R. Pearson and A. Sousa, Clinical Spectrum of Leishmaniasis, Clinical Infectious Diseases, vol.22, issue.1, pp.1-13, 1996.
DOI : 10.1093/clinids/22.1.1

E. Zijlstra and A. Hassan, 3. Visceral leishmaniasis, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol.6, issue.supplement 1, pp.27-58, 2001.
DOI : 10.1046/j.1365-3156.2001.00680.x

F. Chappuis, S. Sundar, and A. Hailu, Visceral leishmaniasis: what are the needs for diagnosis, treatment and control?, Nature Reviews Microbiology, vol.14, issue.11, pp.873-82, 2007.
DOI : 10.1128/CMR.14.2.229-243.2001

S. El-safi, B. Bucheton, and M. Kheir, Epidemiology of visceral leishmaniasis in Atbara River area, eastern Sudan: the outbreak of Barbar El Fugara village (1996???1997), Microbes and Infection, vol.4, issue.14, pp.1439-1486, 1996.
DOI : 10.1016/S1286-4579(02)00026-6

B. Bucheton, M. Kheir, and S. El-safi, The interplay between environmental and host factors during an outbreak of visceral leishmaniasis in eastern Sudan, Microbes and Infection, vol.4, issue.14, pp.1449-57, 2002.
DOI : 10.1016/S1286-4579(02)00027-8

B. Bucheton, L. Abel, and S. El-safi, A Major Susceptibility Locus on Chromosome 22q12 Plays a Critical Role in the Control of Kala-Azar, The American Journal of Human Genetics, vol.73, issue.5, pp.1052-60, 2003.
DOI : 10.1086/379084

S. Rijal, B. Ostyn, and S. Uranw, Increasing Failure of Miltefosine in the Treatment of Kala-azar in Nepal and the Potential Role of Parasite Drug Resistance, Reinfection, or Noncompliance, Clinical Infectious Diseases, vol.377, issue.11, pp.1530-1538, 2013.
DOI : 10.1016/S0140-6736(10)62050-8

H. Yang and K. Wang, Genomic variant annotation and prioritization with ANNOVAR and wANNOVAR, Nature Protocols, vol.10, issue.10, pp.1556-66, 2015.
DOI : 10.1093/bioinformatics/btr330

URL : http://europepmc.org/articles/pmc4718734?pdf=render

C. Ncbi-resource, Database resources of the National Center for Biotechnology Information, Nucleic Acids Res, vol.44, pp.7-19, 2016.

D. Salgado, J. Desvignes, and G. Rai, UMD-Predictor: A High-Throughput Sequencing Compliant System for Pathogenicity Prediction of any Human cDNA Substitution, Human Mutation, vol.452, issue.Database issue, pp.439-485, 2016.
DOI : 10.1038/nature06884

URL : https://hal.archives-ouvertes.fr/hal-01670164

H. Thorvaldsdóttir, J. Robinson, and J. Mesirov, Integrative Genomics Viewer (IGV): high-performance genomics data visualization and exploration, Briefings in Bioinformatics, vol.463, issue.7283, pp.178-92, 2013.
DOI : 10.1038/nature08822

A. Untergasser, I. Cutcutache, and T. Koressaar, Primer3???new capabilities and interfaces, Primer3?new capabilities and interfaces, p.115, 2012.
DOI : 10.6026/97320630003282

URL : https://academic.oup.com/nar/article-pdf/40/15/e115/16967451/gks596.pdf

K. Watschinger, J. Fuchs, and V. Yarov-yarovoy, Catalytic residues and a predicted structure of tetrahydrobiopterin-dependent alkylglycerol mono-oxygenase, Biochemical Journal, vol.247, issue.1, pp.279-86, 2012.
DOI : 10.1093/nar/gkp885

M. Auer-grumbach, S. Toegel, and M. Schabhüttl, Rare Variants in MME , Encoding Metalloprotease Neprilysin, Are Linked to Late-Onset Autosomal-Dominant Axonal Polyneuropathies, The American Journal of Human Genetics, vol.99, issue.3, pp.607-630, 2016.
DOI : 10.1016/j.ajhg.2016.07.008

URL : https://doi.org/10.1016/j.ajhg.2016.07.008

N. Orr, R. Arnaout, and L. Gula, A mutation in the atrial-specific myosin light chain gene (MYL4) causes familial atrial fibrillation, Nature Communications, vol.24, p.11303, 2016.
DOI : 10.1007/s10822-010-9352-6

H. Kuehn, W. Ouyang, and B. Lo, Immune dysregulation in human subjects with heterozygous germline mutations in CTLA4, Science, vol.37, issue.12, pp.1623-1630, 2014.
DOI : 10.1002/gepi.21743

Q. Zhou, H. Wang, and D. Schwartz, Loss-of-function mutations in TNFAIP3 leading to A20 haploinsufficiency cause an early-onset autoinflammatory disease, Nature Genetics, vol.21, issue.1, pp.67-73, 2016.
DOI : 10.1016/j.jaci.2014.02.013

X. Kong, G. Vogt, and Y. Itan, Haploinsufficiency at the human IFNGR2 locus contributes to mycobacterial disease, Human Molecular Genetics, vol.115, issue.4, pp.769-81, 2013.
DOI : 10.1172/JCI200523139

A. Sakthianandeswaren, S. Foote, and E. Handman, The role of host genetics in leishmaniasis, Trends in Parasitology, vol.25, issue.8, pp.383-91, 2009.
DOI : 10.1016/j.pt.2009.05.004

B. Bucheton, L. Argiro, and C. Chevillard, Identification of a novel G245R polymorphism in the IL-2 receptor ?? membrane proximal domain associated with human visceral leishmaniasis, Genes & Immunity, vol.171, issue.Suppl 1, pp.79-83, 2007.
DOI : 10.1084/jem.171.5.1821

URL : https://hal.archives-ouvertes.fr/hal-01592707

B. Bucheton, L. Abel, and M. Kheir, Genetic control of visceral leishmaniasis in a Sudanese population: candidate gene testing indicates a linkage to the NRAMP1 region, Genes & Immunity, vol.67, issue.Suppl 1, pp.104-113, 2003.
DOI : 10.1086/316895

URL : https://hal.archives-ouvertes.fr/hal-01592716

H. Mohamed, M. Ibrahim, and E. Miller, SLC11A1 (formerly NRAMP1) and susceptibility to visceral leishmaniasis in The Sudan, European Journal of Human Genetics, vol.2, issue.Suppl 1, pp.66-74, 2004.
DOI : 10.1016/S1286-4579(00)00295-1

M. Fakiola, A. Strange, and H. Cordell, Common variants in the HLA-DRB1???HLA-DQA1 HLA class II region are associated with susceptibility to visceral leishmaniasis, Nature Genetics, vol.21, issue.2, pp.208-221, 2013.
DOI : 10.1214/09-STS311

S. Jamieson, E. Miller, and C. Peacock, Genome-wide scan for visceral leishmaniasis susceptibility genes in Brazil, Genes & Immunity, vol.117, issue.1, pp.84-90, 2007.
DOI : 10.1038/75514

M. Fakiola, E. Miller, and M. Fadl, Genetic and Functional Evidence Implicating DLL1 as the Gene That Influences Susceptibility to Visceral Leishmaniasis at Chromosome 6q27, The Journal of Infectious Diseases, vol.165, issue.3, pp.467-77, 2011.
DOI : 10.1093/infdis/165.3.535

A. Mishra, S. Nizamuddin, and G. Arekatla, IL10 Variant g.5311A Is Associated with Visceral Leishmaniasis in Indian Population, PLOS ONE, vol.2011, issue.36, p.124559, 2015.
DOI : 10.1371/journal.pone.0124559.t006

D. Kumar, P. Tiwary, J. Chakravarty, and S. Sundar, Association of interleukin-18 gene polymorphism with susceptibility to visceral leishmaniasis in endemic area of Bihar, an Indian population, Sci World J, vol.2014, p.852104, 2014.

H. Mohamed, M. Ibrahim, and E. Miller, Genetic susceptibility to visceral leishmaniasis in The Sudan: linkage and association with IL4 and IFNGR1, Genes & Immunity, vol.52, issue.5, pp.351-356, 2003.
DOI : 10.1038/75514

M. Rasouli, M. Keshavarz, M. Kalani, A. Moravej, S. Kiany et al., Toll-like receptor 4 (TLR4) polymorphisms in Iranian patients with visceral leishmaniasis, Molecular Biology Reports, vol.53, issue.12, pp.10795-802, 2012.
DOI : 10.1016/j.jinf.2005.10.018

S. Mehrotra, M. Fakiola, and J. Oommen, Genetic and functional evaluation of the role of CXCR1 and CXCR2 in susceptibility to visceral leishmaniasis in north-east India, BMC Medical Genetics, vol.16, issue.9, p.162, 2011.
DOI : 10.1038/nm.2209

A. Frade, L. Oliveira, and D. Costa, TGFB1 and IL8 gene polymorphisms and susceptibility to visceral leishmaniasis, Infection, Genetics and Evolution, vol.11, issue.5, pp.912-918, 2011.
DOI : 10.1016/j.meegid.2011.02.014

URL : https://lirias.kuleuven.be/bitstream/123456789/347370/2/InfGenEvol2011TGFbIL8Leish.pdf

A. Grant, A. Sabri, and A. Abid, A genome-wide association study of pulmonary tuberculosis in Morocco, Human Genetics, vol.132, issue.3, pp.299-307, 2016.
DOI : 10.1007/s00439-013-1282-7

J. Casanova and L. Abel, : The Human Model, Annual Review of Immunology, vol.20, issue.1, pp.581-620, 2002.
DOI : 10.1146/annurev.immunol.20.081501.125851

K. Watschinger and E. Werner, Alkylglycerol monooxygenase, IUBMB Life, vol.42, issue.4, pp.366-72, 2013.
DOI : 10.1021/bi035656u

URL : http://onlinelibrary.wiley.com/doi/10.1002/iub.1143/pdf

S. Tokuoka, Y. Kita, H. Shindou, and T. Shimizu, Alkylglycerol monooxygenase as a potential modulator for PAF synthesis in macrophages, Biochemical and Biophysical Research Communications, vol.436, issue.2, pp.306-318, 2013.
DOI : 10.1016/j.bbrc.2013.05.099

S. Ishii and T. Shimizu, Platelet-activating factor (PAF) receptor and genetically engineered PAF receptor mutant mice, Progress in Lipid Research, vol.39, issue.1, pp.41-82, 2000.
DOI : 10.1016/S0163-7827(99)00016-8

M. Lonardoni, M. Russo, and S. Jancar, Essential Role of Platelet-Activating Factor in Control of Leishmania (Leishmania) amazonensis Infection, Infection and Immunity, vol.68, issue.11, pp.6355-61, 2000.
DOI : 10.1128/IAI.68.11.6355-6361.2000

H. Santiago, B. Pires, M. Souza, and D. , Platelet activating factor receptor-deficient mice present delayed interferon-?? upregulation and high susceptibility to Leishmania amazonensis infection, Microbes and Infection, vol.8, issue.11, pp.2569-77, 2006.
DOI : 10.1016/j.micinf.2006.06.011

S. Das, K. Pandey, V. Rabidas, A. Mandal, and P. Das, Effectiveness of miltefosine treatment in targeting anti-leishmanial HO-1/Nrf-2-mediated oxidative responses in visceral leishmaniasis patients, Journal of Antimicrobial Chemotherapy, vol.28, issue.9, pp.2059-65, 2013.
DOI : 10.1016/j.it.2007.07.004

P. Gangalum, W. De-castro, and L. Vieira, Platelet-Activating Factor Receptor Contributes to Antileishmanial Function of Miltefosine, The Journal of Immunology, vol.194, issue.12, pp.5961-5968, 2015.
DOI : 10.4049/jimmunol.1401890

P. Wadhone, M. Maiti, R. Agarwal, V. Kamat, S. Martin et al., Miltefosine Promotes IFN-??-Dominated Anti-Leishmanial Immune Response, The Journal of Immunology, vol.182, issue.11, pp.7146-54, 2009.
DOI : 10.4049/jimmunol.0803859

URL : http://www.jimmunol.org/content/jimmunol/182/11/7146.full.pdf

D. Berkovic, U. Grunwald, W. Menzel, C. Unger, W. Hiddemann et al., Effects of hexadecylphosphocholine on membrane phospholipid metabolism in human tumour cells, European Journal of Cancer, vol.31, issue.12, pp.2080-2085, 1995.
DOI : 10.1016/0959-8049(95)00350-9

S. Croft and J. Engel, Miltefosine ??? discovery of the antileishmanial activity of phospholipid derivatives, Transactions of the Royal Society of Tropical Medicine and Hygiene, vol.73, issue.Suppl. 1, pp.4-8, 2006.
DOI : 10.1007/BF02535532

P. Hilgard, E. Kampherm, L. Nolan, J. Pohl, and T. Reissmann, Investigation into the immunological effects of miltefosine, a new anticancer agent under development, Journal of Cancer Research and Clinical Oncology, vol.84, issue.5, pp.403-411, 1991.
DOI : 10.1007/BF01612758

I. Eue, Hexadecylphosphocholine selectively upregulates expression of intracellular adhesion molecule-1 and class I major histocompatibility complex antigen in human monocytes, Journal of Experimental Therapeutics and Oncology, vol.1, issue.5, pp.333-339, 2002.
DOI : 10.1006/bbrc.1996.0854