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.315-325, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02682017

L. G. Ng, A. Hsu, M. A. Mandell, B. Roediger, C. Hoeller et al., Migratory dermal dendritic cells act as rapid sensors of protozoan parasites, PLoS Pathog, vol.4, p.1000222, 2008.

N. C. Peters, J. G. Egen, N. Secundino, A. Debrabant, N. Kimblin et al., In vivo imaging reveals an essential role for neutrophils in leishmaniasis transmitted by sand flies, Science, vol.321, pp.970-974, 2008.

A. C. Misslitz, K. Bonhagen, D. Harbecke, C. Lippuner, T. Kamradt et al., Two waves of antigen-containing dendritic cells in vivo in experimental Leishmania major infection, Eur. J. Immunol, vol.34, pp.715-725, 2004.

A. Kissenpfennig, S. Henri, B. Dubois, C. Laplace-builhe, P. Perrin et al., Dynamics and function of Langerhans cells in vivo: dermal dendritic cells colonize lymph node areas distinct from slower migrating Langerhans cells, Immunity, vol.22, pp.643-654, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00165695

L. Xin, K. Li, and L. Soong, Down-regulation of dendritic cell signaling pathways by Leishmania amazonensis amastigotes, Mol. Immunol, vol.45, pp.3371-3382, 2008.

C. Auffray, M. H. Sieweke, and F. Geissmann, Blood monocytes: development, heterogeneity, and relationship with dendritic cells, Annu. Rev. Immunol, vol.27, pp.669-692, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00407757

Y. Xu, Y. Zhan, A. M. Lew, S. H. Naik, and M. H. Kershaw, Differential development of murine dendritic cells by GM-CSF versus Flt3 ligand has implications for inflammation and trafficking, J. Immunol, vol.179, pp.7577-7584, 2007.

B. J. Bevis and B. S. Glick, Rapidly maturing variants of the Discosoma red fluorescent protein (DsRed), Nat. Biotechnol, vol.20, pp.83-87, 2000.

I. Schmid, C. Lambert, D. Ambrozak, G. E. Marti, D. M. Moss et al., International society for analytical cytology biosafety standard for sorting of unfixed cells, vol.71, pp.414-437, 2007.

I. Schmid, J. K. Nicholson, J. V. Giorgi, G. Janossy, A. Kunkl et al., Biosafety guidelines for sorting of unfixed cells, Cytometry, vol.28, pp.99-117, 1997.

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, pp.31-42, 2003.

M. C. Bosetto and S. Giorgio, Leishmania amazonensis: multiple receptorligand interactions are involved in amastigote infection of human dendritic cells, Exp. Parasitol, vol.116, pp.306-310, 2007.

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

H. Lecoeur, A. Langonne, L. Baux, D. Rebouillat, P. Rustin et al., Real-time flow cytometry analysis of permeability transition in isolated mitochondria, Exp. Cell Res, vol.294, pp.106-117, 2004.

I. Méderlé, I. Bourguin, D. Ensergueix, E. Badell, J. Moniz-peireira et al., Plasmidic versus insertional cloning of heterologous genes in mycobacterium bovis BCG: impact on in vivo antigen persistence and immune responses, Infect. Immun, vol.70, pp.303-314, 2002.

T. Zal, A. Volkmann, and B. Stockinger, Mechanisms of tolerance induction in major histocompatibility complex class ii-restricted t-cells specific for a blood-borne self-antigen, J. Exp. Med, vol.180, pp.2089-2099, 1994.

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, 2006.

S. Fleige, V. Walf, S. Huch, C. Prgomet, J. Sehm et al., Comparison of relative mRNA quantification models and the impact of RNA integrity in quantitative real-time RT-PCR, Biotechnol. Lett, vol.28, pp.1601-1613, 2006.

O. Wagner-ballon, H. Chagraoui, E. Prina, M. Tulliez, G. Milon et al., Monocyte/macrophage dysfunctions do not impair the promotion of myelofibrosis by high levels of thrombopoietin, J. Immunol, vol.176, pp.6425-6433, 2006.

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

J. Vandesompele, K. De, F. Preter, B. Pattyn, N. Poppe et al., Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biol, vol.18, 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, pp.5245-5250, 2004.

J. Osorio-y-fortea, E. Prina, T. Lang, G. Milon, C. Davory et al., Affycqc: a web interface to detect outlying genechips with extreme studentized deviate tests, J. Bioinform. Comput. Biol, vol.6, pp.317-334, 2008.

N. Jain, J. Thatte, T. Braciale, K. Ley, M. O'connell et al., Localpooled-error test for identifying differentially expressed genes with a small number of replicated microarrays, Bioinformatics, vol.19, pp.1945-1951, 2003.

Y. Benjamini and Y. Hochberg, Controlling the false discovery rate e a practical and powerful approach to multiple testing, J. R. Stat. Soc., Ser. B, Stat. Methodol, vol.19, pp.289-300, 1995.

E. Prina, E. Roux, D. Mattei, and G. Milon, Leishmania DNA is rapidly degraded following parasite death: an analysis by microscopy and realtime PCR, Microbes Infect, vol.9, pp.1307-1315, 2007.
URL : https://hal.archives-ouvertes.fr/pasteur-00181350

B. Leon, M. Lopez-bravo, and C. Ardavin, Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania, Immunity, vol.26, pp.519-531, 2007.

T. Gotoh and M. Mori, Arginase II downregulates nitric oxide (NO) production and prevents NO-mediated apoptosis in murine macrophagederived RAW 264.7 cells, J. Cell Biol, vol.144, pp.427-434, 1999.

S. C. Roberts, M. J. Tancer, M. R. Polinsky, K. M. Gibson, O. Heby et al., Arginase plays a pivotal role in polyamine precursor metabolism in Leishmania. Characterization of gene deletion mutants, J. Biol. Chem, vol.279, pp.23668-23678, 2004.

A. E. Pegg, Spermidine/spermine-N(1)-acetyltransferase: a key metabolic regulator, Am J Physiol Endocrinol Metab, vol.294, pp.995-1010, 2008.

C. De-trez, S. Magez, S. Akira, B. Ryffel, Y. Carlier et al., iNOSproducing inflammatory dendritic cells constitute the major infected cell type during the chronic Leishmania major infection phase of C57BL/6 resistant mice, PLoS Pathog, vol.5, p.1000494, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00591434