A. Tran and P. Gual, Non-alcoholic steatohepatitis in morbidly obese patients, Clin Res Hepatol Gastroenterol, vol.37, pp.17-29, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00583896

Z. M. Younossi, A. B. Koenig, D. Abdelatif, Y. Fazel, L. Henry et al., Global epidemiology of nonalcoholic fatty liver disease-Meta-analytic assessment of prevalence, incidence, and outcomes, Hepatology, vol.64, pp.73-84, 2016.

H. Tilg, P. D. Cani, and E. A. Mayer, Gut microbiome and liver diseases, Gut, vol.65, pp.2035-2079, 2016.

M. V. Machado and A. M. Diehl, Pathogenesis of nonalcoholic steatohepatitis, Gastroenterology, vol.150, pp.1769-77, 2016.

M. J. Hubler and A. J. Kennedy, Role of lipids in the metabolism and activation of immune cells, J Nutr Biochem, vol.34, pp.1-7, 2016.

C. Luci, A. Reynders, . Ivanov, . Ii, C. Cognet et al., Influence of the transcription factor RORgammat on the development of NKp46+ cell populations in gut and skin, Nat Immunol, vol.10, pp.75-82, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00403251

M. Cella, K. Otero, and M. Colonna, Expansion of human NK-22 cells with IL-7, IL-2, and IL-1beta reveals intrinsic functional plasticity, Proc Natl Acad Sci USA, vol.107, pp.10961-10967, 2010.

F. Vely, V. Barlogis, B. Vallentin, B. Neven, C. Piperoglou et al., Evidence of innate lymphoid cell redundancy in humans, Nat Immunol, vol.17, pp.1291-1300, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01482373

Y. Huang, K. Mao, and R. N. Germain, Thinking differently about ILCs-Not just tissue resident and not just the same as CD4(+) T-cell effectors, Immunol Rev, vol.286, pp.160-71, 2018.

H. Spits, D. Artis, M. Colonna, A. Diefenbach, D. Santo et al., Innate lymphoid cells-a proposal for uniform nomenclature, Nat Rev Immunol, vol.13, pp.145-154, 2013.

E. Vivier, D. Artis, M. Colonna, A. Diefenbach, D. Santo et al., Innate Lymphoid Cells: 10 Years On, Cell, vol.174, pp.1054-66, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-02101002

M. Ebbo, A. Crinier, F. Vely, and E. Vivier, Innate lymphoid cells: major players in inflammatory diseases, Nat Rev Immunol, vol.17, pp.665-78, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01764670

T. E. O'sullivan and J. C. Sun, Innate lymphoid cell immunometabolism, J Mol Biol, vol.429, pp.3577-86, 2017.

C. Possot, S. Schmutz, S. Chea, L. Boucontet, L. A. Cumano et al., Notch signaling is necessary for adult, but not fet al., development of RORgammat(+) innate lymphoid cells, Nat Immunol, vol.12, pp.949-58, 2011.

X. Yu, Y. Wang, M. Deng, Y. Li, K. A. Ruhn et al., The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor, Elife, p.3, 2014.

C. Klose, M. Flach, L. Mohle, L. Rogell, T. Hoyler et al., Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages, Cell, vol.157, pp.340-56, 2014.

M. G. Constantinides, B. D. Mcdonald, P. A. Verhoef, and A. Bendelac, A committed precursor to innate lymphoid cells, Nature, vol.508, pp.397-401, 2014.

I. E. Ishizuka, S. Chea, H. Gudjonson, M. G. Constantinides, A. R. Dinner et al., Single-cell analysis defines the divergence between the innate lymphoid cell lineage and lymphoid tissue-inducer cell lineage, Nat Immunol, vol.17, pp.269-76, 2016.
URL : https://hal.archives-ouvertes.fr/pasteur-01289322

S. M. Gordon, J. Chaix, L. J. Rupp, J. Wu, S. Madera et al., The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation, Immunity, vol.36, pp.55-67, 2012.

C. Daussy, F. Faure, K. Mayol, S. Viel, G. Gasteiger et al., Tbet and Eomes instruct the development of two distinct natural killer cell lineages in the liver and in the bone marrow, J Exp Med, vol.211, pp.563-77, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00972070

F. D. Shi, H. G. Ljunggren, L. Cava, A. , V. Kaer et al., Organ-specific features of natural killer cells, Nat Rev Immunol, vol.11, pp.658-71, 2011.

D. K. Sojka, B. Plougastel-douglas, L. Yang, M. A. Pak-wittel, M. N. Artyomov et al., Tissue-resident natural killer (NK) cells are cell lineages distinct from thymic and conventional splenic NK cells, Elife, vol.3, p.1659, 2014.

V. S. Cortez, A. Fuchs, M. Cella, S. Gilfillan, and M. Colonna, Cutting edge: salivary gland NK cells develop independently of Nfil3 in steady-state, J Immunol, vol.192, pp.4487-91, 2014.

Y. Gao, F. Souza-fonseca-guimaraes, T. Bald, S. S. Ng, A. Young et al., Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells, Nat Immunol, vol.18, pp.1004-1019, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01765092

V. S. Cortez, T. K. Ulland, L. Cervantes-barragan, J. K. Bando, M. L. Robinette et al., SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing non-canonical TGF-beta signaling, Nat Immunol, vol.18, pp.995-1003, 2017.

O. E. Weizman, N. M. Adams, I. S. Schuster, C. Krishna, Y. Pritykin et al., ILC1 confer early host protection at initial sites of viral infection, vol.171, pp.795-808, 2017.

T. Perchet, S. Chea, M. Hasan, A. Cumano, and R. Golub, Single-cell gene expression using multiplex RT-qPCR to characterize heterogeneity of rare lymphoid populations, J Vis Exp, vol.19, p.119, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01473735

O. Pikovskaya, J. Chaix, N. J. Rothman, A. Collins, Y. H. Chen et al., Cutting edge: eomesodermin is sufficient to direct type 1 innate lymphocyte development into the conventional NK Lineage, J Immunol, vol.196, pp.1449-54, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01438533

L. K. Mackay, M. Minnich, N. A. Kragten, Y. Liao, B. Nota et al., Hobit and Blimp1 instruct a universal transcriptional program of tissue residency in lymphocytes, Science, vol.352, pp.459-63, 2016.

T. Perchet, M. Petit, E. G. Banchi, S. Meunier, A. Cumano et al., The notch signaling pathway is balancing type 1 innate lymphoid cell immune functions, Front Immunol, vol.9, p.1252, 2018.
URL : https://hal.archives-ouvertes.fr/pasteur-01828956

H. Peng and Z. Tian, Diversity of tissue-resident NK cells, Semin Immunol, vol.31, pp.3-10, 2017.

L. Tang, H. Peng, J. Zhou, Y. Chen, H. Wei et al., Differential phenotypic and functional properties of liver-resident NK cells and mucosal ILC1s, J Autoimmun, vol.67, pp.29-35, 2016.

C. A. Vosshenrich, T. Ranson, S. I. Samson, E. Corcuff, F. Colucci et al., Roles for common cytokine receptor gamma-chain-dependent cytokines in the generation, differentiation, and maturation of NK cell precursors and peripheral NK cells in vivo, J Immunol, vol.174, pp.1213-1234, 2005.

N. S. Williams, J. Klem, I. J. Puzanov, P. V. Sivakumar, M. Bennett et al., Differentiation of NK1.1+, Ly49+ NK cells from flt3+ multipotent marrow progenitor cells, J Immunol, vol.163, pp.2648-56, 1999.

A. Fuchs, W. Vermi, J. S. Lee, S. Lonardi, S. Gilfillan et al., Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12-and IL-15-responsive IFN-gamma-producing cells, Immunity, vol.38, pp.769-81, 2013.

J. H. Bernink, C. P. Peters, M. Munneke, A. A. Te-velde, S. L. Meijer et al., Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues, Nat Immunol, vol.14, pp.221-230, 2013.

G. S. Hotamisligil, Inflammation and metabolic disorders, Nature, vol.444, pp.860-867, 2006.

S. Nishimura, I. Manabe, M. Nagasaki, K. Eto, H. Yamashita et al., CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity, Nat Med, vol.15, pp.914-934, 2009.

M. Feuerer, L. Herrero, D. Cipolletta, A. Naaz, J. Wong et al., but not obese, fat is enriched for a unique population of regulatory T cells that affect metabolic parameters, Nat Med, vol.15, pp.930-939, 2009.

T. E. O'sullivan, M. Rapp, X. Fan, O. E. Weizman, P. Bhardwaj et al., Adipose-Resident group 1 innate lymphoid cells promote obesity-associated insulin resistance, Immunity, vol.45, pp.428-469, 2016.

S. Boulenouar, X. Michelet, D. Duquette, D. Alvarez, A. E. Hogan et al., Adipose type one innate lymphoid cells regulate macrophage homeostasis through targeted cytotoxicity, vol.46, pp.273-86, 2017.

F. M. Wensveen, V. Jelencic, S. Valentic, M. Sestan, T. T. Wensveen et al., NK cells link obesity-induced adipose stress to inflammation and insulin resistance, Nat Immunol, vol.16, pp.376-85, 2015.

B. C. Lee, M. S. Kim, M. Pae, Y. Yamamoto, D. Eberle et al., Adipose natural killer cells regulate adipose tissue macrophages to promote insulin resistance in obesity, Cell Metab, vol.23, pp.685-98, 2016.

R. W. O'rourke, K. A. Meyer, C. K. Neeley, G. D. Gaston, P. Sekhri et al., Systemic NK cell ablation attenuates intra-abdominal adipose tissue macrophage infiltration in murine obesity, Obesity, vol.22, pp.2109-2123, 2014.

N. Satoh-takayama, C. A. Vosshenrich, S. Lesjean-pottier, S. Sawa, M. Lochner et al., Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense, Immunity, vol.29, pp.958-70, 2008.
URL : https://hal.archives-ouvertes.fr/pasteur-01402754

L. C. Rankin, M. J. Girard-madoux, C. Seillet, L. A. Mielke, Y. Kerdiles et al., Complementarity and redundancy of IL-22-producing innate lymphoid cells, Nat Immunol, vol.17, pp.179-86, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01440245

A. Fuchs, ILC1s in Tissue inflammation and infection, Front Immunol, vol.7, p.104, 2016.

C. Vonarbourg, A. Mortha, V. L. Bui, P. P. Hernandez, E. A. Kiss et al., Regulated expression of nuclear receptor RORgammat confers distinct functional fates to NK cell receptorexpressing RORgammat(+) innate lymphocytes, Immunity, vol.33, pp.736-51, 2010.

C. S. Klose, E. A. Kiss, V. Schwierzeck, K. Ebert, T. Hoyler et al., A T-bet gradient controls the fate and function of CCR6-RORgammat+ innate lymphoid cells, Nature, vol.494, pp.261-266, 2013.

J. H. Bernink, L. Krabbendam, K. Germar, J. E. De, K. Gronke et al., Interleukin-12 and?23 control plasticity of CD127(+) group 1 and group 3 innate lymphoid cells in the intestinal lamina propria, Immunity, vol.43, pp.146-60, 2015.

A. Reynders, N. Yessaad, T. P. Vu-manh, M. Dalod, A. Fenis et al., Identity, regulation and in vivo function of gut NKp46+RORgammat+ and NKp46+RORgammat-lymphoid cells, EMBO J, vol.30, pp.2934-2981, 2011.

S. Li, J. W. Bostick, J. Ye, J. Qiu, B. Zhang et al., Aryl hydrocarbon receptor signaling cell intrinsically inhibits intestinal group 2 innate lymphoid cell function, Immunity, vol.49, pp.915-943, 2018.

J. H. Shin, L. Zhang, O. Murillo-sauca, J. Kim, H. E. Kohrt et al., Modulation of natural killer cell antitumor activity by the aryl hydrocarbon receptor, Proc Natl Acad Sci, vol.110, pp.12391-12397, 2013.

E. T. Saltzman, T. Palacios, M. Thomsen, and L. Vitetta, Intestinal microbiome shifts, dysbiosis, inflammation, and non-alcoholic fatty liver disease, Front Microbiol, vol.9, p.61, 2018.

G. Den-besten, K. Van-eunen, A. K. Groen, K. Venema, D. J. Reijngoud et al., The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, J Lipid Res, vol.54, 2013.

A. Trompette, E. S. Gollwitzer, K. Yadava, A. K. Sichelstiel, N. Sprenger et al., Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, Nat Med, vol.20, pp.159-66, 2014.

A. Trompette, E. S. Gollwitzer, C. Pattaroni, I. C. Lopez-mejia, R. E. Pernot et al., Dietary fiber confers protection against flu by shaping Ly6c(-) patrolling monocyte hematopoiesis and CD8(+) T cell metabolism, Immunity, vol.48, pp.992-1005, 2018.

M. L. Balmer, E. H. Ma, G. R. Bantug, J. Grahlert, S. Pfister et al., Memory CD8(+) T cells require increased concentrations of acetate induced by stress for optimal function, Immunity, vol.44, pp.1312-1336, 2016.

K. Takeda, E. Cretney, Y. Hayakawa, T. Ota, H. Akiba et al., TRAIL identifies immature natural killer cells in newborn mice and adult mouse liver, Blood, vol.105, pp.2082-2091, 2005.

J. Zhang, M. Marotel, S. Fauteux-daniel, A. L. Mathieu, S. Viel et al., Tbet and Eomes govern differentiation and function of mouse and human NK cells and ILC1, Eur J Immunol, vol.48, pp.738-50, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01911191

H. Peng, X. Jiang, Y. Chen, D. K. Sojka, H. Wei et al., Liver-resident NK cells confer adaptive immunity in skin-contact inflammation, J Clin Invest, vol.123, pp.1444-56, 2013.

S. Paust, B. Senman, and U. H. Von-andrian, Adaptive immune responses mediated by natural killer cells, Immunol Rev, vol.235, pp.286-96, 2010.

M. Forkel, L. Berglin, E. Kekalainen, A. Carlsson, E. Svedin et al., Composition and functionality of the intrahepatic innate lymphoid cellcompartment in human nonfibrotic and fibrotic livers, Eur J Immunol, vol.47, pp.1280-94, 2017.

C. Harmon, M. W. Robinson, R. Fahey, S. Whelan, D. D. Houlihan et al., Tissue-resident Eomes(hi) T-bet(lo) CD56(bright) NK cells with reduced proinflammatory potential are enriched in the adult human liver, Eur J Immunol, vol.46, pp.2111-2131, 2016.

K. A. Stegmann, F. Robertson, N. Hansi, U. Gill, C. Pallant et al., CXCR6 marks a novel subset of T-bet(lo)Eomes(hi) natural killer cells residing in human liver, Sci Rep, vol.6, p.26157, 2016.

K. Hudspeth, M. Donadon, M. Cimino, E. Pontarini, P. Tentorio et al., Human liver-resident CD56(bright)/CD16(neg) NK cells are retained within hepatic sinusoids via the engagement of CCR5 and CXCR6 pathways, J Autoimmun, vol.66, pp.40-50, 2016.

N. Marquardt, V. Beziat, S. Nystrom, J. Hengst, M. A. Ivarsson et al., Cutting edge: identification and characterization of human intrahepatic CD49a+ NK cells, J Immunol, vol.194, pp.2467-71, 2015.

Z. Tian, Y. Chen, and B. Gao, Natural killer cells in liver disease, Hepatology, vol.57, pp.1654-62, 2013.

S. Radaeva, R. Sun, B. Jaruga, V. T. Nguyen, Z. Tian et al., Natural killer cells ameliorate liver fibrosis by killing activated stellate cells in NKG2D-dependent and tumor necrosis factor-related apoptosisinducing ligand-dependent manners, Gastroenterology, vol.130, pp.435-52, 2006.

C. Gur, S. Doron, S. Kfir-erenfeld, E. Horwitz, L. Abu-tair et al., NKp46-mediated killing of human and mouse hepatic stellate cells attenuates liver fibrosis, Gut, vol.61, pp.885-93, 2012.

T. Li, Y. Yang, H. Song, H. Li, A. Cui et al., Activated NK cells kill hepatic stellate cells via p38/PI3K signaling in a TRAIL-involved degranulation manner, J Leukoc Biol, vol.105, pp.695-704, 2019.

Z. Yang, T. Tang, X. Wei, S. Yang, and Z. Tian, Type 1 innate lymphoid cells contribute to the pathogenesis of chronic hepatitis B, Innate Immun, vol.21, pp.665-73, 2015.

P. D. Krueger, S. Narayanan, F. A. Surette, M. G. Brown, S. J. Sung et al., Murine liver-resident group 1 innate lymphoid cells regulate optimal priming of anti-viral CD8+ T cells, J Leukoc Biol, vol.101, pp.329-367, 2017.

Y. Cepero-donates, G. Lacraz, F. Ghobadi, V. Rakotoarivelo, S. Orkhis et al., Interleukin-15-mediated inflammation promotes non-alcoholic fatty liver disease, Cytokine, vol.82, pp.102-113, 2016.

A. Bertola, S. Bonnafous, R. Anty, S. Patouraux, M. C. Saint-paul et al., Hepatic expression patterns of inflammatory and immune response genes associated with obesity and NASH in morbidly obese patients, PLoS ONE, vol.5, 2010.

A. Marcais, S. Viel, M. Grau, T. Henry, J. Marvel et al., Regulation of mouse NK cell development and function by cytokines, Front Immunol, vol.4, p.450, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00972074

Y. Kamari, A. Shaish, E. Vax, S. Shemesh, M. Kandel-kfir et al., Lack of interleukin-1alpha or interleukin-1beta inhibits transformation of steatosis to steatohepatitis and liver fibrosis in hypercholesterolemic mice, J Hepatol, vol.55, pp.1086-94, 2011.

S. Stojsavljevic, G. Palcic, M. , V. Jukic, L. et al., Adipokines and proinflammatory cytokines, the key mediators in the pathogenesis of nonalcoholic fatty liver disease, World J Gastroenterol, vol.20, pp.18070-91, 2014.

A. Bertola, V. Deveaux, S. Bonnafous, D. Rousseau, R. Anty et al., Elevated expression of osteopontin may be related to adipose tissue macrophage accumulation and liver steatosis in morbid obesity, Diabetes, vol.58, pp.125-158, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00419058

S. Mimura, S. Mochida, M. Inao, A. Matsui, S. Nagoshi et al., Massive liver necrosis after provocation of imbalance between Th1 and Th2 immune reactions in osteopontin transgenic mice, J Gastroenterol, vol.39, pp.867-72, 2004.

S. A. Lund, C. M. Giachelli, and M. Scatena, The role of osteopontin in inflammatory processes, J Cell Commun Signal, vol.3, pp.311-333, 2009.

J. W. Chung, M. S. Kim, Z. H. Piao, M. Jeong, S. R. Yoon et al., Osteopontin promotes the development of natural killer cells from hematopoietic stem cells, Stem Cells, vol.26, pp.2114-2137, 2008.

J. W. Leavenworth, B. Verbinnen, Q. Wang, E. Shen, and H. Cantor, Intracellular osteopontin regulates homeostasis and function of natural killer cells, Proc Natl Acad Sci, vol.112, pp.494-503, 2015.

M. Greter, J. Helft, A. Chow, D. Hashimoto, A. Mortha et al., GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells, Immunity, vol.36, pp.1031-1077, 2012.

Y. Wolf, A. Shemer, M. Polonsky, M. Gross, A. Mildner et al., Autonomous TNF is critical for in vivo monocyte survival in steady state and inflammation, J Exp Med, vol.214, pp.905-922, 2017.

S. Patouraux, D. Rousseau, S. Bonnafous, C. Lebeaupin, C. Luci et al., CD44 is a key player in non-alcoholic steatohepatitis, J Hepatol, vol.67, pp.328-366, 2017.

K. Kazankov, S. Jorgensen, K. L. Thomsen, H. J. Moller, H. Vilstrup et al., The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, Nat Rev Gastroenterol Hepatol, 2018.

A. Tosello-trampont, F. A. Surette, S. E. Ewald, and Y. S. Hahn, Immunoregulatory role of NK cells in tissue inflammation and regeneration. Front Immunol, vol.8, p.301, 2017.

A. Marcais, J. Cherfils-vicini, C. Viant, S. Degouve, S. Viel et al., The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells, Nat Immunol, vol.15, pp.749-57, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01911540

M. P. Keppel, N. Saucier, A. Y. Mah, T. P. Vogel, and M. A. Cooper, Activation-specific metabolic requirements for NK Cell IFN-gamma production, J Immunol, vol.194, pp.1954-62, 2015.

S. Viel, L. Besson, E. Charrier, A. Marcais, E. Disse et al., Alteration of Natural Killer cell phenotype and function in obese individuals, Clin Immunol, vol.177, pp.12-19, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01796182

A. Marcais, M. Marotel, S. Degouve, A. Koenig, S. Fauteux-daniel et al., High mTOR activity is a hallmark of reactive natural killer cells and amplifies early signaling through activating receptors, p.6, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01764666

N. Assmann, K. L. O'brien, R. P. Donnelly, L. Dyck, V. Zaiatz-bittencourt et al., Srebp-controlled glucose metabolism is essential for NK cell functional responses, Nat Immunol, vol.18, pp.1197-206, 2017.

E. E. Calle, R. Kaaks, and . Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms, Nat Rev Cancer, vol.4, pp.579-91, 2004.

M. Kompoti, A. Mariolis, A. Alevizos, I. Kyrazis, I. Protopsaltis et al., Elevated serum triglycerides is the strongest single indicator for the presence of metabolic syndrome in patients with type 2 diabetes, Cardiovasc Diabetol, vol.5, p.21, 2006.

X. Michelet, L. Dyck, A. Hogan, R. M. Loftus, D. Duquette et al., Metabolic reprogramming of natural killer cells in obesity limits antitumor responses, Nat Immunol, vol.19, pp.1330-1370, 2018.