40 ligand interactions results in an enhanced susceptibility to Leishmania amazonensis infection, Immunity, vol.4, issue.3, pp.263-73, 1996.

S. J. Green, M. S. Meltzer, J. B. Hibbs, and C. A. Nacy, Activated macrophages destroy intracellular Leishmania major amastigotes by an L-arginine-dependent killing mechanism, J Immunol, vol.144, issue.1, pp.278-83, 1990.

E. Linares, O. Augusto, S. C. Barao, and G. S. , Leishmania amazonensis infection does not inhibit systemic nitric oxide levels elicited by lipopolysaccharide in vivo, J Parasitol, vol.86, issue.1, pp.78-82, 2000.

L. Rozsa, J. Reiczigel, and G. Majoros, Quantifying parasites in samples of hosts, J Parasitol, vol.86, issue.2, pp.228-260, 2000.

H. Guo, C. Q. Cai, R. A. Schroeder, and P. C. Kuo, Osteopontin is a negative feedback regulator of nitric oxide synthesis in murine macrophages, J Immunol, vol.166, issue.2, pp.1079-86, 2001.

S. M. Hwang, C. A. Lopez, D. E. Heck, C. R. Gardner, D. L. Laskin et al., Osteopontin inhibits induction of nitric oxide synthase gene expression by inflammatory mediators in mouse kidney epithelial cells, J Biol Chem, vol.269, issue.1, pp.711-716, 1994.

F. Takahashi, K. Takahashi, K. Maeda, S. Tominaga, and Y. Fukuchi, Osteopontin is induced by nitric oxide in RAW 264.7 cells, IUBMB Life, vol.49, issue.3, pp.217-238, 2000.

T. Van-assche, M. Deschacht, R. A. Da-luz, L. Maes, and P. Cos, Leishmaniamacrophage interactions: insights into the redox biology, Free Radic Biol Med, vol.51, issue.2, pp.337-51, 2011.

N. Bortell, C. Flynn, B. Conti, H. S. Fox, and M. Marcondes, Osteopontin impacts West Nile virus pathogenesis and resistance by regulating Inflammasome components and cell death in the central nervous system at early time points, Mediat Inflamm, p.7582437, 2017.

O. Donnell, H. Pham, O. H. Li, L. X. Atif, S. M. Lee et al., Toll-like receptor and inflammasome signals converge to amplify the innate bactericidal capacity of T helper 1 cells, Immunity, vol.40, issue.2, pp.213-237, 2014.

D. J. Krysan, F. S. Sutterwala, and M. Wellington, Catching fire: Candida albicans, macrophages, and pyroptosis, PLoS Pathog, vol.10, issue.6, p.1004139, 2014.

D. Gris, Z. Ye, H. A. Iocca, H. Wen, R. R. Craven et al., NLRP3 plays a critical role in the development of experimental autoimmune encephalomyelitis by mediating Th1 and Th17 responses, J Immunol, vol.185, issue.2, pp.974-81, 2010.

P. J. Shaw, J. R. Lukens, S. Burns, H. Chi, M. A. Mcgargill et al., Cutting edge: critical role for PYCARD/ASC in the development of experimental autoimmune encephalomyelitis, J Immunol, vol.184, issue.9, pp.4610-4614, 2010.

S. K. Ippagunta, D. D. Brand, J. Luo, K. L. Boyd, C. Calabrese et al., Inflammasomeindependent role of apoptosis-associated speck-like protein containing a CARD (ASC) in T cell priming is critical for collagen-induced arthritis, J Biol Chem, vol.285, issue.16, pp.12454-62, 2010.

C. A. Yang and B. L. Chiang, Inflammasomes and human autoimmunity: a comprehensive review, J Autoimmun, vol.61, pp.1-8, 2015.

C. Dostert, K. Ludigs, and G. Guarda, Innate and adaptive effects of inflammasomes on T cell responses, Curr Opin Immunol, vol.25, issue.3, pp.359-65, 2013.

M. Yang, C. H. Hearnden, E. Oleszycka, and E. C. Lavelle, NLRP3 inflammasome activation and cytotoxicity induced by particulate adjuvants, Methods Mol Biol, vol.1040, pp.41-63, 2013.

P. Sagoo, Z. Garcia, B. Breart, F. Lemaitre, D. Michonneau et al., In vivo imaging of inflammasome activation reveals a subcapsular macrophage burst response that mobilizes innate and adaptive immunity, Nat Med, vol.22, issue.1, pp.64-71, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01253201

L. Liaw, D. E. Birk, C. B. Ballas, J. S. Whitsitt, J. M. Davidson et al., Altered wound healing in mice lacking a functional osteopontin gene (spp1), J Clin Invest, vol.101, issue.7, pp.1468-78, 1998.

T. Lang, S. Goyard, M. Lebastard, and G. Milon, Bioluminescent Leishmania expressing luciferase for rapid and high throughput screening of drugs acting on amastigote-harbouring macrophages and for quantitative realtime monitoring of parasitism features in living mice, Cell Microbiol, vol.7, issue.3, pp.383-92, 2005.

H. Lecoeur, E. De-la-llave, Y. Osorio, S. Goyard, H. Kiefer-biasizzo et al., Sorting of Leishmania-bearing dendritic cells reveals subtle parasite-induced modulation of host-cell gene expression, Microbes Infect, vol.12, issue.1, pp.46-54, 2010.

J. C. Antoine, C. Jouanne, and A. Ryter, Megasomes as the targets of leucine methyl ester in Leishmania amazonensis amastigotes, Parasitology, vol.99, pp.1-9, 1989.

N. Aulner, A. Danckaert, E. Rouault-hardoin, J. Desrivot, O. Helynck et al., High content analysis of primary macrophages hosting proliferating Leishmania amastigotes: application to anti-leishmanial drug discovery, PLoS Negl Trop Dis, vol.7, issue.4, p.2154, 2013.
URL : https://hal.archives-ouvertes.fr/pasteur-01433415

S. Goyard, H. Segawa, J. Gordon, M. Showalter, R. Duncan et al., An in vitro system for developmental and genetic studies of Leishmania donovani phosphoglycans, Mol Biochem Parasitol, vol.130, issue.1, pp.31-42, 2003.

G. F. Spath and S. M. Beverley, A lipophosphoglycan-independent method for isolation of infective Leishmania metacyclic promastigotes by density gradient centrifugation, Exp Parasitol, vol.99, issue.2, pp.97-103, 2001.

E. Giraud, T. Lestinova, T. Derrick, O. Martin, R. J. Dillon et al., Leishmania proteophosphoglycans regurgitated from infected sand flies accelerate dermal wound repair and exacerbate leishmaniasis via insulin-like growth factor 1-dependent signalling, PLoS Pathog, vol.14, issue.1, p.1006794, 2018.

E. Giraud, H. Lecoeur, E. Rouault, S. Goyard, G. Milon et al., A combined luciferase-expressing Leishmania imaging/RT-qPCR assay provides new insights into the sequential bilateral processes deployed in the ear pinna of C57BL/6 mice, Parasitol Int, vol.63, issue.1, pp.245-53, 2014.

E. De-la-llave, H. Lecoeur, A. Besse, G. Milon, E. Prina et al., A combined luciferase imaging and reverse transcription polymerase chain reaction assay for the study of Leishmania amastigote burden and correlated mouse tissue transcript fluctuations, Cell Microbiol, vol.13, issue.1, pp.81-91, 2011.

E. Prina, S. Z. Abdi, M. Lebastard, E. Perret, N. Winter et al., Dendritic cells as host cells for the promastigote and amastigote stages of Leishmania amazonensis: the role of opsonins in parasite uptake and dendritic cell maturation, J Cell Sci, vol.117, pp.315-340, 2004.

D. Biegel, G. Topper, and M. Rabinovitch, Leishmania mexicana: temperature sensitivity of isolated amastigotes and of amastigotes infecting macrophages in culture, Exp Parasitol, vol.56, issue.3, pp.289-97, 1983.

U. K. Laemmli, Cleavage of structural proteins during the assembly of the head of bacteriophage T4, Nature, vol.227, issue.5259, pp.680-685, 1970.

L. Lai, N. Alaverdi, L. Maltais, and H. C. Morse, Mouse cell surface antigens: nomenclature and immunophenotyping, J Immunol, vol.160, issue.8, pp.3861-3869, 1998.

A. J. Mcknight, A. J. Macfarlane, P. Dri, L. Turley, A. C. Willis et al., Molecular cloning of F4/80, a murine macrophage-restricted cell surface glycoprotein with homology to the G-protein-linked transmembrane 7 hormone receptor family, J Biol Chem, vol.271, issue.1, pp.486-495, 1996.

A. Schroeder, O. Mueller, S. Stocker, R. Salowsky, M. Leiber et al., The RIN: an RNA integrity number for assigning integrity values to RNA measurements, BMC Mol Biol, vol.7, p.3, 2006.

J. Hellemans, G. Mortier, D. Paepe, A. Speleman, F. Vandesompele et al., qBase relative quantification framework and software for management and automated analysis of real-time quantitative PCR data, Genome Biol, vol.8, issue.2, p.19, 2007.

J. Vandesompele, D. Preter, K. Pattyn, F. Poppe, B. Van-roy et al., Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biol, vol.3, issue.7, p.34, 2002.

C. L. Andersen, J. L. Jensen, and T. F. Orntoft, Normalization of real-time quantitative reverse transcription-PCR data: a model-based variance estimation approach to identify genes suited for normalization, applied to bladder and colon cancer data sets, Cancer Res, vol.64, issue.15, pp.5245-50, 2004.

M. Neuhauser and R. Poulin, Comparing parasite numbers between samples of hosts, J Parasitol, vol.90, issue.4, pp.689-91, 2004.

M. Neuhauser, J. Kotzmann, M. Walier, and R. Poulin, The comparison of mean crowding between two groups, J Parasitol, vol.96, issue.3, pp.477-81, 2010.

J. Reiczigel, Z. Lang, L. Rozsa, and B. Tothmeresz, Properties of crowding indices and statistical tools to analyze parasite crowding data, J Parasitol, vol.91, issue.2, pp.245-52, 2005.