, As an alarmin, IL-33 early contributes to I/R liver injuries by recruiting iNKT cells that would in turn amplify the recruitment of immuno-inflammatory cells together with IL-33

R. J. Ploeg, D. 'alessandro, A. M. Knechtle, S. J. Stegall, M. D. Pirsch et al., Risk factors for primary dysfunction after liver transplantation--a multivariate analysis, Transplantation, vol.55, pp.807-820, 1993.

G. Varotti, G. L. Grazi, G. Vetrone, G. Ercolani, M. Cescon et al., Causes of early acute graft failure after liver transplantation: analysis of a 17-year single-centre experience, Clin. Transplant, vol.19, pp.492-500, 2005.

P. Kron, A. Schlegel, X. Muller, A. Gaspert, P. Clavien et al., Hypothermic Oxygenated Perfusion: A Simple and Effective Method to Modulate the Immune Response in Kidney Transplantation, Transplantation, vol.103, pp.128-136, 2019.

A. Schlegel, P. Kron, R. Graf, P. A. Clavien, and P. Dutkowski, Hypothermic Oxygenated Perfusion (HOPE) downregulates the immune response in a rat model of liver transplantation, Ann. Surg, vol.260, pp.931-938, 2014.

S. O'neill, A. Roebuck, E. Khoo, S. J. Wigmore, and E. M. Harrison, A meta-analysis and metaregression of outcomes including biliary complications in donation after cardiac death liver transplantation, Transpl. Int, vol.27, pp.1159-1174, 2014.

D. Hide, A. Warren, A. Fernández-iglesias, R. Maeso-díaz, C. Peralta et al.,

, Ischemia/reperfusion injury in the aged liver: The importance of the sinusoidal endothelium in developing therapeutic strategies for the elderly, J. Gerontol. A. Biol. Sci. Med. Sci, 2019.

P. Dutkowski, J. V. Guarrera, J. De-jonge, P. N. Martins, R. J. Porte et al., Evolving Trends in Machine Perfusion for Liver Transplantation, Gastroenterology, vol.94, pp.1542-1547, 2019.

C. Cayrol and J. Girard, Interleukin-33 (IL-33): A nuclear cytokine from the IL-1 family, Immunol. Rev, vol.281, pp.154-168, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02363721

C. Moussion, N. Ortega, and J. Girard, The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel "alarmin, PLoS One, vol.3, p.3331, 2008.

L. Barbier, M. Ferhat, E. Salamé, A. Robin, A. Herbelin et al.,

, Family Cytokines: Keystones in Liver Inflammatory Diseases. Front. Immunol, vol.10, 2014.

M. Ferhat, A. Robin, S. Giraud, S. Sena, J. Goujon et al., , p.33

, Contributes to Kidney Ischemia-Reperfusion Injury as an Alarmin, J. Am. Soc. Nephrol, vol.29, pp.1272-1288, 2018.

H. O. Yazdani, H. Chen, S. Tohme, S. Tai, D. J. Van-der-windt et al., IL-33 exacerbates liver sterile inflammation by amplifying neutrophil extracellular trap formation, J. Hepatol, vol.68, pp.130-139, 2017.

N. Sakai, H. L. Van-sweringen, R. C. Quillin, R. Schuster, J. Blanchard et al., Interleukin-33 is hepatoprotective during liver ischemia/reperfusion in mice

, Hepatology, vol.56, pp.1468-78, 2012.

M. Roth-g-a,-zimmermann, . Lubsczyk-b-a, J. Pilz, P. Faybik, and H. Hetz, Up-regulation of interleukin 33 and soluble ST2 serum levels in liver failure, J. Surg. Res, vol.163, pp.79-83, 2010.

Y. Zhai, H. Petrowsky, J. C. Hong, R. W. Busuttil, and J. W. Kupiec-weglinski, Ischaemiareperfusion injury in liver transplantation--from bench to bedside, Nat. Rev

, Gastroenterol. Hepatol, vol.10, pp.79-89, 2013.

L. Panqueva, R. Del, and P. , Biopsia hepática en la patología del trasplante, período postrasplante temprano, enfoque dirigido al diagnóstico histopatológico y su correlación clínicopatológica, Rev. Colomb. Gastroenterol, vol.31, p.169, 2017.

A. Matton and R. J. Porte, Opportunities for scientific expansion of the deceased donor pool, Liver Transpl, vol.20, issue.2, p.5, 2014.

V. Racanelli and B. Rehermann, The liver as an immunological organ, Hepatology, vol.43, 2006.

N. Aizarani, A. Saviano, . Sagar, L. Mailly, S. Durand et al., A human liver cell atlas reveals heterogeneity and epithelial progenitors, Nature, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02309750

F. Winau, G. Hegasy, R. Weiskirchen, S. Weber, C. Cassan et al., Ito Cells Are Liver-Resident Antigen-Presenting Cells for Activating T Cell Responses, Immunity, vol.26, pp.117-129, 2007.

R. Charles, H. S. Chou, L. Wang, J. J. Fung, L. Lu et al., Human hepatic stellate cells inhibit t-cell response through B7-H1 pathway, Transplantation, vol.96, pp.17-24, 2013.

E. Gomez-perdiguero, K. Klapproth, C. Schulz, K. Busch, E. Azzoni et al., Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors, Nature, vol.518, pp.547-551, 2015.

R. Soysa, X. Wu, and I. N. Crispe, Dendritic cells in hepatitis and liver transplantation, Liver Transplant, vol.23, pp.1433-1439, 2017.

F. Sierro, M. Evrard, S. Rizzetto, M. Melino, A. J. Mitchell et al., A Liver Capsular Network of Monocyte-Derived Macrophages Restricts Hepatic Dissemination of Intraperitoneal Bacteria by Neutrophil Recruitment, Immunity, vol.47, pp.374-388, 2017.

F. Heymann and F. Tacke, Immunology in the liver-from homeostasis to disease, Nat. Rev. Gastroenterol. Hepatol, vol.13, pp.88-110, 2016.

W. De-graaf, K. P. Van-lienden, T. M. Van-gulik, and R. J. Bennink, (99m)Tc-mebrofenin hepatobiliary scintigraphy with SPECT for the assessment of hepatic function and liver functional volume before partial hepatectomy, J. Nucl. Med, vol.51, pp.229-265, 2010.

S. Dinant, W. De-graaf, B. J. Verwer, R. J. Bennink, K. P. Van-lienden et al., Risk assessment of posthepatectomy liver failure using hepatobiliary scintigraphy and CT volumetry, J. Nucl. Med, vol.48, pp.685-92, 2007.

N. Fausto, J. S. Campbell, and K. J. Riehle, Liver regeneration, Hepatology, vol.43, pp.45-53, 2006.

G. K. Michalopoulos, Hepatostat: Liver regeneration and normal liver tissue maintenance, Hepatology, vol.65, pp.1384-1392, 2017.

Y. Tao, M. Wang, E. Chen, and H. Tang, Liver Regeneration: Analysis of the Main Relevant Signaling Molecules, Mediators Inflamm, 2017.

W. Li, X. Liang, J. I. Leu, K. Kovalovich, G. Ciliberto et al., Global changes in interleukin-6-dependent gene expression patterns in mouse livers after partial hepatectomy, Hepatology, vol.33, pp.1377-86, 2001.

G. K. Michalopoulos, Liver regeneration, J. Cell. Physiol, vol.213, pp.286-300, 2007.

N. Selzner, M. Selzner, B. Odermatt, Y. Tian, N. Van-rooijen et al., ICAM-1 triggers liver regeneration through leukocyte recruitment and Kupffer cell-dependent release of TNFalpha/IL-6 in mice, Gastroenterology, vol.124, pp.692-700, 2003.

J. W. Grisham, A morphologic study of deoxyribonucleic acid synthesis and cell proliferation in regenerating rat liver; autoradiography with thymidine-H3, Cancer Res, vol.22, pp.842-851, 1962.

G. K. Michalopoulos and Z. Khan, Liver regeneration, growth factors, and amphiregulin, Gastroenterology, vol.128, pp.503-509, 2005.

P. Skov-olsen, S. Boesby, P. Kirkegaard, K. Therkelsen, T. Almdal et al., Influence of epidermal growth factor on liver regeneration after partial hepatectomy in rats, Hepatology, vol.8, pp.992-998

M. Lesurtel, R. Graf, B. Aleil, D. J. Walther, Y. Tian et al., Platelet-derived serotonin mediates liver regeneration, Science (80-. ), vol.312, pp.104-107, 2006.

J. S. Campbell, L. Prichard, F. Schaper, J. Schmitz, A. Stephenson-famy et al., Expression of suppressors of cytokine signaling during liver regeneration, J. Clin. Invest, vol.107, pp.1285-92, 2001.

T. Ichikawa, Y. Q. Zhang, K. Kogure, Y. Hasegawa, H. Takagi et al., Transforming growth factor beta and activin tonically inhibit DNA synthesis in the rat liver, Hepatology, vol.34, pp.918-943, 2001.

R. Manco, I. A. Leclercq, and L. A. Clerbaux, Liver regeneration: Different sub-populations of parenchymal cells at play choreographed by an injury-specific microenvironment, Int. J. Mol. Sci, vol.19, 2018.

R. E. Billingham, L. Brent, and P. B. Medawar, Actively acquired tolerance of foreign cells, Nature, vol.172, pp.603-609, 1953.

R. Y. Calne, R. A. Sells, J. R. Pena, D. R. Davis, P. R. Millard et al., Induction of immunological tolerance by porcine liver allografts, Nature, vol.223, pp.472-478, 1969.

A. Sugioka, M. Morita, J. Fujita, A. Hasumi, and T. Shiroishi, Graft acceptance and tolerance induction in mouse liver transplantation using wild mice, Transplant. Proc, vol.33, pp.137-146

N. Kamada, H. S. Davies, and B. Roser, Reversal of transplantation immunity by liver grafting, Nature, vol.292, pp.840-842, 1981.

N. Kamada, H. S. Davies, D. Wight, L. Culank, and B. Roser, Liver transplantation in the rat. Biochemical and histological evidence of complete tolerance induction in non-rejector strains, Transplantation, vol.35, pp.304-315, 1983.

E. Kobayashi, N. Kamada, S. Enosawa, N. Toyama, N. I. Walker et al., Prevention by liver transplantation of the graft-versus-host reaction and allograft rejection in a rat model of small bowel transplantation, Transplantation, vol.57, pp.177-81, 1994.

Y. Sugawara, M. Makuuchi, J. Kaneko, Y. Kishi, S. Hata et al., Positive T lymphocytotoxic cross-match in living donor liver transplantation, Liver Transpl, vol.9, pp.1062-1068, 2003.

X. Lan, Impact of human leukocyte antigen mismatching on outcomes of liver transplantation: A meta-analysis, World J. Gastroenterol, vol.16, p.3457, 2010.

D. Bello, A. , C. , N. Danjoux, M. Muscari et al., De novo donor-specific anti-HLA antibodies mediated rejection in liver-transplant patients, Transpl. Int, vol.28, 2015.

D. Bello, A. , C. , N. Muscari, F. Lavayssière et al., Prevalence, incidence and risk factors for donor-specific anti-HLA antibodies in maintenance liver transplant patients, Am. J. Transplant, vol.14, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01915082

S. Feng and J. Bucuvalas, Tolerance after liver transplantation: Where are we? Liver Transplant, vol.23, pp.1601-1614, 2017.

A. J. Demetris, C. Bellamy, C. R. Gandhi, S. Prost, Y. Nakanuma et al., Functional Immune Anatomy of the Liver-As an Allograft, Am. J. Transplant, vol.16, pp.1653-80, 2016.

A. Albillos, A. Gottardi, . De, and M. Rescigno, The gut-liver axis in liver disease: pathophysiological basis for therapy, J. Hepatol, 2019.

Y. Kurashima and H. Kiyono, Mucosal Ecological Network of Epithelium and Immune Cells for Gut Homeostasis and Tissue Healing, Annu. Rev. Immunol, vol.35, pp.119-147, 2017.

S. I. Sayin, A. Wahlström, J. Felin, S. Jäntti, H. Marschall et al., Gut Microbiota Regulates Bile Acid Metabolism by Reducing the Levels of Tauro-beta-muricholic Acid, a Naturally Occurring FXR Antagonist, Cell Metab, vol.17, pp.225-235, 2013.

A. Wahlström, S. I. Sayin, H. U. Marschall, and F. Bäckhed, Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism, Cell Metab, vol.24, pp.41-50, 2016.

M. W. Robinson, C. Harmon, O. 'farrelly, and C. , Liver immunology and its role in inflammation and homeostasis, Cell. Mol. Immunol, vol.13, pp.267-276, 2016.

S. Bekkering, R. Arts, B. Novakovic, I. Kourtzelis, C. Van-der-heijden et al., Metabolic Induction of Trained Immunity through the Mevalonate Pathway, Cell, vol.172, pp.135-146, 2018.

L. Barbier, M. Ferhat, E. Salamé, A. Robin, A. Herbelin et al., Interleukin-1 Family Cytokines: Keystones in Liver Inflammatory Diseases, Front. Immunol, vol.10, 2014.

A. Rubartelli, M. T. Lotze, E. Latz, and A. Manfredi, Mechanisms of sterile inflammation, Front. Immunol, vol.4, p.398, 2013.

P. Kubes and W. Z. Mehal, Sterile inflammation in the liver, Gastroenterology, vol.143, pp.1158-1172, 2012.

B. Mcdonald and P. Kubes, Innate Immune Cell Trafficking and Function During Sterile Inflammation of the Liver, Gastroenterology, vol.151, pp.1087-1095, 2016.

M. Hossain and P. Kubes, Innate immune cells orchestrate the repair of sterile injury in the liver and beyond, Eur. J. Immunol, vol.49, pp.831-841, 2019.

Y. Zhai, H. Petrowsky, J. C. Hong, R. W. Busuttil, and J. W. Kupiec-weglinski, Ischaemia-reperfusion injury in liver transplantation--from bench to bedside, Nat. Rev. Gastroenterol. Hepatol, vol.10, pp.79-89, 2013.

R. F. Van-golen, T. M. Van-gulik, and M. Heger, The sterile immune response during hepatic ischemia/reperfusion, Cytokine Growth Factor Rev, vol.23, pp.69-84, 2012.

J. J. Oppenheim and D. Yang, Alarmins: Chemotactic activators of immune responses, Curr. Opin. Immunol, vol.17, pp.359-365, 2005.

L. Lu, H. Zhou, M. Ni, X. Wang, R. Busuttil et al., Innate Immune Regulations and Liver Ischemia-Reperfusion Injury, Transplantation, vol.100, pp.2601-2610, 2016.

E. B. Thorgersen, A. Barratt-due, H. Haugaa, M. Harboe, S. E. Pischke et al., The Role of Complement in Liver Injury, Regeneration, and Transplantation, Hepatology, vol.70, pp.725-736, 2019.

M. Bonneville, R. L. O'brien, and W. K. Born, Gammadelta T cell effector functions: a blend of innate programming and acquired plasticity, Nat. Rev. Immunol, vol.10, pp.467-78, 2010.

J. Turner, C. Rickassel, H. Healy, and A. J. Kassianos, Natural Killer Cells in Kidney Health and Disease, Front. Immunol, vol.10, p.587, 2019.

M. Ferhat, A. Robin, S. Giraud, S. Sena, J. Goujon et al., Endogenous IL-33 Contributes to Kidney Ischemia-Reperfusion Injury as an Alarmin, J. Am. Soc. Nephrol, vol.29, pp.1272-1288, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01929544

E. S. Baekkevold, M. Roussigné, T. Yamanaka, F. Johansen, F. L. Jahnsen et al., Molecular characterization of NF-HEV, a nuclear factor preferentially expressed in human high endothelial venules, Am. J. Pathol, vol.163, pp.69-79, 2003.

J. Schmitz, A. Owyang, E. Oldham, Y. Song, E. Murphy et al., IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines, Immunity, vol.23, pp.479-90, 2005.

C. Cayrol and J. Girard, Interleukin-33 (IL-33): A nuclear cytokine from the IL-1 family, Immunol. Rev, vol.281, pp.154-168, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02363721

G. Bandara, M. A. Beaven, A. Olivera, A. M. Gilfillan, and D. D. Metcalfe, Activated mast cells synthesize and release soluble ST2-a decoy receptor for IL-33, Eur. J. Immunol, vol.45, pp.3034-3078, 2015.

F. Y. Liew, J. P. Girard, and H. R. Turnquist, Interleukin-33 in health and disease, Nat. Rev. Immunol, vol.16, pp.676-689, 2016.

C. Moussion, N. Ortega, and J. Girard, The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel "alarmin, PLoS One, vol.3, p.3331, 2008.

C. Michaudel, C. Mackowiak, I. Maillet, L. Fauconnier, C. A. Akdis et al., Ozone exposure induces respiratory barrier biphasic injury and inflammation controlled by IL-33, J. Allergy Clin. Immunol, vol.142, pp.942-958, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02126403

S. P. Gadani, J. T. Walsh, I. Smirnov, J. Zheng, and J. Kipnis, The glia-derived alarmin IL-33 orchestrates the immune response and promotes recovery following CNS injury, Neuron, vol.85, pp.703-712, 2015.

C. Shimokawa, T. Kanaya, M. Hachisuka, K. Ishiwata, H. Hisaeda et al., Mast Cells Are Crucial for Induction of Group 2 Innate Lymphoid Cells and Clearance of Helminth Infections, Immunity, vol.46, pp.863-874, 2017.

A. B. Molofsky, A. K. Savage, and R. M. Locksley, Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation, Immunity, vol.42, pp.1005-1024, 2015.

N. E. Humphreys, D. Xu, M. R. Hepworth, F. Y. Liew, and R. K. Grencis, IL-33, a potent inducer of adaptive immunity to intestinal nematodes, J. Immunol, vol.180, pp.2443-2452, 2008.

O. Rostan, J. Gangneux, C. Piquet-pellorce, C. Manuel, A. Mckenzie et al., The IL-33/ST2 axis is associated with human visceral leishmaniasis and suppresses Th1 responses in the livers of BALB/c mice infected with Leishmania donovani, MBio, vol.4, pp.383-396, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00871493

E. Ayimba, J. Hegewald, A. Y. Ségbéna, R. G. Gantin, C. J. Lechner et al., Proinflammatory and regulatory cytokines and chemokines in infants with uncomplicated and severe Plasmodium falciparum malaria, Clin. Exp. Immunol, vol.166, pp.218-244, 2011.

L. A. Jones, F. Roberts, M. B. Nickdel, F. Brombacher, A. Mckenzie et al., IL-33 receptor (T1/ST2) signalling is necessary to prevent the development of encephalitis in mice infected with Toxoplasma gondii, Eur. J. Immunol, vol.40, pp.426-462, 2010.

K. J. Baker, A. Houston, and E. Brint, IL-1 Family Members in Cancer; Two Sides to Every Story, Front. Immunol, vol.10, p.1197, 2019.

Z. Yang, D. Ling, Y. Xie, W. Wu, J. Li et al., The Association of Serum IL-33 and sST2 with Breast Cancer, Dis. Markers, p.516895, 2015.

C. Wang, Z. Chen, X. Bu, Y. Han, S. Shan et al., IL-33 signaling fuels outgrowth and metastasis of human lung cancer, Biochem. Biophys. Res. Commun, vol.479, pp.461-468, 2016.

X. Tong, M. Barbour, K. Hou, C. Gao, S. Cao et al., Interleukin-33 predicts poor prognosis and promotes ovarian cancer cell growth and metastasis through regulating ERK and JNK signaling pathways, Mol. Oncol, vol.10, pp.113-138, 2016.

R. L. Maywald, S. K. Doerner, L. Pastorelli, D. Salvo, C. Benton et al., IL-33 activates tumor stroma to promote intestinal polyposis, Proc. Natl. Acad. Sci. U. S. A, vol.112, pp.2487-96, 2015.

N. L. Millar, C. O'donnell, I. B. Mcinnes, and E. Brint, Wounds that heal and wounds that don't -The role of the IL-33/ST2 pathway in tissue repair and tumorigenesis, Semin. Cell Dev. Biol, vol.61, pp.41-50, 2017.

L. Wang, H. Li, F. Liang, Y. Hong, S. Jiang et al., Examining IL-33 expression in the cervix of HPV-infected patients: a preliminary study comparing IL-33 levels in different stages of disease and analyzing its potential association with IFN-?, Med. Oncol, vol.31, pp.143-147, 2014.

C. Musolino, A. A. Profita, M. Alonci, A. Saitta, S. Russo et al., Reduced IL-33 plasma levels in multiple myeloma patients are associated with more advanced stage of disease, Br. J. Haematol, vol.160, pp.709-719, 2013.

D. Dominguez, C. Ye, Z. Geng, S. Chen, J. Fan et al., Exogenous IL-33 Restores Dendritic Cell Activation and Maturation in Established Cancer, J. Immunol, vol.198, pp.1365-1375, 2017.

V. Lucarini, G. Ziccheddu, I. Macchia, L. Sorsa, V. Peschiaroli et al., IL-33 restricts tumor growth and inhibits pulmonary metastasis in melanoma-bearing mice through eosinophils, Oncoimmunology, vol.6, p.1317420, 2017.

Y. Xia, T. Ohno, N. Nishii, A. Bhingare, H. Tachinami et al., Endogenous IL-33 exerts CD8+ T cell antitumor responses overcoming pro-tumor effects by regulatory T cells in a colon carcinoma model, Biochem. Biophys. Res. Commun, vol.518, pp.331-336, 2019.

H. O. Yazdani, H. Chen, S. Tohme, S. Tai, D. J. Van-der-windt et al., IL-33 exacerbates liver sterile inflammation by amplifying neutrophil extracellular trap formation, J. Hepatol, vol.68, pp.130-139, 2017.

N. Sakai, H. L. Van-sweringen, R. C. Quillin, R. Schuster, J. Blanchard et al., Interleukin-33 is hepatoprotective during liver ischemia/reperfusion in mice, Hepatology, vol.56, pp.1468-78, 2012.

N. M. Heller, R. Berga-bolanos, L. Naler, and J. M. Sen, Natural Killer T (NKT) Cells in Mice and Men, Signaling Mechanisms Regulating T Cell Diversity and Function, pp.119-146, 2017.

K. J. Wood, C. Mariat, O. Thaunat, C. Mousson, and G. Rifle, Bridging innate with adaptive immunity in transplantation: Triggers, sensors, and modulators, Transplantation, vol.98, pp.1021-1024, 2014.

P. J. Brennan, M. Brigl, and M. B. Brenner, Invariant natural killer T cells: An innate activation scheme linked to diverse effector functions, Nat. Rev. Immunol, vol.13, pp.101-117, 2013.

P. X. Liew, W. Lee, and P. Kubes, iNKT Cells Orchestrate a Switch from Inflammation to Resolution of Sterile Liver Injury, Immunity, vol.47, pp.752-765, 2017.

D. I. Godfrey, S. Stankovic, and A. G. Baxter, Raising the NKT cell family, Nat. Immunol, vol.11, pp.197-206, 2010.

T. M. Hill, J. S. Bezbradica, L. Van-kaer, and J. S. , CD1d-Restricted Natural Killer T Cells, In: eLS. Chichester, pp.1-27, 2016.

B. Gao, S. Radaeva, and O. Park, Liver natural killer and natural killer T cells: immunobiology and emerging roles in liver diseases, J. Leukoc. Biol, vol.86, pp.513-541, 2009.

C. Liao, M. I. Zimmer, and C. Wang, The functions of type I and type II natural killer T cells in inflammatory bowel diseases, Inflamm. Bowel Dis, vol.19, pp.1330-1338, 2013.

C. Agrati, F. Martini, C. Nisii, A. Oliva, D. Offizi et al., CD1d expression by hepatocytes is a main restriction element for intrahepatic T-cell recognition, J. Biol. Regul. Homeost. Agents, vol.19, pp.41-49

S. Fujii, K. Shimizu, C. Smith, L. Bonifaz, and R. M. Steinman, Activation of natural killer T cells by alpha-galactosylceramide rapidly induces the full maturation of dendritic cells in vivo and thereby acts as an adjuvant for combined CD4 and CD8 T cell immunity to a coadministered protein, J. Exp. Med, vol.198, pp.267-79, 2003.

C. Carnaud, D. Lee, O. Donnars, S. H. Park, A. Beavis et al., Cutting edge: Cross-talk between cells of the innate immune system: NKT cells rapidly activate NK cells, J. Immunol, vol.163, pp.4647-50, 1999.

J. M. Coquet, S. Chakravarti, K. Kyparissoudis, F. W. Mcnab, L. A. Pitt et al., Diverse cytokine production by NKT cell subsets and identification of an IL-17-producing CD4-NK1.1-NKT cell population, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.11287-92, 2008.

C. M. Lappas, Y. Day, M. A. Marshall, V. H. Engelhard, and J. Linden, Adenosine A2A receptor 148 activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation, J. Exp. Med, vol.203, pp.2639-2687, 2006.

H. Matsuda, K. Takeda, T. Koya, M. Okamoto, Y. Shiraishi et al., Plasticity of invariant NKT cell regulation of allergic airway disease is dependent on IFN-gamma production, J. Immunol, vol.185, pp.253-62, 2010.

E. Y. Kim, J. T. Battaile, A. C. Patel, Y. You, E. Agapov et al., Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease, Nat. Med, vol.14, pp.633-673, 2008.

O. Akbari, P. Stock, E. Meyer, M. Kronenberg, S. Sidobre et al., Essential role of NKT cells producing IL-4 and IL-13 in the development of allergen-induced airway hyperreactivity, Nat. Med, vol.9, pp.582-590, 2003.

G. Bricard, V. Cesson, E. Devevre, H. Bouzourene, C. Barbey et al., Enrichment of human CD4+ V(alpha)24/Vbeta11 invariant NKT cells in intrahepatic malignant tumors, J. Immunol, vol.182, pp.5140-51, 2009.

A. Waddell, J. Zhao, and M. T. Cantorna, NKT cells can help mediate the protective effects of 1,25-dihydroxyvitamin D3 in experimental autoimmune encephalomyelitis in mice, Int. Immunol, vol.27, pp.237-281, 2015.

D. Chen, H. Zhao, X. Gao, S. Chen, H. Liu et al., Subcutaneous administration of ?-GalCer activates iNKT10 cells to promote M2 macrophage polarization and ameliorates chronic inflammation of obese adipose tissue, Int. Immunopharmacol, vol.77, p.105948, 2019.

S. Zhu, H. Zhang, and L. Bai, NKT cells in liver diseases, Front. Med, vol.12, pp.249-261, 2018.

Y. Kinjo, E. Tupin, D. Wu, M. Fujio, R. Garcia-navarro et al., Natural killer T cells recognize diacylglycerol antigens from pathogenic bacteria, Nat. Immunol, vol.7, pp.978-86, 2006.

K. Kawakami, N. Yamamoto, Y. Kinjo, K. Miyagi, C. Nakasone et al., Critical role of Valpha14+ natural killer T cells in the innate phase of host protection against Streptococcus pneumoniae infection, Eur. J. Immunol, vol.33, pp.3322-3352, 2003.

S. Fujii, K. Shimizu, Y. Okamoto, N. Kunii, T. Nakayama et al., NKT cells as an ideal anti-tumor immunotherapeutic, Front. Immunol, vol.4, p.409, 2013.

N. A. Braun, R. Covarrubias, and A. S. Major, Natural killer T cells and atherosclerosis: form and function meet pathogenesis, J. Innate Immun, vol.2, pp.316-340, 2010.

J. Mattner, Natural killer T (NKT) cells in autoimmune hepatitis, Curr. Opin. Immunol, vol.25, pp.697-703, 2013.

M. Lisbonne, L. 'helgoualc'h, A. Nauwelaers, G. Turlin, B. Lucas et al., Invariant natural killer T-cell-deficient mice display increased CCl? -induced hepatitis associated with CXCL1 over-expression and neutrophil infiltration, Eur. J. Immunol, vol.41, pp.1720-1752, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00597242

M. H. Ferhat, A. Robin, L. Barbier, A. Thierry, J. Gombert et al., The Impact of Invariant NKT Cells in Sterile Inflammation: The Possible Contribution of the Alarmin/Cytokine IL-33, Front. Immunol, vol.9, p.2308, 2018.

S. Kuboki, N. Sakai, J. Tschop, M. J. Edwards, A. B. Lentsch et al., Distinct contributions of CD4+ T cell subsets in hepatic ischemia/reperfusion injury, Am. J. Physiol. -Gastrointest. Liver Physiol, vol.296, pp.1054-1063, 2009.

P. Arrenberg, I. Maricic, and V. Kumar, Sulfatide-mediated activation of type II natural killer T cells prevents hepatic ischemic reperfusion injury in mice, Gastroenterology, vol.140, pp.646-55, 2011.

Z. Cao, Y. Yuan, G. Jeyabalan, Q. Du, A. Tsung et al., Preactivation of NKT cells with alpha-GalCer protects against hepatic ischemia-reperfusion injury in mouse by a mechanism involving IL-13 and adenosine A2A receptor, Am. J. Physiol. Gastrointest. liver Physiol, vol.297, pp.249-58, 2009.

E. Bourgeois, L. P. Van, M. Samson, S. Diem, A. Barra et al., The pro-Th2 cytokine, p.149
URL : https://hal.archives-ouvertes.fr/hal-00356489

, directly interacts with invariant NKT and NK cells to induce IFN-gamma production, Eur. J. Immunol, vol.39, pp.1046-55, 2009.

A. Thierry, S. Giraud, A. Robin, A. Barra, F. Bridoux et al., The Alarmin Concept Applied to Human Renal Transplantation: Evidence for a Differential Implication of HMGB1 and IL-33, PLoS One, vol.9, p.88742, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01928093

Y. Abe, I. N. Hines, G. Zibari, K. Pavlick, L. Gray et al., Mouse Model of Liver Ischemia and Reperfusion Injury: Method to Study Reactive Oxygen and Nitrogen Metabolites in vivo, Nat. Med, vol.46, pp.1-7, 2009.

H. Yin, B. Huang, H. Yang, Y. Huang, P. Xiong et al., Pretreatment with soluble ST2 reduces warm hepatic ischemia/reperfusion injury, Biochem. Biophys. Res. Commun, vol.351, pp.940-946, 2006.

N. Sakai, H. L. Van-sweringen, R. M. Belizaire, R. C. Quillin, R. Schuster et al., Interleukin-37 reduces liver inflammatory injury via effects on hepatocytes and nonparenchymal cells, J. Gastroenterol. Hepatol, vol.27, pp.1609-1625, 2012.

G. Beldi, Y. Banz, A. Kroemer, X. Sun, Y. Wu et al., Deletion of CD39 on natural killer cells attenuates hepatic ischemia/reperfusion injury in mice, Hepatology, vol.51, pp.1702-1713, 2010.

J. G. Brockmann, C. August, H. H. Wolters, R. Hömme, D. Palmes et al., Sequence of reperfusion influences ischemia/reperfusion injury and primary graft function following porcine liver transplantation, Liver Transplant, vol.11, pp.1214-1222, 2005.

Q. Liu, K. Vekemans, L. Iania, M. Komuta, J. Parkkinen et al., Assessing warm ischemic injury of pig livers at hypothermic machine perfusion, J. Surg. Res, vol.186, pp.379-89, 2014.

S. Suzuki, L. H. Toledo-pereyra, F. J. Rodriguez, and D. Cejalvo, Neutrophil infiltration as an important factor in liver ischemia and reperfusion injury. Modulating effects of FK506 and cyclosporine, Transplantation, vol.55, pp.1265-72, 1993.

N. Shahbazi, H. Haeri, N. Toosi, M. Jafarian, A. Shahsiah et al., Correlation of Histopathologic Findings of Non-Graft Threatening Preservation/Reperfusion Injury in Time-Zero Liver Needle Biopsies With Short-Term Post-transplantation Laboratory Alterations, Hepat. Mon, vol.15, pp.4-9, 2015.

D. A. Neil and S. G. Hubscher, Are parenchymal changes in early post-transplant biopsies related to preservation-reperfusion injury or rejection?, Transplantation, vol.71, pp.1566-72, 2001.

L. Ricca, A. Lemoine, F. Cauchy, J. Hamelin, M. Sebagh et al., Ischemic Postconditioning of the Liver Graft in Adult Liver Transplantation, Transplantation, vol.99, pp.1633-1676, 2015.

T. Senoner, S. Schindler, S. Stättner, D. Öfner, J. Troppmair et al., Associations of Oxidative Stress and Postoperative Outcome in Liver Surgery with an Outlook to Future Potential Therapeutic Options, Oxid. Med. Cell. Longev, 2019.

Y. Li, D. Ma, Z. Wang, and J. Yang, MicroRNA-155 Deficiency in Kupffer Cells Ameliorates Liver Ischemia-Reperfusion Injury in Mice, Transplantation, vol.101, pp.1600-1608, 2017.

H. Bai, J. Wen, J. Gong, H. Wu, F. Yuan et al., Blockade of the Notch1/Jagged1 pathway in Kupffer cells aggravates ischemia-reperfusion injury of orthotopic liver transplantation in mice, Autoimmunity, vol.52, pp.176-184, 2019.

P. Marvie, M. Lisbonne, L. 'helgoualc'h, A. Rauch, M. Turlin et al., Interleukin-33 overexpression is associated with liver fibrosis in mice and humans, J. Cell. Mol. Med, vol.14, pp.1726-1765, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00660257

A. B. Al-khafaji, S. Tohme, H. O. Yazdani, D. Miller, H. Huang et al., Superoxide induces neutrophil extracellular trap formation in a TLR-4 and NOX-dependent mechanism, Mol, p.150

. Med, , vol.22, pp.621-631, 2016.

O. Strauss, A. Phillips, K. Ruggiero, A. Bartlett, and P. R. Dunbar, Immunofluorescence identifies distinct subsets of endothelial cells in the human liver, Sci. Rep, vol.7, p.44356, 2017.

M. I. Arshad, C. Piquet-pellorce, L. 'helgoualc'h, A. Rauch, M. Patrat-delon et al., TRAIL but not FasL and TNF?, regulates IL-33 expression in murine hepatocytes during acute hepatitis, Hepatology, vol.56, pp.2353-2362, 2012.

C. Moussion, N. Ortega, and J. P. Girard, The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: A novel "Alarmin, PLoS One, vol.3, pp.1-8, 2008.

M. Cottagiri, M. Nyandjo, M. Stephens, J. Mantilla, H. Saito et al., In drug-induced, immune-mediated hepatitis, interleukin-33 reduces hepatitis and improves survival independently and as a consequence of FoxP3+ T-cell activity, Cell. Mol. Immunol, 2018.

M. I. Arshad, M. Rauch, L. 'helgoualc'h, A. , J. V. Leite-de-moraes et al., NKT cells are required to induce high IL-33 expression in hepatocytes during ConA-induced acute hepatitis, Eur. J. Immunol, vol.41, pp.2341-2348, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00681990

R. Fahrner, F. Dondorf, M. Ardelt, U. Settmacher, and F. Rauchfuss, Role of NK, NKT cells and macrophages in liver transplantation, World J. Gastroenterol, vol.22, pp.6135-6144, 2016.

V. Menkin, Studies on the isolation of the factor responsible for tissue injury in inflammation, Science, vol.97, pp.165-172, 1943.

P. B. Beeson, Temperature-elevating effect of a substance obtained from polymorphonuclear leucocytes, J Clin Invest, vol.27, p.524, 1948.

E. Atkins and W. B. Wood, Studies on the pathogenesis of fever. II. Identification of an endogenous pyrogen in the blood stream following the injection of typhoid vaccine, J Exp Med, vol.102, pp.499-516, 1955.

J. M. Dayer, From supernatants to cytokines: a personal view on the early history of IL-1, IL-1Ra, TNF and its inhibitor in rheumatology, Arthritis Res Ther, vol.20, pp.1-6, 2018.

C. A. Dinarello, Interleukin-1, Cytokine Growth Factor Rev, vol.8, pp.253-65, 1997.
URL : https://hal.archives-ouvertes.fr/pasteur-01721887

C. A. Dinarello, IL-1: discoveries, controversies and future directions, Eur J Immunol, vol.40, pp.599-606, 2010.

H. Tsutsui, X. Cai, and S. Hayashi, Interleukin-1 family cytokines in liver diseases, Mediators Inflamm, p.630265, 2015.

C. A. Dinarello, Overview of the IL-1 family in innate inflammation and acquired immunity, Immunol Rev, vol.281, pp.8-27, 2018.

A. Mantovani, C. A. Dinarello, M. Molgora, and C. Garlanda, Interleukin-1 and related cytokines in the regulation of inflammation and immunity, Immunity, vol.50, pp.778-95, 2019.

M. Molgora, E. Bonavita, A. Ponzetta, F. Riva, M. Barbagallo et al., IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity, Nature, vol.551, pp.110-114, 2017.

M. Molgora, D. Supino, A. Mantovani, and C. Garlanda, Tuning inflammation and immunity by the negative regulators IL-1R2 and IL-1R8, Immunol Rev, vol.281, pp.233-280, 2018.

C. Moussion, N. Ortega, and J. P. Girard, The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel 'alarmin'?, PLoS ONE, vol.3, pp.1-8, 2008.

C. Cayrol and J. Girard, Interleukin-33 (IL-33): a nuclear cytokine from the IL-1 family, Immunol Rev, vol.281, pp.154-68, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02363721

G. Bandara, M. A. Beaven, A. Olivera, A. M. Gilfillan, and D. D. Metcalfe, Activated mast cells synthesize and release soluble ST2-a decoy receptor for IL-33, Eur J Immunol, vol.45, pp.3034-3078, 2015.

H. Hayakawa, M. Hayakawa, A. Kume, and S. Tominaga, Soluble ST2 blocks interleukin-33 signaling in allergic airway inflammation, J Biol Chem, vol.282, pp.26369-80, 2007.

S. Sanada, D. Hakuno, L. J. Higgins, E. R. Schreiter, A. Mckenzie et al., IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system, J Clin Invest, vol.117, pp.1538-1587, 2007.

J. Bessa, C. A. Meyer, M. C. De-vera-mudry, S. Schlicht, S. H. Smith et al., Altered subcellular localization of IL-33 leads to non-resolving lethal inflammation, J Autoimmun, vol.55, pp.33-41, 2014.

I. Cohen, P. Rider, Y. Carmi, A. Braiman, S. Dotan et al., Differential release of chromatin-bound IL-1alpha discriminates between necrotic and apoptotic cell death by the ability to induce sterile inflammation, Proc Natl Acad Sci, vol.107, pp.2574-2583, 2010.

S. Bekkering, R. Arts, B. Novakovic, I. Kourtzelis, C. Van-der-heijden et al., Metabolic induction of trained immunity through the mevalonate pathway, Cell, vol.172, pp.135-181, 2018.

I. Mitroulis, K. Ruppova, B. Wang, L. Chen, M. Grzybek et al., Modulation of myelopoiesis progenitors is an integral component of trained immunity, Cell, vol.172, pp.147-61, 2018.

S. Moorlag, R. J. Röring, L. Joosten, and M. G. Netea, The role of the interleukin-1 family in trained immunity, Immunol Rev, vol.281, pp.28-39, 2018.

J. Dupaul-chicoine, A. Arabzadeh, M. Dagenais, T. Douglas, C. Champagne et al., The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity, Immunity, vol.43, pp.751-63, 2015.

N. M. Adams, T. E. O'sullivan, C. D. Geary, J. M. Karo, R. A. Amezquita et al., NK cell responses redefine immunological memory, J Immunol, vol.197, pp.2963-70, 2016.

S. Madera and J. C. Sun, Cutting edge: stage-specific requirement of IL-18 for antiviral NK cell expansion, J Immunol, vol.194, pp.1408-1420, 2015.

T. Nabekura, J. Girard, and L. L. Lanier, IL-33 receptor ST2 amplifies the expansion of NK cells and enhances host defense during mouse cytomegalovirus infection, J Immunol, vol.194, pp.5948-52, 2015.

R. Romee, S. E. Schneider, J. W. Leong, J. M. Chase, C. R. Keppel et al., Cytokine activation induces human memory-like NK cells, Blood, vol.120, pp.4751-60, 2012.

X. Huang, L. D. Hazlett, W. Du, and R. P. Barrett, SIGIRR promotes resistance against Pseudomonas aeruginosa keratitis by down-regulating type-1 immunity and IL-1R1 and TLR4 signaling, J Immunol, vol.177, pp.548-56, 2006.

T. Véliz-rodriguez, F. Moalli, N. Polentarutti, M. Paroni, E. Bonavita et al., Role of Toll interleukin-1 receptor (IL-1R) 8, a negative regulator of IL-1R/Toll-like receptor signaling, in resistance to acute Pseudomonas aeruginosa lung infection, Infect Immun, vol.80, pp.100-109, 2012.

L. F. Campesato, A. Silva, L. Cordeiro, B. R. Correa, F. Navarro et al., High IL-1R8 expression in breast tumors promotes tumor growth and contributes to impaired antitumor immunity, Oncotarget, vol.8, pp.49470-83, 2017.

M. Bachmann, J. Pfeilschifter, and H. Mühl, A prominent role of interleukin-18 in acetaminophen-induced liver injury advocates its blockage for therapy of hepatic necroinflammation, Front Immunol, vol.9, pp.1-7, 2018.

S. Belkaya, E. Michailidis, C. B. Korol, M. Kabbani, A. Cobat et al., Inherited IL-18BP deficiency in human fulminant viral hepatitis, J Exp Med, vol.216, pp.1777-90, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02359432

C. L. Langrish, Y. Chen, W. M. Blumenschein, J. Mattson, B. Basham et al., IL-23 drives a pathogenic T cell population that induces autoimmune inflammation, J Exp Med, vol.201, pp.233-273, 2005.

K. Mills, L. S. Dungan, S. A. Jones, and J. Harris, The role of inflammasomederived IL-1 in driving IL-17 responses, J Leukoc Biol, vol.93, pp.489-97, 2013.

C. Sutton, C. Brereton, B. Keogh, K. Mills, and E. C. Lavelle, A crucial role for interleukin (IL)-1 in the induction of IL-17-producing T cells that mediate autoimmune encephalomyelitis, J Exp Med, vol.203, pp.1685-91, 2006.

C. E. Sutton, S. J. Lalor, C. M. Sweeney, C. F. Brereton, E. C. Lavelle et al., Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity, Immunity, vol.31, pp.331-372, 2009.

N. Arpaia, J. A. Green, B. Moltedo, A. Arvey, S. Hemmers et al., A distinct function of regulatory T cells in tissue protection, Cell, vol.162, pp.1078-89, 2015.

L. A. Monticelli, L. C. Osborne, M. Noti, S. V. Tran, D. Zaiss et al., IL-33 promotes an innate immune pathway of intestinal tissue protection dependent on amphiregulin-EGFR interactions, Proc Natl Acad Sci, vol.112, pp.10762-10769, 2015.

E. Bourgeois, L. P. Van, M. Samson, S. Diem, A. Barra et al., The pro-Th2 cytokine IL-33 directly interacts with invariant NKT and NK cells to induce IFN-gamma production, Eur J Immunol, vol.39, pp.1046-55, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00676402

M. D. Smithgall, M. R. Comeau, B. Yoon, D. Kaufman, R. Armitage et al., IL-33 amplifies both Th1-and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells, Int Immunol, vol.20, pp.1019-1049, 2008.

J. Kearley, J. S. Silver, C. Sanden, Z. Liu, A. A. Berlin et al., Cigarette smoke silences innate lymphoid cell function and facilitates an exacerbated type I interleukin-33-dependent response to infection, Immunity, vol.42, pp.566-79, 2015.

W. V. Bonilla, A. Fröhlich, K. Senn, S. Kallert, M. Fernandez et al., The alarmin interleukin-33 drives protective antiviral CD8+ T cell responses, Science, vol.335, pp.984-993, 2012.

E. A. Bourgeois, A. Levescot, S. Diem, A. Chauvineau, H. Bergès et al., A natural protective function of invariant NKT cells in a mouse model of innate-cell-driven lung inflammation, Eur J Immunol, vol.41, pp.299-305, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00539574

M. Ferhat, A. Robin, S. Giraud, S. Sena, J. Goujon et al., Endogenous IL-33 contributes to kidney ischemia-reperfusion injury as an alarmin, J Am Soc Nephrol, vol.29, pp.1272-88, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01929544

M. H. Ferhat, A. Robin, L. Barbier, A. Thierry, J. Gombert et al., The impact of invariant NKT cells in sterile inflammation: the possible contribution of the alarmin/cytokine IL-33, Front Immunol, vol.9, p.2308, 2018.

A. Thierry, S. Giraud, A. Robin, A. Barra, F. Bridoux et al., The alarmin concept applied to human renal transplantation: evidence for a differential implication of HMGB1 and IL-33, PLoS ONE, vol.9, p.88742, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01928093

V. Sabapathy, M. E. Stremska, S. Mohammad, R. L. Corey, P. R. Sharma et al., Novel immunomodulatory cytokine regulates inflammation, diabetes, and obesity to protect from diabetic nephropathy, Front Pharmacol, vol.10, p.572, 2019.

T. Mahlakõiv, A. Flamar, L. K. Johnston, S. Moriyama, G. G. Putzel et al., Stromal cells maintain immune cell homeostasis in adipose tissue via production of interleukin-33, Sci Immunol, vol.4, p.416, 2019.

R. G. Spallanzani, D. Zemmour, T. Xiao, T. Jayewickreme, C. Li et al., Distinct immunocyte-promoting and adipocyte-generating stromal components coordinate adipose tissue immune and metabolic tenors, Sci Immunol, vol.4, p.3658, 2019.

J. A. Green, N. Arpaia, M. Schizas, A. Dobrin, and A. Y. Rudensky, A nonimmune function of T cells in promoting lung tumor progression, J Exp Med, vol.214, pp.3565-75, 2017.

A. Christ, P. Günther, M. Lauterbach, P. Duewell, D. Biswas et al., Western diet triggers NLRP3-dependent innate immune reprogramming, Cell, vol.172, pp.162-75, 2018.

E. Kaufmann, J. Sanz, J. L. Dunn, N. Khan, L. E. Mendonça et al., BCG educates hematopoietic stem cells to generate protective innate immunity against tuberculosis, Cell, vol.172, pp.176-90, 2018.

M. I. Arshad, M. Rauch, L. 'helgoualc'h, A. , J. V. Leite-de-moraes et al., NKT cells are required to induce high IL-33 expression in hepatocytes during ConA-induced acute hepatitis, Eur J Immunol, vol.41, pp.2341-2349, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00681990

A. Matsukawa, T. Fukumoto, T. Maeda, S. Ohkawara, and M. Yoshinaga, Detection and characterization of IL-1 receptor antagonist in tissues from healthy rabbits: IL-1 receptor antagonist is probably involved in health, Cytokine, vol.9, pp.307-322, 1997.

C. A. Dinarello, Immunological and inflammatory functions of the interleukin-1 family, Annu Rev Immunol, vol.27, pp.519-50, 2009.

N. Sakai, H. L. Van-sweringen, R. C. Quillin, R. Schuster, J. Blanchard et al., Interleukin-33 is hepatoprotective during liver ischemia/reperfusion in mice, Hepatology, vol.56, pp.1468-78, 2012.

W. Bernal and J. Wendon, Acute liver failure, N Engl J Med, vol.369, pp.2525-2559, 2013.

D. Gotthardt, C. Riediger, K. H. Weiss, J. Encke, P. Schemmer et al., Fulminant hepatic failure: etiology and indications for liver transplantation, Nephrol Dial Transplant, pp.5-8, 2007.

T. Sakurai, G. He, A. Matsuzawa, G. Yu, S. Maeda et al., Hepatocyte necrosis induced by oxidative stress and IL-1? release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis, Cancer Cell, vol.14, pp.156-65, 2008.

C. Chen, H. Kono, D. Golenbock, G. Reed, S. Akira et al., Identification of a key pathway required for the sterile inflammatory response triggered by dying cells, Nat Med, vol.13, pp.851-857, 2007.

M. Sultan, Z. Ben-ari, R. Masoud, O. Pappo, D. Harats et al., Interleukin-1? and interleukin-1? play a central role in the pathogenesis of fulminant hepatic failure in mice, PLoS ONE, vol.12, p.184084, 2017.

C. Gabay, M. F. Smith, D. Eidlen, and W. P. Arend, Interleukin 1 receptor antagonist (IL-1Ra) is an acute-phase protein, J Clin Invest, vol.99, pp.2930-2970, 1997.

H. Tsutsui, N. Kayagaki, K. Kuida, H. Nakano, N. Hayashi et al., Caspase-1-independent, Fas/Fas ligand-mediated IL-18 secretion from macrophages causes acute liver injury in mice, Immunity, vol.11, pp.80111-80120, 1999.

C. Yan, L. Zhou, and Y. Han, Contribution of hepatic stellate cells and matrix metalloproteinase 9 in acute liver failure, Liver Int, vol.28, pp.959-71, 2008.

M. Shinoda, A. W. Tilles, N. Kobayashi, G. Wakabayashi, A. Takayanagi et al., A bioartificial liver device secreting interleukin-1 receptor antagonist for the treatment of hepatic failure in rats, J Surg Res, vol.137, pp.130-170, 2007.

B. Shakoory, J. A. Carcillo, W. W. Chatham, R. L. Amdur, H. Zhao et al., Interleukin-1 receptor blockade is associated with reduced mortality in sepsis patients with features of macrophage activation syndrome: reanalysis of a prior phase III trial *, Crit Care Med, vol.44, pp.275-81, 2016.

H. Tsutsui, K. Matsui, N. Kawada, Y. Hyodo, N. Hayashi et al., IL-18 accounts for both TNF-alpha-and Fas ligand-mediated hepatotoxic pathways in endotoxin-induced liver injury in mice, J Immunol, vol.159, pp.3961-3968, 1997.

S. Finotto, J. Siebler, M. Hausding, M. Schipp, S. Wirtz et al., Severe hepatic injury in interleukin 18 (IL-18) transgenic mice: a key role for IL-18 in regulating hepatocyte apoptosis in vivo, Gut, vol.53, pp.392-400, 2004.

A. B. Imaeda, A. Watanabe, M. A. Sohail, S. Mahmood, M. Mohamadnejad et al., Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9 and the Nalp3 inflammasome, J Clin Invest, vol.119, pp.305-319, 2009.

M. M. Antunes, A. M. Araújo, A. B. Diniz, R. Pereira, D. M. Alvarenga et al., IL-33 signalling in liver immune cells enhances druginduced liver injury and inflammation, Inflamm Res, vol.67, pp.77-88, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02126492

P. Scheiermann, M. Bachmann, L. Härdle, T. Pleli, A. Piiper et al., Application of IL-36 receptor antagonist weakens CCL20 expression and impairs recovery in the late phase of murine acetaminophen-induced liver injury, Sci Rep, vol.5, pp.1-7, 2015.

M. I. Arshad, S. Patrat-delon, C. Piquet-pellorce, L. 'helgoualc'h, A. Rauch et al., Pathogenic mouse hepatitis virus or poly(I:C) induce IL-33 in hepatocytes in murine models of hepatitis, PLoS ONE, vol.8, pp.1-10, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00874120

N. Kamo, B. Ke, A. A. Ghaffari, X. Shen, R. W. Busuttil et al., ASC/caspase-1/IL-1? signaling triggers inflammatory responses by promoting HMGB1 induction in liver ischemia/reperfusion injury, Hepatology, vol.58, pp.351-62, 2013.

H. Y. Kim, S. J. Kim, and S. M. Lee, Activation of NLRP3 and AIM2 inflammasomes in Kupffer cells in hepatic ischemia/reperfusion, FEBS J, vol.282, pp.259-70, 2015.

Y. Inoue, K. Shirasuna, H. Kimura, F. Usui, A. Kawashima et al., NLRP3 regulates neutrophil functions and contributes to hepatic ischemiareperfusion injury independently of inflammasomes, J Immunol, vol.192, pp.4342-51, 2014.

A. Sadatomo, Y. Inoue, H. Ito, T. Karasawa, H. Kimura et al., Interaction of neutrophils with macrophages promotes IL-1? maturation and contributes to hepatic ischemia-reperfusion injury, J Immunol, vol.199, pp.3306-3321, 2017.

H. O. Yazdani, H. Chen, S. Tohme, S. Tai, D. J. Van-der-windt et al., IL-33 exacerbates liver sterile inflammation by amplifying neutrophil extracellular trap formation, J Hepatol, vol.68, pp.130-139, 2017.

P. Arrenberg, I. Maricic, and V. Kumar, Sulfatide-mediated activation of type II natural killer T cells prevents hepatic ischemic reperfusion injury in mice, Gastroenterology, vol.140, pp.646-55, 2011.

N. Sakai, H. L. Van-sweringen, R. M. Belizaire, R. C. Quillin, R. Schuster et al., Interleukin-37 reduces liver inflammatory injury via effects on hepatocytes and non-parenchymal cells, J Gastroenterol Hepatol, vol.27, pp.1609-1625, 2012.

Y. Muto, K. T. Nouri-aria, A. Meager, G. J. Alexander, A. L. Eddleston et al., Enhanced tumour necrosis factor and interleukin-1 in fulminant hepatic failure, Lancet, vol.2, pp.90006-90008, 1988.

K. D. Sekiyama, M. Yoshiba, and A. W. Thomson, Circulating proinflammatory cytokines (IL-1 beta, TNF-alpha, and IL-6) and IL-1 receptor antagonist (IL-1Ra) in fulminant hepatic failure and acute hepatitis, Clin Exp Immunol, vol.98, pp.71-78, 1994.

G. A. Roth, P. Faybik, H. Hetz, S. Hacker, H. J. Ankersmit et al., Proinflammatory interleukin-18 and Caspase-1 serum levels in liver failure are unaffected by MARS treatment, Dig Liver Dis, vol.41, pp.417-440, 2009.

G. A. Roth, M. Zimmermann, B. A. Lubsczyk, J. Pilz, P. Faybik et al., Upregulation of interleukin 33 and soluble ST2 serum levels in liver failure, J Surg Res, vol.163, pp.79-83, 2010.

J. Kim, D. Chang, H. W. Lee, H. Lee, J. H. Kim et al., Innatelike cytotoxic function of bystander-activated CD8+ T cells is associated with liver injury in acute hepatitis A, Immunity, vol.48, pp.161-73, 2018.

A. Iracheta-vellve, J. Petrasek, A. Satishchandran, B. Gyongyosi, B. Saha et al., Inhibition of sterile danger signals, uric acid and ATP, prevents inflammasome activation and protects from alcoholic steatohepatitis in mice, J Hepatol, vol.63, pp.1147-55, 2015.

J. Petrasek, S. Bala, T. Csak, D. Lippai, K. Kodys et al., IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice, J Clin Invest, vol.122, pp.3476-89, 2012.

K. Cui, G. Yan, C. Xu, Y. Chen, J. Wang et al., Invariant NKT cells promote alcohol-induced steatohepatitis through interleukin-1? in mice, J Hepatol, vol.62, pp.1311-1319, 2015.

M. J. Heo, T. H. Kim, J. S. You, D. Blaya, P. Sancho-bru et al., Alcohol dysregulates miR-148a in hepatocytes through FoxO1, facilitating pyroptosis via TXNIP overexpression, Gut, vol.68, pp.708-728, 2019.

Z. Sun, B. Chang, A. Huang, S. Hao, M. Gao et al., Plasma levels of soluble ST2, but not IL-33, correlate with the severity of alcoholic liver disease, J Cell Mol Med, vol.23, pp.887-97, 2018.

M. Wang, G. Shen, L. Xu, X. Liu, J. M. Brown et al., IL-1 receptor like 1 protects against alcoholic liver injury by limiting NF-?B activation in hepatic macrophages, J Hepatol, vol.68, pp.109-126, 2018.

I. Maricic, H. Sheng, I. Marrero, E. Seki, T. Kisseleva et al., Inhibition of type I natural killer T cells by retinoids or following sulfatidemediated activation of type II natural killer T cells attenuates alcoholic liver disease in mice, Hepatology, vol.61, pp.1357-69, 2015.

S. Mathews, D. Feng, I. Maricic, C. Ju, V. Kumar et al., Invariant natural killer T cells contribute to chronic-plus-binge ethanol-mediated liver injury by promoting hepatic neutrophil infiltration, Cell Mol Immunol, vol.13, pp.206-222, 2016.

E. Bugianesi, C. Rosso, and H. Cortez-pinto, How to diagnose NAFLD in 2016, J Hepatol, vol.65, pp.643-647, 2016.

S. Bellentani, The epidemiology of non-alcoholic fatty liver disease, Liver Int, vol.37, pp.81-85, 2017.

, EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease, European Association for the Study of the Liver (EASL), vol.59, pp.1121-1161, 2016.

J. Yu, S. Marsh, J. Hu, W. Feng, and C. Wu, The pathogenesis of nonalcoholic fatty liver disease: interplay between diet, gut microbiota, and genetic background

, Gastroenterol Res Pract, p.2862173, 2016.

K. A. Negrin, R. Flach, M. T. Distefano, A. Matevossian, R. H. Friedline et al., IL-1 Signaling in obesity-induced hepatic lipogenesis and steatosis, PLoS ONE, vol.9, p.107265, 2014.

K. Miura, L. Yang, N. Van-rooijen, D. A. Brenner, H. Ohnishi et al., Toll-like receptor 2 and palmitic acid cooperatively contribute to the development of nonalcoholic steatohepatitis through inflammasome activation in mice, Hepatology, vol.57, pp.577-89, 2013.

T. Csak, M. Ganz, J. Pespisa, K. Kodys, A. Dolganiuc et al., Fatty acid and endotoxin activate inflammasomes in mouse hepatocytes that release danger signals to stimulate immune cells, Hepatology, vol.54, pp.133-177, 2011.

A. R. Moschen, C. Molnar, B. Enrich, S. Geiger, C. F. Ebenbichler et al., Adipose and liver expression of interleukin (IL)-1 family members in morbid obesity and effects of weight loss, Mol Med Camb Mass, vol.17, pp.840-845, 2011.

A. L. Patrick, J. Rullo, S. Beaudin, P. Liaw, and A. E. Fox-robichaud, Hepatic leukocyte recruitment in response to time-limited expression of TNF-? and IL-1?, Am J Physiol-Gastrointest Liver Physiol, vol.293, pp.663-72, 2007.

H. Tilg, A. R. Moschen, and G. Szabo, Interleukin-1 and inflammasomes in alcoholic liver disease/acute alcoholic hepatitis and nonalcoholic fatty liver disease/nonalcoholic steatohepatitis, Hepatology, vol.64, pp.955-65, 2016.

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

J. Jager, T. Grémeaux, M. Cormont, L. Marchand-brustel, Y. Tanti et al., Interleukin-1?-induced insulin resistance in adipocytes through downregulation of insulin receptor substrate-1 expression, Endocrinology, vol.148, pp.241-51, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00139856

O. Nov, A. Kohl, E. C. Lewis, N. Bashan, I. Dvir et al., Interleukin-1beta may mediate insulin resistance in liver-derived cells in response to adipocyte inflammation, Endocrinology, vol.151, pp.4247-56, 2010.

H. Wen, D. Gris, Y. Lei, S. Jha, L. Zhang et al., Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling, Nat Immunol, vol.12, pp.408-423, 2011.

T. Matsuki, R. Horai, K. Sudo, and Y. Iwakura, IL-1 plays an important role in lipid metabolism by regulating insulin levels under physiological conditions, J Exp Med, vol.198, pp.877-88, 2003.

K. Isoda, S. Sawada, M. Ayaori, T. Matsuki, R. Horai et al., Deficiency of interleukin-1 receptor antagonist deteriorates fatty liver and cholesterol metabolism in hypercholesterolemic mice, J Biol Chem, vol.280, pp.7002-7011, 2005.

J. Henao-mejia, E. Elinav, J. C. Hao, L. Mehal, W. Z. Strowig et al., Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity, Nature, vol.482, pp.179-85, 2012.

P. Vasseur, S. Dion, A. Filliol, V. Genet, G. Jean-philippe et al., Endogenous IL-33 has no effect on the progression of fibrosis during experimental steatohepatitis, Oncotarget, vol.8, pp.48563-74, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01578584

Y. Gao, Y. Liu, M. Yang, X. Guo, M. Zhang et al., IL-33 treatment attenuated diet-induced hepatic steatosis but aggravated hepatic fibrosis, Oncotarget, vol.7, pp.1-13, 2016.

I. Jeftic, N. Jovicic, J. Pantic, N. Arsenijevic, M. L. Lukic et al., Galectin-3 ablation enhances liver steatosis, but attenuates inflammation and IL-33-dependent fibrosis in obesogenic mouse model of nonalcoholic steatohepatitis, Mol Med Camb Mass, vol.21, pp.453-65, 2015.

B. Jaruga, F. Hong, R. Sun, S. Radaeva, and B. Gao, Crucial role of IL-4/STAT6 in T cell-mediated hepatitis: up-regulating eotaxins and IL-5 and recruiting leukocytes, J Immunol, vol.171, pp.3233-3277, 2003.

Y. Tagawa, K. Sekikawa, and Y. Iwakura, Suppression of concanavalin Ainduced hepatitis in IFN-gamma(-/-) mice, but not in TNF-alpha(-/-) mice: role for IFN-gamma in activating apoptosis of hepatocytes, J Immunol, vol.159, pp.1418-1446, 1997.

R. Zhu, S. Diem, L. M. Araujo, A. Aumeunier, J. Denizeau et al., The Pro-Th1 cytokine IL-12 enhances IL-4 production by invariant NKT cells: relevance for T cell-mediated hepatitis, J Immunol, vol.178, pp.5435-5477, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00314236

K. Takeda, Y. Hayakawa, L. Van-kaer, H. Matsuda, H. Yagita et al., Critical contribution of liver natural killer T cells to a murine model of hepatitis, Proc Natl Acad Sci, vol.97, pp.5498-503, 2000.

M. I. Arshad, C. Piquet-pellorce, L. 'helgoualc'h, A. Rauch, M. Patrat-delon et al., TRAIL but not FasL and TNF?, regulates IL-33 expression in murine hepatocytes during acute hepatitis, Hepatology, vol.56, pp.2353-62, 2012.

J. Schümann, D. Wolf, A. Pahl, K. Brune, T. Papadopoulos et al., Importance of Kupffer cells for T-cell-dependent liver injury in mice, Am J Pathol, vol.157, issue.10, pp.64804-64807, 2000.

J. Luan, X. Zhang, S. Wang, Y. Li, J. Fan et al., NOD-like receptor protein 3 inflammasome-dependent IL-1? accelerated ConA-induced Hepatitis, Front Immunol, vol.9, p.758, 2018.

G. Noel, M. I. Arshad, A. Filliol, V. Genet, M. Rauch et al., Ablation of interaction between IL-33 and ST2+ regulatory T cells increases immune cell-mediated hepatitis and activated NK cell liver infiltration, Am J Physiol Gastrointest Liver Physiol, vol.311, pp.313-336, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01366372

V. Volarevic, M. Mitrovic, M. Milovanovic, I. Zelen, I. Nikolic et al., Protective role of IL-33/ST2 axis in Con A-induced hepatitis, J Hepatol, vol.56, pp.26-33, 2012.

D. Burzyn, W. Kuswanto, D. Kolodin, J. L. Shadrach, M. Cerletti et al., A special population of regulatory T cells potentiates muscle repair, Cell, vol.155, pp.1282-95, 2013.

S. Shrivastava, A. Mukherjee, R. Ray, and R. B. Ray, Hepatitis C virus induces interleukin-1 (IL-1 )/IL-18 in circulatory and resident liver macrophages, J Virol, vol.87, pp.12284-90, 2013.

A. A. Negash, H. J. Ramos, N. Crochet, D. Lau, B. Doehle et al., IL-1? production through the NLRP3 Inflammasome by hepatic macrophages links hepatitis C virus infection with liver inflammation and disease, PLoS Pathog, vol.9, pp.1-13, 2013.

K. Yanagisawa, S. Yue, H. J. Van-der-vliet, R. Wang, N. Alatrakchi et al., Ex vivo analysis of resident hepatic pro-inflammatory CD1d-reactive T cells and hepatocyte surface CD1d expression in hepatitis C, J Viral Hepat, vol.20, pp.556-65, 2013.

P. Fisicaro, C. Valdatta, M. Massari, E. Loggi, E. Biasini et al., Antiviral intrahepatic T-cell responses can be restored by blocking programmed death-1 pathway in chronic hepatitis B, Gastroenterology, vol.138, pp.1-4, 2010.

M. Hofmann and R. Thimme, iNKT cells in chronic HBV: a balancing act, Hepatol Int, vol.10, pp.535-542, 2016.

M. Li, Z. Zhou, X. Sun, X. Zhang, X. Zhu et al., The dynamic changes of circulating invariant natural killer T cells during chronic hepatitis B virus infection, Hepatol Int, vol.10, pp.594-601, 2016.

K. Kimura, K. Kakimi, S. Wieland, L. G. Guidotti, and F. V. Chisari, Interleukin-18 inhibits hepatitis B virus replication in the livers of transgenic mice, J Virol, vol.76, pp.10702-10709, 2002.

X. X. Du, Y. Shi, Y. Yang, Y. Yu, H. G. Lou et al., DAMP molecular IL-33 augments monocytic inflammatory storm in hepatitis B-precipitated acuteon-chronic liver failure, Liver Int, vol.38, pp.229-267, 2018.

S. Huan, J. Zhao, Z. Wang, S. Gao, and K. Wang, Relevance of serum interleukin-33 and ST2 levels and the natural course of chronic hepatitis B virus infection, BMC Infect Dis, vol.16, p.200, 2016.

J. Wang, P. Zhao, H. Guo, X. Sun, Z. Jiang et al., Serum IL-33 levels are associated with liver damage in patients with chronic hepatitis C, Mediators Inflamm, p.819636, 2012.

R. G. Gieling, K. Wallace, and Y. Han, Interleukin-1 participates in the progression from liver injury to fibrosis, Am J Physiol-Gastrointest Liver Physiol, vol.296, pp.1324-1355, 2009.

P. Marvie, M. Lisbonne, L. 'helgoualc'h, A. Rauch, M. Turlin et al., Interleukin-33 overexpression is associated with liver fibrosis in mice and humans, J Cell Mol Med, vol.14, pp.1726-1765, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00660257

T. Mchedlidze, M. Waldner, S. Zopf, J. Walker, A. L. Rankin et al., Interleukin-33-dependent innate lymphoid cells mediate hepatic fibrosis, Immunity, vol.39, pp.357-71, 2013.

C. Ma, M. Han, B. Heinrich, Q. Fu, Q. Zhang et al., Gut microbiomemediated bile acid metabolism regulates liver cancer via NKT cells, Science, vol.360, p.5931, 2018.

T. Miyagi, T. Takehara, T. Tatsumi, T. Kanto, T. Suzuki et al., CD1d-mediated stimulation of natural killer T cells selectively activates hepatic natural killer cells to eliminate experimentally disseminated hepatoma cells in murine liver, Int J Cancer, vol.106, pp.81-90, 2003.

M. C. Leite-de-moraes, A. Hameg, A. Arnould, F. Machavoine, Y. Koezuka et al., A distinct IL-18-induced pathway to fully activate NK T lymphocytes independently from TCR engagement, J Immunol, vol.163, pp.5871-5877, 1999.

G. Bricard, V. Cesson, E. Devevre, H. Bouzourene, C. Barbey et al., Enrichment of human CD4 + V?24/V?11 invariant NKT cells in intrahepatic malignant tumors, J Immunol, vol.182, pp.5140-51, 2009.

X. Gao, X. Wang, Q. Yang, X. Zhao, W. Wen et al., Tumoral expression of IL-33 inhibits tumor growth and modifies the tumor microenvironment through CD8 + T and NK cells, J Immunol, vol.194, pp.438-483, 2015.

C. Hollande, J. Boussier, J. Ziai, T. Nozawa, V. Bondet et al., Inhibition of the dipeptidyl peptidase DPP4 (CD26) reveals IL-33-dependent eosinophil-mediated control of tumor growth, Nat Immunol, vol.20, pp.257-64, 2019.
URL : https://hal.archives-ouvertes.fr/pasteur-02063831

D. O. Villarreal, M. C. Wise, J. N. Walters, E. L. Reuschel, M. J. Choi et al., Alarmin IL-33 acts as an immunoadjuvant to enhance antigen-specific tumor immunity, Cancer Res, vol.74, pp.1789-800, 2014.

J. Zhang, Q. Zhang, Y. Lou, Q. Fu, Q. Chen et al., Hypoxia-inducible factor-1?/interleukin-1? signaling enhances hepatoma epithelialmesenchymal transition through macrophages in a hypoxic-inflammatory microenvironment, Hepatology, vol.67, pp.1872-89, 2018.

J. Zhao, Q. Pan, K. Pan, D. Weng, Q. Wang et al., Interleukin-37 mediates the antitumor activity in hepatocellular carcinoma: role for CD57+ NK cells, Sci Rep, vol.4, p.5177, 2015.

R. Liu, C. Tang, A. Shen, H. Luo, X. Wei et al., IL-37 suppresses hepatocellular carcinoma growth by converting pSmad3 signaling from JNK/pSmad3L/c-Myc oncogenic signaling to pSmad3C/P21 tumor-suppressive signaling, Oncotarget, vol.7, pp.85079-96, 2016.

T. Li, D. Zhu, T. Mou, Z. Guo, J. Pu et al., IL-37 induces autophagy in hepatocellular carcinoma cells by inhibiting the PI3K/AKT/mTOR pathway, Mol Immunol, vol.87, pp.132-172, 2017.

Y. Yang, J. B. Wang, Y. M. Li, Y. Zhao, R. Wang et al., Role of IL-33 expression in oncogenesis and development of human hepatocellular carcinoma, Oncol Lett, vol.12, pp.429-465, 2016.

P. Zhang, X. Liu, Z. Chu, J. Ye, K. Li et al., Detection of interleukin-33 in serum and carcinoma tissue from patients with hepatocellular carcinoma and its clinical implications, J Int Med Res, vol.40, pp.1654-61, 2012.

D. Bergis, V. Kassis, A. Ranglack, V. Koeberle, A. Piiper et al., High serum levels of the interleukin-33 receptor soluble ST2 as a negative prognostic factor in hepatocellular carcinoma, Transl Oncol, vol.6, pp.311-319, 2013.

C. Berasain, J. Castillo, M. J. Perugorria, M. U. Latasa, J. Prieto et al., Inflammation and liver cancer: new molecular links, Ann N Y Acad Sci, vol.1155, pp.206-227, 2009.

X. Wu, L. Dong, X. Lin, and J. Li, Relevance of the NLRP3 inflammasome in the pathogenesis of chronic liver disease. Front Immunol, vol.8, p.1728, 2017.

Y. Yang, H. Wang, M. Kouadir, H. Song, and F. Shi, Recent advances in the mechanisms of NLRP3 inflammasome activation and its inhibitors, Cell Death Dis, vol.10, p.128, 2019.

A. M. Adkison, S. Z. Raptis, D. G. Kelley, and C. Pham, Dipeptidyl peptidase I activates neutrophil-derived serine proteases and regulates the development of acute experimental arthritis, J Clin Invest, vol.109, pp.363-71, 2002.

J. A. Lust, M. Q. Lacy, S. R. Zeldenrust, T. E. Witzig, L. L. Moon-tasson et al., Reduction in C-reactive protein indicates successful targeting of the IL-1/IL-6 axis resulting in improved survival in early stage multiple myeloma, Am J Hematol, vol.91, pp.571-575, 2016.

T. Wu, K. Xu, J. Martinek, R. R. Young, R. Banchereau et al., IL1 receptor antagonist controls transcriptional signature of inflammation in patients with metastatic breast cancer, Cancer Res, vol.78, pp.5243-58, 2018.

P. M. Ridker, B. M. Everett, T. Thuren, J. G. Macfadyen, W. H. Chang et al., Antiinflammatory therapy with canakinumab for atherosclerotic disease, N Engl J Med, vol.377, pp.1119-1150, 2017.

M. E. Stremska, S. Jose, V. Sabapathy, L. Huang, A. Bajwa et al., A novel IL-2 and IL-33 hybrid cytokine, ameliorates renal injury, J Am Soc Nephrol, vol.28, pp.2681-93, 2017.

, Reference(s) REFERENCES

K. Buchmann, Evolution of innate immunity: clues from invertebrates via fish to mammals, Front Immunol, vol.5, p.459, 2014.

B. Mcdonald and P. Kubes, Innate immune cell trafficking and function during sterile inflammation of the liver, Gastroenterology, vol.151, pp.1087-95, 2016.

H. Shen, D. Kreisel, and D. R. Goldstein, Processes of sterile inflammation, J Immunol, vol.191, pp.2857-63, 2013.

J. R. Lukens, J. M. Gross, and T. Kanneganti, IL-1 family cytokines trigger sterile inflammatory disease, Front Immunol, vol.3, p.315, 2012.

G. Y. Chen and G. Nuñez, Sterile inflammation: sensing and reacting to damage, Nat Rev Immunol, vol.10, pp.826-863, 2010.

M. A. Sugimoto, L. P. Sousa, V. Pinho, M. Perretti, and M. M. Teixeira, Resolution of inflammation: what controls its onset? Front Immunol, vol.7, p.160, 2016.

M. Ferhat, A. Robin, S. Giraud, S. Sena, J. M. Goujon et al., Endogenous IL-33 contributes to kidney ischemia-reperfusion injury as an alarmin, J Am Soc Nephrol, vol.29, pp.1272-88, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01929544

L. Li, L. Huang, S. J. Sung, P. I. Lobo, M. G. Brown et al., NKT cell activation mediates neutrophil IFN-gamma production and renal ischemia-reperfusion injury, J Immunol, vol.178, pp.5899-911, 2007.

K. Shimamura, H. Kawamura, T. Nagura, T. Kato, T. Naito et al., Association of NKT cells and granulocytes with liver injury after reperfusion of the portal vein, Cell Immunol, vol.234, pp.31-39, 2005.

M. A. Zimmerman, A. Martin, J. Yee, J. Schiller, and J. C. Hong, Natural killer T cells in liver ischemia-reperfusion injury, J Clin Med, vol.6, p.41, 2017.

C. M. Lappas, Y. J. Day, M. A. Marshall, V. H. Engelhard, and J. Linden, Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation, J Exp Med, vol.203, pp.2639-2687, 2006.

P. Arrenberg, I. Maricic, and V. Kumar, Sulfatide-mediated activation of type II natural killer T cells prevents hepatic ischemic reperfusion injury in mice, Gastroenterology, vol.140, pp.646-55, 2011.

A. K. Sharma, D. J. Lapar, Y. Zhao, L. Li, C. L. Lau et al., Natural killer T cell-derived IL-17 mediates lung ischemia-reperfusion injury, Am J Respir Crit Care Med, vol.183, pp.1539-1588, 2011.

Z. K. Wang, L. Xue, T. Wang, X. J. Wang, and Z. Q. Su, Infiltration of invariant natural killer T cells occur and accelerate brain infarction in permanent ischemic stroke in mice, Neurosci Lett, vol.633, pp.62-70, 2016.

T. Homma, S. Kinugawa, M. Takahashi, M. A. Sobirin, A. Saito et al., Activation of invariant natural killer T cells by ?-galactosylceramide ameliorates myocardial ischemia/reperfusion injury in mice, J Mol Cell Cardiol, vol.62, pp.179-88, 2013.

J. A. Richards, S. J. Wigmore, S. M. Anderton, and S. Howie, NKT cells are important mediators of hepatic ischemia-reperfusion injury, Transpl Immunol, vol.45, pp.15-21, 2017.

S. Kuboki, N. Sakai, J. Tschöp, M. J. Edwards, A. B. Lentsch et al., Distinct contributions of CD4+ T cell subsets in hepatic ischemia/reperfusion injury, Am J Physiol Gastrointest Liver Physiol, vol.296, pp.1054-1063, 2009.

H. O. Yazdani, H. W. Chen, S. Tohme, S. Tai, D. J. Van-der-windt et al., IL-33 exacerbates liver sterile inflammation by amplifying neutrophil extracellular trap formation, J Hepatol, 2017.

M. I. Arshad, M. Rauch, L. 'helgoualc'h, A. , J. V. Leite-de-moraes et al., NKT cells are required to induce high IL-33 expression in hepatocytes during ConA-induced acute hepatitis, Eur J Immunol, vol.41, pp.2341-2349, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00681990

M. I. Arshad, C. Piquet-pellorce, L. 'helgoualc'h, A. Rauch, M. Patrat-delon et al., TRAIL but not FasL and TNF?, regulates IL-33 expression in murine hepatocytes during acute hepatitis, Hepatology, vol.56, pp.2353-62, 2012.

Z. Cao, Y. Yuan, G. Jeyabalan, Q. Du, A. Tsung et al., Preactivation of NKT cells with alpha-GalCer protects against hepatic ischemia-reperfusion injury in mouse by a mechanism involving IL-13 and adenosine A2A receptor, Am J Physiol Gastrointest Liver Physiol, vol.297, pp.249-58, 2009.

L. Cheng, Q. You, H. Yin, M. P. Holt, and C. Ju, Involvement of natural killer T cells in halothane-induced liver injury in mice, Biochem Pharmacol, vol.80, pp.255-61, 2010.

M. M. Antunes, A. M. Araújo, A. B. Diniz, R. Pereira, D. M. Alvarenga et al., IL-33 signalling in liver immune cells enhances druginduced liver injury and inflammation, Inflamm Res, vol.67, pp.77-88, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02126492

P. X. Liew, W. Y. Lee, and P. Kubes, iNKT Cells orchestrate a switch from inflammation to resolution of sterile liver injury, Immunity, vol.47, pp.752-65, 2017.

A. K. Sharma, D. J. Lapar, M. L. Stone, Y. Zhao, C. K. Mehta et al., NOX2 activation of natural killer T cells is blocked by the adenosine A2A receptor to inhibit lung ischemia-reperfusion injury, Am J Respir Crit Care Med, vol.193, pp.988-99, 2016.

F. Grabarz, C. F. Aguiar, M. Correa-costa, T. T. Braga, M. I. Hyane et al., Protective role of NKT cells and macrophage M2-driven phenotype in bleomycin-induced pulmonary fibrosis, Inflammopharmacology, vol.26, pp.491-504, 2018.

J. H. Kim, H. Y. Kim, S. Kim, J. H. Chung, W. S. Park et al., Natural killer T (NKT) cells attenuate bleomycin-induced pulmonary fibrosis by producing interferon-gamma, Am J Pathol, vol.167, issue.10, pp.61211-61215, 2005.

E. A. Bourgeois, A. Levescot, S. Diem, A. Chauvineau, H. Bergès et al., A natural protective function of invariant NKT cells in a mouse model of innate-cell-driven lung inflammation, Eur J Immunol, vol.41, pp.299-305, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00539574

D. Li, R. Guabiraba, A. G. Besnard, M. Komai-koma, M. S. Jabir et al., IL-33 promotes ST2-dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice, J Allergy Clin Immunol, vol.134, pp.1422-1454, 2014.

I. G. Luzina, P. Kopach, V. Lockatell, P. H. Kang, A. Nagarsekar et al., Interleukin-33 potentiates bleomycin-induced lung injury, Am J Respir Cell Mol Biol, vol.49, pp.999-1008, 2013.

C. Michaudel, C. Mackowiak, I. Maillet, L. Fauconnier, C. A. Akdis et al., Ozone exposure induces respiratory barrier biphasic injury and inflammation controlled by IL-33, J Allergy Clin Immunol, vol.142, pp.942-58, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02126403

M. Pichavant, S. Goya, E. H. Meyer, R. A. Johnston, H. Y. Kim et al., Ozone exposure in a mouse model induces airway hyperreactivity that requires the presence of natural killer T cells and IL-17, J Exp Med, vol.205, pp.385-93, 2008.

S. Kubala, M. Q. Ge, I. Redai, M. Soni, B. Chen et al., Ozone inhalation induces epithelial IL-33 and Thymic Stromal Lymphopoietin (TSLP) and leads to eosinophilic airway inflammation, J Allergy Clin Immunol, vol.133, p.145, 2014.

V. Savransky, R. R. Molls, M. Burne-taney, C. C. Chien, L. Racusen et al., Role of the T-cell receptor in kidney ischemia-reperfusion injury, Kidney Int, vol.69, pp.233-241, 2006.

K. Hochegger, T. Schätz, P. Eller, A. Tagwerker, D. Heininger et al., Role of alpha/beta and gamma/delta T cells in renal ischemiareperfusion injury, Am J Physiol Renal Physiol, vol.293, pp.741-748, 2007.

C. Kleinschnitz, N. Schwab, P. Kraft, I. Hagedorn, A. Dreykluft et al., Early detrimental T-cell effects in experimental cerebral ischemia are neither related to adaptive immunity nor thrombus formation, Blood, vol.115, pp.3835-3877, 2010.

Z. X. Zhang, S. Wang, X. Huang, W. P. Min, H. Sun et al., NK cells induce apoptosis in tubular epithelial cells and contribute to renal ischemia-reperfusion injury, J Immunol, vol.181, pp.7489-98, 2008.

J. C. Vahl, K. Heger, N. Knies, M. Y. Hein, L. Boon et al., NKT Cell-TCR Expression activates conventional T Cells in vivo, but is largely dispensable for mature NKT cell biology, PLOS Biol, vol.11, 2013.

K. L. Holzapfel, A. J. Tyznik, M. Kronenberg, and K. A. Hogquist, Antigen-dependent versus -independent activation of iNKT cells during infection, J Immunol, vol.192, pp.5490-5498, 2014.

M. C. Leite-de-moraes, A. Hameg, A. Arnould, F. Machavoine, Y. Koezuka et al., A distinct IL-18-induced pathway to fully activate NK T lymphocytes independently from TCR engagement, J Immunol, vol.163, pp.5871-5877, 1999.

M. C. Leite-de-moraes, A. Hameg, M. Pacilio, Y. Koezuka, M. Taniguchi et al., IL-18 enhances IL-4 production by ligand-activated NKT lymphocytes: a pro-Th2 effect of IL-18 exerted through NKT cells, J Immunol, vol.166, pp.945-51, 2001.

E. Bourgeois, L. P. Van, M. Samson, S. Diem, A. Barra et al., The pro-Th2 cytokine IL-33 directly interacts with invariant NKT and NK cells to induce IFN-gamma production, Eur J Immunol, vol.39, pp.1046-55, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00676402

R. Zhu, S. Diem, L. M. Araujo, A. Aumeunier, J. Denizeau et al., The Pro-Th1 cytokine IL-12 enhances IL-4 production by invariant nkt cells: relevance for t cell-mediated hepatitis, J Immunol, vol.178, pp.5435-5477, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00314236

V. P. Marques, G. M. Gonçalves, C. Q. Feitoza, M. A. Cenedeze, F. Bertocchi et al., Influence of TH1/TH2 switched immune response on renal ischemia-reperfusion injury, Nephron Exp Nephrol, vol.104, pp.48-56, 2006.

C. Shimokawa, T. Kanaya, M. Hachisuka, K. Ishiwata, H. Hisaeda et al., Mast cells are crucial for induction of group 2 innate lymphoid cells and clearance of helminth infections, Immunity, vol.46, pp.863-74, 2017.

C. Cayrol, A. Duval, P. Schmitt, S. Roga, M. Camus et al., Environmental allergens induce allergic inflammation through proteolytic maturation of IL-33, Nat Immunol, vol.19, pp.375-85, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02179492

S. P. Gadani, J. T. Walsh, I. Smirnov, J. Zheng, and J. Kipnis, The glia-derived alarmin IL-33 orchestrates the immune response and promotes recovery following CNS injury, Neuron, vol.85, pp.703-712, 2015.

J. Schmitz, A. Owyang, E. Oldham, Y. Song, E. Murphy et al., IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines, Immunity, vol.23, pp.479-90, 2005.

M. D. Smithgall, M. R. Comeau, B. Yoon, D. Kaufman, R. Armitage et al., IL-33 amplifies both Th1-and Th2-type responses through its activity on human basophils, allergen-reactive Th2 cells, iNKT and NK cells, Int Immunol, vol.20, pp.1019-1049, 2008.

F. Y. Liew, J. P. Girard, and H. R. Turnquist, Interleukin-33 in health and disease, Nat Rev Immunol, vol.16, pp.676-89, 2016.

Q. Luo, Y. Fan, L. Lin, J. Wei, Z. Li et al., Interleukin-33 protects ischemic brain injury by regulating specific microglial activities, Neuroscience, vol.385, pp.75-89, 2018.

A. Akcay, Q. Nguyen, Z. He, K. Turkmen, W. Lee et al., IL-33 Exacerbates acute kidney injury, J Am Soc Nephrol, vol.22, pp.2057-67, 2011.

A. Thierry, S. Giraud, A. Robin, A. Barra, F. Bridoux et al., The alarmin concept applied to human renal transplantation: evidence for a differential implication of HMGB1 and IL-33, PLoS ONE, vol.9, p.88742, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01928093

A. Thierry, A. Robin, S. Giraud, S. Minouflet, A. Barra et al., Identification of invariant natural killer T cells in porcine peripheral blood, Vet Immunol Immunopathol, vol.149, pp.272-281, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00871261

M. Y. Zeng, D. Pham, J. Bagaitkar, J. Liu, K. Otero et al., An efferocytosisinduced, IL-4-dependent macrophage-iNKT cell circuit suppresses sterile inflammation and is defective in murine CGD, Blood, vol.121, pp.3473-83, 2013.

Y. Kaneko, M. Harada, T. Kawano, M. Yamashita, Y. Shibata et al., Augmentation of Valpha14 NKT cell-mediated cytotoxicity by interleukin 4 in an autocrine mechanism resulting in the development of concanavalin A-induced hepatitis, J Exp Med, vol.191, pp.105-119, 2000.

K. Takeda, Y. Hayakawa, L. Van-kaer, H. Matsuda, H. Yagita et al., Critical contribution of liver natural killer T cells to a murine model of hepatitis, Proc Natl Acad Sci, vol.97, pp.5498-503, 2000.

B. V. Martin-murphy, D. J. Kominsky, D. J. Orlicky, T. M. Donohue, and C. Ju, Increased susceptibility of natural killer T-cell-deficient mice to acetaminopheninduced liver injury, Hepatology, vol.57, pp.1575-84, 2013.

V. Kumar, NKT-cell subsets: promoters and protectors in inflammatory liver disease, J Hepatol, vol.59, pp.618-638, 2013.

S. H. Yang, J. P. Lee, H. R. Jang, R. Cha, S. S. Han et al., Sulfatide-reactive natural killer T cells abrogate ischemia-reperfusion injury, J Am Soc Nephrol, vol.22, pp.1305-1319, 2011.

Y. S. Choi, H. J. Choi, J. K. Min, B. J. Pyun, Y. S. Maeng et al., Interleukin-33 induces angiogenesis and vascular permeability through ST2/TRAF6-mediated endothelial nitric oxide production, Blood, vol.114, pp.3117-3143, 2009.

C. Schiering, T. Krausgruber, A. Chomka, A. Fröhlich, K. Adelmann et al., The alarmin IL-33 promotes regulatory T-cell function in the intestine, Nature, vol.513, pp.564-572, 2014.

S. Schiaffino, M. G. Pereira, S. Ciciliot, and P. Rovere-querini, Regulatory T cells and skeletal muscle regeneration, FEBS J, vol.284, pp.517-541

G. D. Rak, L. C. Osborne, M. C. Siracusa, B. S. Kim, K. Wang et al., IL-33-dependent group 2 innate lymphoid cells promote cutaneous wound healing, J Invest Dermatol, vol.136, pp.487-96, 2016.

Y. Yang, H. Liu, H. Zhang, Q. Ye, J. Wang et al., ST2/IL-33-Dependent microglial response limits acute ischemic brain injury, J Neurosci, vol.37, pp.4692-704, 2017.

G. Wicher, U. Wallenquist, Y. Lei, M. Enoksson, X. Li et al., Interleukin-33 promotes recruitment of microglia/macrophages in response to traumatic brain injury, J Neurotrauma, vol.34, pp.3173-82, 2017.

L. Cava, A. Van-kaer, L. , and F. Shi, CD4+CD25+ Tregs and NKT cells: regulators regulating regulators, vol.27, pp.322-329, 2006.

K. Venken, T. Decruy, S. Aspeslagh, S. Van-calenbergh, B. N. Lambrecht et al., Bacterial CD1d-restricted glycolipids induce IL-10 production by human regulatory T cells upon cross-talk with invariant NKT cells, J Immunol, vol.191, pp.2174-83, 2013.

H. Y. Kim, Y. J. Chang, S. Subramanian, H. H. Lee, L. A. Albacker et al., Innate lymphoid cells responding to IL-33 mediate airway hyperreactivity independently of adaptive immunity, J Allergy Clin Immunol, vol.129, pp.216-243, 2012.

B. Nandi and S. M. Behar, Regulation of neutrophils by interferon-? limits lung inflammation during tuberculosis infection, J Exp Med, vol.208, pp.2251-62, 2011.

A. B. Molofsky, F. Van-gool, H. E. Liang, S. J. Van-dyken, J. C. Nussbaum et al., Interleukin-33 and interferon-? counter-regulate group 2 innate lymphoid cell activation during immune perturbation, Immunity, vol.43, pp.161-74, 2015.

L. Li, L. Huang, A. L. Vergis, H. Ye, A. Bajwa et al., IL-17 produced by neutrophils regulates IFN-gamma-mediated neutrophil migration in mouse kidney ischemia-reperfusion injury, J Clin Invest, vol.120, pp.331-373, 2010.

N. Sakai, H. L. Van-sweringen, R. C. Quillin, R. Schuster, J. Blanchard et al., Interleukin-33 is hepatoprotective during liver ischemia/reperfusion in mice, Hepatology, vol.56, pp.1468-78, 2012.

K. Seki, S. Sanada, A. Y. Kudinova, M. L. Steinhauser, V. Handa et al., Interleukin-33 prevents apoptosis and improves survival after experimental myocardial infarction through ST2 signaling, Circ Heart Fail, vol.2, pp.684-91, 2009.

T. Rui, J. Zhang, X. Xu, Y. Yao, R. Kao et al., Reduction in IL-33 expression exaggerates ischaemia/reperfusion-induced myocardial injury in mice with diabetes mellitus, Cardiovasc Res, vol.94, pp.370-378, 2012.