. Bonnel, F. Ird, and M. Kong-hap, IRD, France) for their helpful contribution in the immunological studies. We are grateful to the staff of the Regional Directorate of Public Health of Kairouan, Tunisia. Consortium members and affiliations R, Chamakh-Ayari, W. Markikou-Ouni, K. Aoun, M. Chenik and A

J. Meddeb-garnaoui, R. Pagniez, J. Bras-gonçalves, U. Lemesre, . Ird et al., Laboratory of Transmission, Control and Immunobiology of Infection, LR11-IPT-02, Institut Pasteur de Tunis, Tunisie; and recently joined UPSP EGEAL Institut Polytechnique LaSalle Beauvais, France. Faten Bel Haj Rhouma Safdarjung Hospital Campus Instituto de Medicina Tropical, National Institute of Pathology (ICMR) Lima, Peru. GM. Papierok: Virbac, pp.11-17

J. Alvar, I. Velez, C. Bern, M. Herrero, and P. Desjeux, Leishmaniasis Worldwide and Global Estimates of Its Incidence, PLoS ONE, vol.83, issue.30, p.35671, 2012.
DOI : 10.1371/journal.pone.0035671.s101

A. Salah, Y. Kamarianakis, S. Chlif, N. Alaya, and P. Prastacos, Zoonotic cutaneous leishmaniasis in central Tunisia: spatio temporal dynamics, International Journal of Epidemiology, vol.36, issue.5, 2007.
DOI : 10.1093/ije/dym125

URL : https://hal.archives-ouvertes.fr/pasteur-00871665

C. Lucas, E. Franke, M. Cachay, A. Tejada, and M. Cruz, Geographic distribution and clinical description of leishmaniasis cases in Peru, Am J Trop Med Hyg, vol.59, pp.312-317, 1998.

A. Stauch, R. Sarkar, A. Picado, B. Ostyn, and S. Sundar, Visceral Leishmaniasis in the Indian Subcontinent: Modelling Epidemiology and Control, PLoS Neglected Tropical Diseases, vol.3, issue.Pt 1, p.1405, 2011.
DOI : 10.1371/journal.pntd.0001405.s004

K. Aoun, F. Jeddi, F. Amri, J. Ghrab, and A. Bouratbine, Actualit??s ??pid??miologiques de la leishmaniose visc??rale en Tunisie, M??decine et Maladies Infectieuses, vol.39, issue.10, pp.775-779, 2009.
DOI : 10.1016/j.medmal.2009.08.010

M. Den-boer, D. Argaw, J. Jannin, and J. Alvar, Leishmaniasis impact and treatment access, Clinical Microbiology and Infection, vol.17, issue.10, pp.1471-1477, 2011.
DOI : 10.1111/j.1469-0691.2011.03635.x

D. Sacks and N. Noben-trauth, The immunology of susceptibility and resistance to leishmania major in mice, Nature Reviews Immunology, vol.2, issue.11, pp.845-858, 2002.
DOI : 10.1038/nri933

P. Mansueto, G. Vitale, D. Lorenzo, G. Rini, G. Mansueto et al., Immunopathology of Leishmaniasis: An Update, International Journal of Immunopathology and Pharmacology, vol.189, issue.3, pp.435-445, 2007.
DOI : 10.1177/039463200702000302

S. Nylen and S. Gautam, Immunological perspectives of leishmaniasis, Journal of Global Infectious Diseases, vol.2, issue.2, pp.135-146, 2010.
DOI : 10.4103/0974-777X.62876

E. Carvalho, C. Filho, D. Bacellar, O. Almeida, R. Lessa et al., Characterization of the immune response in subjects with self-healing cutaneous leishmaniasis, Am J Trop Med Hyg, vol.53, pp.273-277, 1995.

A. Kharazmi, K. Kemp, A. Ismail, S. Gasim, and A. Gaafar, T-cell response in human leishmaniasis, Immunology Letters, vol.65, issue.1-2, pp.105-108, 1999.
DOI : 10.1016/S0165-2478(98)00132-1

A. Sassi, H. Louzir, B. Salah, A. Mokni, M. et al., Leishmanin skin test lymphoproliferative responses and cytokine production after symptomatic or asymptomatic Leishmania major infection in Tunisia, Clinical and Experimental Immunology, vol.62, issue.1, pp.127-132, 1999.
DOI : 10.1084/jem.174.5.1209

URL : https://hal.archives-ouvertes.fr/pasteur-00878412

S. Ajdary, M. Alimohammadian, M. Eslami, K. Kemp, and A. Kharazmi, Comparison of the Immune Profile of Nonhealing Cutaneous Leishmaniasis Patients with Those with Active Lesions and Those Who Have Recovered from Infection, Infection and Immunity, vol.68, issue.4, pp.1760-1764, 2000.
DOI : 10.1128/IAI.68.4.1760-1764.2000

R. Coler, Y. Goto, L. Bogatzki, V. Raman, and S. Reed, Leish-111f, a Recombinant Polyprotein Vaccine That Protects against Visceral Leishmaniasis by Elicitation of CD4+ T Cells, Infection and Immunity, vol.75, issue.9, pp.4648-4654, 2007.
DOI : 10.1128/IAI.00394-07

P. Kushawaha, R. Gupta, S. Sundar, A. Sahasrabuddhe, and A. Dube, Elongation Factor-2, a Th1 Stimulatory Protein of Leishmania donovani, Generates Strong IFN-?? and IL-12 Response in Cured Leishmania-Infected Patients/Hamsters and Protects Hamsters against Leishmania Challenge, The Journal of Immunology, vol.187, issue.12, pp.6417-6427, 2011.
DOI : 10.4049/jimmunol.1102081

O. Singh, C. Stober, A. Singh, J. Blackwell, and S. Sundar, Cytokine Responses to Novel Antigens in an Indian Population Living in an Area Endemic for Visceral Leishmaniasis, PLoS Neglected Tropical Diseases, vol.21, issue.10, p.1874, 2012.
DOI : 10.1371/journal.pntd.0001874.s007

J. Mestas and C. Hughes, Of Mice and Not Men: Differences between Mouse and Human Immunology, The Journal of Immunology, vol.172, issue.5, pp.2731-2738, 2004.
DOI : 10.4049/jimmunol.172.5.2731

A. Devault and A. Banuls, The promastigote surface antigen gene family of the Leishmania parasite: differential evolution by positive selection and recombination, BMC Evolutionary Biology, vol.8, issue.1, p.292, 2008.
DOI : 10.1186/1471-2148-8-292

P. Murray, T. Spithill, and E. Handman, The PSA-2 glycoprotein complex of Leishmania major is a glycosylphosphatidylinositol-linked promastigote surface antigen, J Immunol, vol.143, pp.4221-4226, 1989.

K. Lohman, P. Langer, and D. Mcmahon-pratt, Molecular cloning and characterization of the immunologically protective surface glycoprotein GP46/M-2 of Leishmania amazonensis., Proceedings of the National Academy of Sciences, vol.87, issue.21, pp.8393-8397, 1990.
DOI : 10.1073/pnas.87.21.8393

T. Hanekamp and P. Langer, Molecular karyotype and chromosomal localization of genes encoding two major surface glycoproteins, gp63 and gp46/M2, hsp70, and ??-tubulin in cloned strains of several Leishmania species, Molecular and Biochemical Parasitology, vol.48, issue.1, pp.27-37, 1991.
DOI : 10.1016/0166-6851(91)90161-X

P. Murray and T. Spithill, Variants of a Leishmania surface antigen derived from a multigenic family, J Biol Chem, vol.266, pp.24477-24484, 1991.

F. Symons, P. Murray, J. H. Simpson, R. Osborn, and A. , Characterization of a polymorphic family of integral membrane proteins in promastigotes of different Leishmania species, Molecular and Biochemical Parasitology, vol.67, issue.1, pp.103-113, 1994.
DOI : 10.1016/0166-6851(94)90100-7

J. Beetham, K. Myung, J. Mccoy, M. Wilson, and J. Donelson, Glycoprotein 46 mRNA Abundance Is Post-transcriptionally Regulated during Development of Leishmania chagasiPromastigotes to an Infectious Form, Journal of Biological Chemistry, vol.272, issue.28, pp.17360-17366, 1997.
DOI : 10.1074/jbc.272.28.17360

A. Jimenez-ruiz, C. Boceta, P. Bonay, J. Requena, and C. Alonso, Cloning, sequencing, and expression of the PSA genes from Leishmania infantum, European Journal of Biochemistry, vol.251, issue.1-2, pp.389-397, 1998.
DOI : 10.1046/j.1432-1327.1998.2510389.x

L. Kedzierski, J. Montgomery, D. Bullen, C. J. Gardiner, and E. , A Leucine-Rich Repeat Motif of Leishmania Parasite Surface Antigen 2 Binds to Macrophages through the Complement Receptor 3, The Journal of Immunology, vol.172, issue.8, pp.4902-4906, 2004.
DOI : 10.4049/jimmunol.172.8.4902

D. Mcmahon-pratt, D. Rodriguez, J. Rodriguez, Y. Zhang, and K. Manson, Recombinant vaccinia viruses expressing GP46/M-2 protect against Leishmania infection, Infect Immun, vol.61, pp.3351-3359, 1993.

E. Handman, F. Symons, T. Baldwin, J. Curtis, and J. Scheerlinck, Protective vaccination with promastigote surface antigen 2 from Leishmania major is mediated by a TH1 type of immune response, Infect Immun, vol.63, pp.4261-4267, 1995.

C. Boceta, C. Alonso, and A. Jimenez-ruiz, Leucine rich repeats are the main epitopes in Leishmania infantum PSA during canine and human visceral leishmaniasis, Parasite Immunology, vol.85, issue.2, pp.55-62, 2000.
DOI : 10.1016/0300-9084(94)90009-4

J. Beetham, J. Donelson, and R. Dahlin, Surface glycoprotein PSA (GP46) expression during short- and long-term culture of Leishmania chagasi, Molecular and Biochemical Parasitology, vol.131, issue.2, pp.109-117, 2003.
DOI : 10.1016/S0166-6851(03)00197-X

E. Handman, A. Osborn, F. Symons, R. Van-driel, and R. Cappai, The Leishmania promastigote surface antigen 2 complex is differentially expressed during the parasite life cycle, Molecular and Biochemical Parasitology, vol.74, issue.2, pp.189-200, 1995.
DOI : 10.1016/0166-6851(95)02500-6

D. Mcmahon-pratt, Y. Traub-cseko, K. Lohman, D. Rogers, and S. Beverley, Loss of the GP46/M-2 surface membrane glycoprotein gene family in the Leishmania braziliensis complex, Molecular and Biochemical Parasitology, vol.50, issue.1, pp.151-160, 1992.
DOI : 10.1016/0166-6851(92)90252-F

K. Myung, J. Beetham, M. Wilson, and J. Donelson, Comparison of the Post-transcriptional Regulation of the mRNAs for the Surface Proteins PSA (GP46) and MSP (GP63) ofLeishmania chagasi, Journal of Biological Chemistry, vol.277, issue.19, pp.16489-16497, 2002.
DOI : 10.1074/jbc.M200174200

L. Lincoln, M. Ozaki, J. Donelson, and J. Beetham, Genetic complementation of Leishmania deficient in PSA (GP46) restores their resistance to lysis by complement, Molecular and Biochemical Parasitology, vol.137, issue.1, pp.185-189, 2004.
DOI : 10.1016/j.molbiopara.2004.05.004

J. Lemesre, P. Holzmuller, M. Cavaleyra, R. Goncalves, and G. Hottin, Protection against experimental visceral leishmaniasis infection in dogs immunized with purified excreted secreted antigens of Leishmania infantum promastigotes, Vaccine, vol.23, issue.22, pp.2825-2840, 2005.
DOI : 10.1016/j.vaccine.2004.11.061

J. Lemesre, P. Holzmuller, R. Goncalves, G. Bourdoiseau, and C. Hugnet, Long-lasting protection against canine visceral leishmaniasis using the LiESAp-MDP vaccine in endemic areas of France: Double-blind randomised efficacy field trial, Vaccine, vol.25, issue.21, pp.4223-4234, 2007.
DOI : 10.1016/j.vaccine.2007.02.083

J. Lemesre, Methods for the culture in vitro of different stages of tissue parasites. International publication nu WO94/226899, 1994.

T. Merlen, D. Sereno, N. Brajon, F. Rostand, and J. Lemesre, Leishmania spp: completely defined medium without serum and macromolecules (CDM/LP) for the continuous in vitro cultivation of infective promastigote forms, Am J Trop Med Hyg, vol.60, pp.41-50, 1999.

E. Morgan, R. Varro, H. Sepulveda, J. Ember, and J. Apgar, Cytometric bead array: a multiplexed assay platform with applications in various areas of biology, Clinical Immunology, vol.110, issue.3, pp.252-266, 2004.
DOI : 10.1016/j.clim.2003.11.017

J. Champsi and D. Mcmahon-pratt, Membrane glycoprotein M-2 protects against Leishmania amazonensis infection, Infect Immun, vol.56, pp.3272-3279, 1988.

A. Sjolander, T. Baldwin, J. Curtis, and E. Handman, Induction of a Th1 immune response and simultaneous lack of activation of a Th2 response are required for generation of immunity to leishmaniasis, J Immunol, vol.160, pp.3949-3957, 1998.

R. Breitling, S. Klingner, N. Callewaert, R. Pietrucha, and A. Geyer, Non-pathogenic trypanosomatid protozoa as a platform for protein research and production, Protein Expression and Purification, vol.25, issue.2, pp.209-218, 2002.
DOI : 10.1016/S1046-5928(02)00001-3

J. Burns, . Jr, J. Scott, E. Carvalho, D. Russo et al., Characterization of a membrane antigen of Leishmania amazonensis that stimulates human immune responses, J Immunol, vol.146, pp.742-748, 1991.

K. Moore, O. Garra, A. De-waal-malefyt, R. Vieira, P. Mosmann et al., Interleukin-10, Annual Review of Immunology, vol.11, issue.1, pp.165-190, 1993.
DOI : 10.1146/annurev.iy.11.040193.001121

D. Jankovic, D. Kugler, and A. Sher, IL-10 production by CD4+ effector T cells: a mechanism for self-regulation, Mucosal Immunology, vol.175, issue.3, pp.239-246, 2010.
DOI : 10.1016/j.immuni.2008.12.009

J. Alexander and F. Brombacher, T Helper1/T Helper2 Cells and Resistance/Susceptibility to Leishmania Infection: Is This Paradigm Still Relevant?, Frontiers in Immunology, vol.3, p.80, 2012.
DOI : 10.3389/fimmu.2012.00080

T. Bousoffara, H. Louzir, B. Salah, A. Dellagi, and K. , Specific Cell???Mediated Cytotoxicity in Zoonotic Cutaneous Leishmaniasis, The Journal of Infectious Diseases, vol.189, issue.7, pp.1265-1273, 2004.
DOI : 10.1086/382031

URL : https://hal.archives-ouvertes.fr/pasteur-00875538

J. Hernandez-ruiz, N. Salaiza-suazo, G. Carrada, S. Escoto, and A. Ruiz-remigio, CD8 Cells of Patients with Diffuse Cutaneous Leishmaniasis Display Functional Exhaustion: The Latter Is Reversed, In Vitro, by TLR2 Agonists, PLoS Neglected Tropical Diseases, vol.124, issue.5, p.871, 2010.
DOI : 10.1371/journal.pntd.0000871.g006

C. Santos, V. Boaventura, R. Cardoso, C. Tavares, N. Lordelo et al., CD8+ Granzyme B+???Mediated Tissue Injury vs. CD4+IFN??+???Mediated Parasite Killing in Human Cutaneous Leishmaniasis, Journal of Investigative Dermatology, vol.133, issue.6, pp.1533-1540, 2013.
DOI : 10.1038/jid.2013.4

W. Grossman, J. Verbsky, B. Tollefsen, C. Kemper, and J. Atkinson, Differential expression of granzymes A and B in human cytotoxic lymphocyte subsets and T regulatory cells, Blood, vol.104, issue.9, pp.2840-2848, 2004.
DOI : 10.1182/blood-2004-03-0859

D. Vignali, L. Collison, and C. Workman, How regulatory T cells work, Nature Reviews Immunology, vol.178, issue.7, pp.523-532, 2008.
DOI : 10.1038/nri2343

I. Afonina, G. Tynan, S. Logue, S. Cullen, and M. Bots, Granzyme B-Dependent Proteolysis Acts as a Switch to Enhance the Proinflammatory Activity of IL-1??, Molecular Cell, vol.44, issue.2, pp.265-278, 2011.
DOI : 10.1016/j.molcel.2011.07.037

P. Hiebert and D. Granville, Granzyme B in injury, inflammation, and repair, Trends in Molecular Medicine, vol.18, issue.12, pp.732-741, 2012.
DOI : 10.1016/j.molmed.2012.09.009

C. Magnani, G. Alberigo, R. Bacchetta, G. Serafini, and M. Andreani, Killing of myeloid APCs via HLA class I, CD2 and CD226 defines a novel mechanism of suppression by human Tr1 cells, European Journal of Immunology, vol.294, issue.6, pp.1652-1662, 2011.
DOI : 10.1002/eji.201041120

B. Dondji, E. Deak, K. Goldsmith-pestana, E. Perez-jimenez, and M. Esteban, Intradermal NKT cell activation during DNA priming in heterologous prime-boost vaccination enhances T cell responses and protection againstLeishmania, European Journal of Immunology, vol.63, issue.3, pp.706-719, 2008.
DOI : 10.1002/eji.200737660

I. Aguilar-be, R. Da-silva-zardo, P. De-souza, E. Borja-cabrera, G. Rosado-vallado et al., Cross-Protective Efficacy of a Prophylactic Leishmania donovani DNA Vaccine against Visceral and Cutaneous Murine Leishmaniasis, Infection and Immunity, vol.73, issue.2, pp.812-819, 2005.
DOI : 10.1128/IAI.73.2.812-819.2005

Y. Goto, A. Bhatia, V. Raman, S. Vidal, and S. Bertholet, Leishmania infantum sterol 24-c-methyltransferase formulated with MPL-SE induces cross-protection against L. major infection, Vaccine, vol.27, issue.21, pp.2884-2890, 2009.
DOI : 10.1016/j.vaccine.2009.02.079

L. Ramirez, D. Santos, A. Souza, E. Coelho, and A. Barral, Evaluation of immune responses and analysis of the effect of vaccination of the Leishmania major recombinant ribosomal proteins L3 or L5 in two different murine models of cutaneous leishmaniasis, Vaccine, vol.31, issue.9, pp.1312-1319, 2013.
DOI : 10.1016/j.vaccine.2012.12.071

P. Melby, Y. J. Zhao, W. Perez, L. Cheng, and J. , Leishmania donovani p36(LACK) DNA Vaccine Is Highly Immunogenic but Not Protective against Experimental Visceral Leishmaniasis, Infection and Immunity, vol.69, issue.8, pp.4719-4725, 2001.
DOI : 10.1128/IAI.69.8.4719-4725.2001

G. Salay, M. Dorta, N. Santos, R. Mortara, and C. Brodskyn, Testing of Four Leishmania Vaccine Candidates in a Mouse Model of Infection with Leishmania (Viannia) braziliensis, the Main Causative Agent of Cutaneous Leishmaniasis in the New World, Clinical and Vaccine Immunology, vol.14, issue.9, pp.1173-1181, 2007.
DOI : 10.1128/CVI.00060-07

W. Tonui and R. Titus, Cross-protection against Leishmania donovani but not L. Braziliensis caused by vaccination with L. Major soluble promastigote exogenous antigens in BALB/c mice, Am J Trop Med Hyg, vol.76, pp.579-584, 2007.

C. Peacock, K. Seeger, D. Harris, L. Murphy, and J. Ruiz, Comparative genomic analysis of three Leishmania species that cause diverse human disease, Nature Genetics, vol.13, issue.7, pp.839-847, 2007.
DOI : 10.1093/bioinformatics/bti1018

URL : https://hal.archives-ouvertes.fr/hal-00169316