J. Kloehn, M. Blume, S. A. Cobbold, E. C. Saunders, M. J. Dagley et al., Using metabolomics to dissect host-parasite interactions. Current Opinion in Microbiology, 2016.

T. Naderer and M. J. Mcconville, The Leishmania-macrophage interaction: A metabolic perspective, Cellular Microbiology, 2008.

L. O'neill, R. J. Kishton, and J. Rathmell, A guide to immunometabolism for immunologists, Nature Reviews Immunology, 2016.

C. Diskin and E. M. Pålsson-mcdermott, Metabolic modulation in macrophage effector function, Frontiers in Immunology, 2018.

P. Italiani and D. Boraschi, From monocytes to M1/M2 macrophages: Phenotypical vs. functional differentiation. Frontiers in Immunology, 2014.

M. Podinovskaia and A. Descoteaux, Leishmania and the macrophage: A multifaceted interaction, Future Microbiology, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01123316

D. Moreira, V. Rodrigues, M. Abengozar, L. Rivas, E. Rial et al., Leishmania infantum Modulates Host Macrophage Mitochondrial Metabolism by Hijacking the SIRT1-AMPK Axis, PLoS Pathog, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01136618

C. Yang, B. Ko, C. T. Hensley, L. Jiang, A. T. Wasti et al., Glutamine oxidation maintains the TCA cycle and cell survival during impaired mitochondrial pyruvate transport, Mol Cell, 2014.

A. K. Jha, S. Huang, A. Sergushichev, V. Lampropoulou, Y. Ivanova et al., Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization, Immunity, 2015.

P. S. Liu, H. Wang, X. Li, T. Chao, T. Teav et al., ?-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming, Nat Immunol, 2017.

J. W. Rob, Arts,1 12, * Boris Novakovic,2 Rob ter Horst,1 Agostinho Carvalho,3,4 Siroon Bekkering, p.1

F. Rodrigues, 4 Ricardo Silvestre,3,4 Shih-Chin Cheng,1,5 Shuang-Yin Wang, Ekta Lachmandas 1, vol.3, p.1

L. David, W. M. Williams-7,-jos, and . Van-der-meer, , p.1

H. G. Stunnenberg, J. Ramnik, and ;. Xavier, Glutaminolysis and Fumarate Accumulation Integrate Immunometabolic and Epigenetic Programs in Trained Immunity, Cell Metab, vol.10, p.11, 2016.

P. Castellano, L. Prevedel, S. Valdebenito, and E. A. Eugenin, HIV infection and latency induce a unique metabolic signature in human macrophages. Sci Rep, p.30850623, 2019.

V. A. Koeken, E. Lachmandas, A. Riza, V. Matzaraki, Y. Li et al., Role of Glutamine Metabolism in Host Defense Against Mycobacterium tuberculosis Infection, J Infect Dis, vol.219, pp.1662-1670, 2019.

L. V. Sinclair, J. Rolf, E. Emslie, Y. B. Shi, P. M. Taylor et al., Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation. Nat Immunol, 2013.

M. Nakaya, Y. Xiao, X. Zhou, J. H. Chang, M. Chang et al., Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation. Immunity, 2014.

D. Moreira, J. Estaquier, A. Cordeiro-da-silva, and R. Silvestre, Evaluating the role of host AMPK in Leishmania burden, Methods in Molecular Biology, 2018.

P. Tsagozis, E. Karagouni, and E. Dotsika, CD8+ T cells with parasite-specific cytotoxic activity and a Tc1 profile of cytokine and chemokine secretion develop in experimental visceral leishmaniasis. Parasite Immunol, 2003.

R. Silvestre, A. Cordeiro-da-silva, . Santaré-m-n, B. Vergnes, D. Sereno et al., SIR2-Deficient Leishmania infantum Induces a Defined IFN-?/IL-10 Pattern That Correlates with Protection, J Immunol, vol.179, pp.3161-3170, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00376744

N. Rolão, C. Melo, and L. Campino, Influence of the inoculation route in BALB/c mice infected by Leishmania infantum, Acta Trop, vol.90, pp.123-126, 2004.

I. Mesquita, C. Ferreira, A. Margarida, C. Machado, D. Moreira et al., Cytokine The impact of IL-10 dynamic modulation on host immune response against visceral leishmaniasis, Cytokine, pp.0-1, 2018.

M. Gleeson, Dosing and Efficacy of Glutamine Supplementation in Human Exercise and Sport Training, J Nutr, 2008.

E. Ward, S. Picton, U. Reid, D. Thomas, C. Gardener et al., Oral glutamine in paediatric oncology patients: A dose finding study, Eur J Clin Nutr, 2003.

E. Mok, B. Constantin, F. Favreau, N. Neveux, C. Magaud et al., L-glutamine administration reduces oxidized glutathione and MAP kinase signaling in dystrophic muscle of mdx mice, Pediatr Res, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02880257

V. Rodrigues, M. Laforge, L. Campillo-gimenez, C. Soundaramourty, A. Correia-de-oliveira et al., Abortive T Follicular Helper Development Is Associated with a Defective Humoral Response in Leishmania infantum-Infected Macaques, PLoS Pathog, 2014.

P. Newsholme, S. Gordon, and E. A. Newsholme, Rates of utilization and fates of glucose, glutamine, pyruvate, fatty acids and ketone bodies by mouse macrophages, Biochem J, 1987.

P. Newsholme, Why Is L-Glutamine Metabolism Important to Cells of the Immune System in Health, Postinjury, Surgery or Infection?2, J Nutr, 2001.

V. F. Cruzat, M. Krause, and P. Newsholme, Amino acid supplementation and impact on immune function in the context of exercise, Journal of the International Society of Sports Nutrition, p.25530736, 2014.

B. Saha, G. Das, H. Vohra, N. K. Ganguly, and G. C. Mishra, Macrophage-T cell interaction in experimental visceral leishmaniasis: failure to express costimulatory molecules on Leishmania-infected macrophages and its implication in the suppression of cell-mediated immunity, Eur J Immunol, 1995.

J. C. Antoine, T. Lang, E. Prina, N. Courret, and R. Hellio, H-2M molecules, like MHC class II molecules, are targeted to parasitophorous vacuoles of Leishmania-infected macrophages and internalized by amastigotes of L. amazonensis and L. mexicana, J Cell Sci, 1999.

K. Roy, S. Mandloi, S. Chakrabarti, and S. Roy, Cholesterol Corrects Altered Conformation of MHC-II Protein in Leishmania donovani Infected Macrophages: Implication in Therapy, PLoS Negl Trop Dis, 2016.

T. Bousoffara, H. Louzir, B. Salah, A. Dellagi, and K. , Analysis of Granzyme B Activity as a Surrogate Marker of Leishmania-Specific Cell-Mediated Cytotoxicity in Zoonotic Cutaneous Leishmaniasis, J Infect Dis, 2004.
URL : https://hal.archives-ouvertes.fr/pasteur-00875538

A. Spittler, S. Winkler, P. Gotzinger, R. Oehler, M. Willhelm et al., Influence of glutamine on the phenotype and function of human monocytes, 1995.

E. L. Carr, A. Kelman, G. S. Wu, R. Gopaul, E. Senkevitch et al., Glutamine Uptake and Metabolism Are Coordinately Regulated by ERK/MAPK during T Lymphocyte Activation, J Immunol, vol.185, pp.1037-1044, 2010.

J. Mo, M. M. Wolf, M. Z. Madden, G. Andrejeva, A. Sugiura et al., Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism, Cell, 2018.

F. A. Harding, J. G. Mcarthur, J. A. Gross, D. H. Raulet, and J. P. Allison, CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones, Nature, 1992.

T. M. Holling, E. Schooten, and P. J. Van-den-elsen, Function and regulation of MHC class II molecules in Tlymphocytes: Of mice and men, Hum Immunol, 2004.

R. Wang, C. P. Dillon, L. Z. Shi, S. Milasta, R. Carter et al., The Transcription Factor Myc Controls Metabolic Reprogramming upon T Lymphocyte Activation, Immunity, 2011.

E. L. Carr, A. Kelman, G. S. Wu, R. Gopaul, E. Senkevitch et al., Glutamine Uptake and Metabolism Are Coordinately Regulated by ERK/MAPK during T Lymphocyte Activation, J Immunol, 2010.

D. Klysz, X. Tai, P. A. Robert, M. Craveiro, G. Cretenet et al., Glutamine-dependent ?-ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation, Sci Signal, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02187398

T. Xu, K. M. Stewart, X. Wang, K. Liu, M. Xie et al., Metabolic control of TH17 and induced Treg cell balance by an epigenetic mechanism, Nature, 2017.

A. Santos, C. A. Correia, D. C. De-oliveira, A. Nogueira-pedro, P. Borelli et al., Intravenous Glutamine Administration Modulates TNF-?/IL-10 Ratio and Attenuates NFkB Phosphorylation in a Protein Malnutrition Model, Inflammation, vol.39, p.27565164, 2016.

E. C. Saunders, W. W. Ng, J. Kloehn, J. M. Chambers, M. Ng et al., Induction of a Stringent Metabolic Response in Intracellular Stages of Leishmania mexicana Leads to Increased Dependence on Mitochondrial Metabolism, PLoS Pathog, 2014.

T. Naderer, E. Wee, and M. J. Mcconville, Role of hexosamine biosynthesis in Leishmania growth and virulence, Mol Microbiol, 2008.

S. Pali?, P. Bhairosing, J. H. Beijnen, and T. Dorlo, Systematic Review of Host-Mediated Activity of Miltefosine in Leishmaniasis through Immunomodulation, Antimicrob Agents Chemother, vol.63, 2019.

S. Srivastava, J. Mishra, A. K. Gupta, A. Singh, P. Shankar et al., Laboratory confirmed miltefosine resistant cases of visceral leishmaniasis from India. Parasites and Vectors, p.28137296, 2017.

V. Cruzat, M. M. Rogero, K. N. Keane, R. Curi, and N. P. Glutamine, Metabolism and immune function, supplementation and clinical translation, Nutrients, 2018.