D. Nutt, L. A. King, W. Saulsbury, and C. Blakemore, Development of a rational scale to assess the harm of drugs of potential misuse. The Lancet, vol.369, pp.1047-53, 2007.

O. Sergent, Role for Membrane Fluidity in Ethanol-Induced Oxidative Stress of Primary Rat Hepatocytes, J Pharmacol Exp Ther, vol.313, issue.1, pp.104-115, 2004.

J. Miñana, Mitochondrial oxidative stress and CD95 ligand: A dual mechanism for hepatocyte apoptosis in chronic alcoholism, Hepatology, vol.35, issue.5, pp.1205-1219, 2002.

N. Nieto, Stimulation and proliferation of primary rat hepatic stellate cells by cytochrome P450 2E1-derived reactive oxygen species, Hepatology, vol.35, issue.1, pp.62-73, 2002.

E. González-reimers, Alcoholism: A systemic proinflammatory condition, World J Gastroenterol, vol.20, issue.40, p.14660, 2014.

Q. Dai and S. B. Pruett, Different Effects of Acute and Chronic Ethanol on LPS-Induced Cytokine Production and TLR4 Receptor Behavior in Mouse Peritoneal Macrophages, J Immunotoxicol, vol.3, issue.4, pp.217-242, 2006.

K. Nurmi, J. Virkanen, K. Rajamäki, K. Niemi, P. T. Kovanen et al., Ethanol Inhibits Activation of NLRP3 and AIM2 Inflammasomes in Human Macrophages-A Novel Anti-Inflammatory Action of Alcohol. Catapano A, editor, PLoS ONE, vol.8, issue.11, p.78537, 2013.

L. R. Hoyt, J. L. Ather, M. J. Randall, D. P. Depuccio, C. C. Landry et al., Ethanol and Other Short-Chain Alcohols Inhibit NLRP3 Inflammasome Activation through Protein Tyrosine Phosphatase Stimulation, J Immunol, vol.197, issue.4, pp.1322-1356, 2016.

L. R. Hoyt, M. J. Randall, J. L. Ather, D. P. Depuccio, C. C. Landry et al., Mitochondrial ROS induced by chronic ethanol exposure promote hyper-activation of the NLRP3 inflammasome, Redox Biol, vol.12, pp.883-96, 2017.

J. S. Kim, K. D. Kim, H. S. Na, S. Y. Jeong, H. R. Park et al., Tumor necrosis factor-? and interleukin-1? expression pathway induced by Streptococcus mutans in macrophage cell line RAW 264.7: Macrophage cytokine induction by S. mutans, Mol Oral Microbiol, vol.27, issue.3, pp.149-59, 2012.

P. A. Keyel, How is inflammation initiated? Individual influences of IL-1, IL-18 and HMGB1, Cytokine, vol.69, issue.1, pp.136-181, 2014.

C. N. Larock and B. T. Cookson, The Yersinia Virulence Effector YopM Binds Caspase-1 to Arrest Inflammasome Assembly and Processing, Cell Host Microbe, vol.12, issue.6, pp.799-805, 2012.

P. Liao, L. K. Chao, J. Chou, W. Dong, C. Lin et al., Lipopolysaccharide/adenosine triphosphate-mediated signal transduction in the regulation of NLRP3 protein expression and caspase-1-mediated interleukin-1? secretion, Inflamm Res, vol.62, issue.1, pp.89-96, 2013.

C. Dostert, V. Petrilli, R. Van-bruggen, C. Steele, B. T. Mossman et al., Innate Immune Activation Through Nalp3 Inflammasome Sensing of Asbestos and Silica, Science, vol.320, issue.5876, pp.674-681, 2008.

K. Rajamäki, J. Lappalainen, K. Öörni, E. Välimäki, S. Matikainen et al., Cholesterol Crystals Activate the NLRP3 Inflammasome in Human Macrophages: A Accepted Article . Novel Link between Cholesterol Metabolism and Inflammation, PLoS ONE, vol.5, issue.7, p.11765, 2010.

H. Hara, K. Tsuchiya, I. Kawamura, R. Fang, E. Hernandez-cuellar et al., Phosphorylation of the adaptor ASC acts as a molecular switch that controls the formation of speck-like aggregates and inflammasome activity, Nat Immunol, vol.14, issue.12, pp.1247-55, 2013.

Y. Lin, D. Huang, J. Wang, Y. Lin, S. Hsieh et al., Syk is involved in NLRP3 inflammasome-mediated caspase-1 activation through adaptor ASC phosphorylation and enhanced oligomerization, J Leukoc Biol, vol.97, issue.5, pp.825-860, 2015.

H. Wen, D. Gris, Y. Lei, S. Jha, L. Zhang et al., Fatty acid-induced NLRP3ASC inflammasome activation interferes with insulin signaling, Nat Immunol, vol.12, issue.5, pp.408-423, 2011.

X. Xu, D. Yin, H. Ren, W. Gao, F. Li et al., Selective NLRP3 inflammasome inhibitor reduces neuroinflammation and improves long-term neurological outcomes in a murine model of traumatic brain injury, Neurobiol Dis, vol.117, pp.15-27, 2018.

A. P. Perera, R. Fernando, T. Shinde, R. Gundamaraju, B. Southam et al., MCC950, a specific small molecule inhibitor of NLRP3 inflammasome attenuates colonic inflammation in spontaneous colitis mice, 2018.

J. Stack, J. Ryan, and G. Mccarthy, Colchicine: New Insights to an Old Drug, Am J Ther, vol.22, issue.5, pp.151-158, 2015.

N. Dalbeth, T. J. Lauterio, and H. R. Wolfe, Mechanism of Action of Colchicine in the Treatment of Gout, Clin Ther, vol.36, issue.10, pp.1465-79, 2014.

G. J. Martínez, D. S. Celermajer, and P. S. Corrigendum, The NLRP3 inflammasome and the emerging role of colchicine to inhibit atherosclerosis-associated inflammation, Atherosclerosis, vol.269, pp.262-271, 2018.

, Atherosclerosis, vol.273, p.157, 2018.

J. Fulp, L. He, S. Toldo, Y. Jiang, A. Boice et al., Structural Insights of Benzenesulfonamide Analogues as NLRP3 Inflammasome Inhibitors: Design, Synthesis, and Biological Characterization, J Med Chem, vol.61, issue.12, pp.5412-5435, 2018.

T. Gicquel, T. Victoni, A. Fautrel, S. Robert, F. Gleonnec et al., Involvement of purinergic receptors and NOD-like receptor-family protein 3-inflammasome pathway in the adenosine triphosphate-induced cytokine release from macrophages, Clin Exp Pharmacol Physiol, vol.41, issue.4, pp.279-86, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01134782

A. Surprenant and R. A. North, Signaling at Purinergic P2X Receptors, Annu Rev Physiol, vol.71, issue.1, pp.333-59, 2009.

V. Ralevic and G. Burnstock, Receptors for purines and pyrimidines, Pharmacol Rev, vol.50, issue.3, pp.413-92, 1998.

M. P. Abbracchio and G. Burnstock, Purinoceptors: are there families of P2X and P2Y purinoceptors?, Pharmacol Ther, vol.64, issue.3, pp.445-75, 1994.

T. Müller, S. Fay, R. P. Vieira, H. Karmouty-quintana, S. Cicko et al., P2Y6 Receptor Activation Promotes Inflammation and Tissue Remodeling in Pulmonary Fibrosis. Front Immunol, vol.8, 2017.

, Accepted Article

M. T. Miras-portugal, Á. Sebastián-serrano, D. De, L. García, and M. Díaz-hernández, Neuronal P2X7 Receptor: Involvement in Neuronal Physiology and Pathology, J Neurosci, vol.37, issue.30, pp.7063-72, 2017.

J. Yin, S. You, H. Liu, L. Chen, C. Zhang et al., Role of P2X7R in the development and progression of pulmonary hypertension, Respir Res, vol.18, issue.1, 2017.

N. Riteau, P. Gasse, L. Fauconnier, A. Gombault, M. Couegnat et al., Extracellular ATP Is a Danger Signal Activating P2X 7 Receptor in Lung Inflammation and Fibrosis, Am J Respir Crit Care Med, vol.182, issue.6, pp.774-83, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00591486

V. Chandrashekaran, S. Das, R. K. Seth, D. Dattaroy, F. Alhasson et al., Purinergic receptor X7 mediates leptin induced GLUT4 function in stellate cells in nonalcoholic steatohepatitis, Biochim Biophys Acta BBA-Mol Basis Dis, vol.1862, issue.1, pp.32-45, 2016.

X. Li, Y. Zhang, J. Q. Xia, K. Jiang, M. Cui et al., Liver kinase B1/AMPactivated protein kinase-mediated regulation by gentiopicroside ameliorates P2X7 receptor-dependent alcoholic hepatosteatosis: Gentiopicroside improves alcoholic hepatosteatosis, Br J Pharmacol, vol.175, issue.9, pp.1451-70, 2018.

T. Gicquel, L. Daré, B. Boichot, E. Lagente, and V. , Purinergic receptors: new targets for the treatment of gout and fibrosis, Fundam Clin Pharmacol, vol.31, issue.2, pp.136-182, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01508430

F. Qian, J. Xiao, B. Hu, N. Sun, W. Yin et al., High expression of P2X7R is an independent postoperative indicator of poor prognosis in colorectal cancer, Hum Pathol, vol.64, pp.61-69, 2017.

L. Asatryan, O. Ostrovskaya, D. Lieu, and D. L. Davies, Ethanol differentially modulates P2X4 and P2X7 receptor activity and function in BV2 microglial cells, Neuropharmacology, vol.128, pp.11-21, 2018.

F. Martinon, V. Pétrilli, A. Mayor, A. Tardivel, and J. Tschopp, Gout-associated uric acid crystals activate the NALP3 inflammasome, Nature, vol.440, issue.7081, pp.237-278, 2006.

V. Sueblinvong, V. E. Kerchberger, R. Saghafi, S. T. Mills, X. Fan et al., Chronic Alcohol Ingestion Primes the Lung for Bleomycin-Induced Fibrosis in Mice, Alcohol Clin Exp Res, vol.38, issue.2, pp.336-379, 2014.

S. V. Siegmund and D. A. Brenner, Molecular pathogenesis of alcohol-induced hepatic fibrosis, Alcohol Clin Exp Res, vol.29, pp.102-109, 2005.

G. Szabo, P. Mandrekar, S. Oak, and J. Mayerle, Effect of Ethanol on Inflammatory Responses, Pancreatology, vol.7, issue.2-3, pp.115-138, 2007.
DOI : 10.1159/000104236

URL : http://europepmc.org/articles/pmc2790780?pdf=render

P. Arvers, Alcool et poumon : des liaisons dangereuses, Rev Mal Respir, 2018.
DOI : 10.1016/j.rmr.2018.02.009

M. Pascual, P. Baliño, C. Aragón, and C. Guerri, Cytokines and chemokines as biomarkers of ethanol-induced neuroinflammation and anxiety-related behavior: role of TLR4 and TLR2, Neuropharmacology, vol.89, pp.352-361, 2015.

M. Rossi, M. J. Anwar, A. Usman, A. Keshavarzian, and F. Bishehsari, Colorectal Cancer and Alcohol Consumption-Populations to Molecules. Cancers, vol.10, p.38, 2018.

P. Ramadori, F. Cubero, C. Liedtke, C. Trautwein, and Y. Nevzorova, Alcohol and Hepatocellular Carcinoma: Adding Fuel to the Flame. Cancers, vol.9, p.130, 2017.

Y. Liu, N. Nguyen, and G. A. Colditz, Links between Alcohol Consumption and Breast Cancer: A Look at the Evidence. Womens Health, vol.11, pp.65-77, 2015.

, IARC monographs on the evaluation of carcinogenic risks to humans: Volume 100E: Personal habits and indoor combustion: consumption of alcoholic beverages, 2012.

B. P. Vaughn, S. C. Robson, and G. Burnstock, Pathological roles of purinergic signaling in the liver, J Hepatol, vol.57, issue.4, pp.916-936, 2012.

G. Burnstock and I. Novak, Purinergic signalling and diabetes, Purinergic Signal, vol.9, issue.3, pp.307-331, 2013.
DOI : 10.1038/sj.bjp.0706429

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1760723

L. K. Parvathenani, S. Tertyshnikova, C. R. Greco, S. B. Roberts, B. Robertson et al., P2X 7 Mediates Superoxide Production in Primary Microglia and Is Up-regulated in a Transgenic Mouse Model of Alzheimer's Disease, J Biol Chem, vol.278, issue.15, pp.13309-13326, 2003.

E. Adinolfi, L. Raffaghello, A. L. Giuliani, L. Cavazzini, M. Capece et al., Expression of P2X7 Receptor Increases In Vivo Tumor Growth, Cancer Res, vol.72, issue.12, pp.2957-69, 2012.

A. Giannuzzo, S. F. Pedersen, and I. Novak, The P2X7 receptor regulates cell survival, migration and invasion of pancreatic ductal adenocarcinoma cells. Mol Cancer, vol.14, 2015.

A. A. Santos, A. R. Cappellari, F. O. De-marchi, M. P. Gehring, A. Zaparte et al., Potential role of P2X7R in esophageal squamous cell carcinoma proliferation. Purinergic Signal, vol.13, pp.279-92, 2017.

C. Tan, L. Han, L. Zou, C. Luo, A. Liu et al., Expression of P2X7R in breast cancer tissue and the induction of apoptosis by the gene-specific shRNA in MCF-7 cells, Exp Ther Med, vol.10, issue.4, pp.1472-1480, 2015.

A. Ghalali, F. Wiklund, H. Zheng, U. Stenius, and J. Högberg, Atorvastatin prevents ATPdriven invasiveness via P2X7 and EHBP1 signaling in PTEN-expressing prostate cancer cells, Carcinogenesis, vol.35, issue.7, pp.1547-55, 2014.

T. Gicquel, S. Robert, P. Loyer, T. Victoni, A. Bodin et al., IL-1? production is dependent on the activation of purinergic receptors and NLRP3 pathway in human macrophages, FASEB J, vol.29, issue.10, pp.4162-73, 2015.

D. A. Mccarthy, A. Ranganathan, S. Subbaram, N. L. Flaherty, N. Patel et al., Redox-control of the alarmin, Interleukin-1?, Redox Biol, vol.1, issue.1, pp.218-243, 2013.

Y. Liang, F. L. Harris, D. P. Jones, and L. Brown, Alcohol induces mitochondrial redox imbalance in alveolar macrophages, Free Radic Biol Med, vol.65, pp.1427-1461, 2013.

M. M. Chen, M. D. Bird, A. Zahs, C. Deburghgraeve, B. Posnik et al., Pulmonary inflammation after ethanol exposure and burn injury is attenuated in the absence of IL-6, Alcohol, vol.47, issue.3, pp.223-232, 2013.

F. Hong, W. Kim, Z. Tian, B. Jaruga, E. Ishac et al., Elevated interleukin-6 during ethanol consumption acts as a potential endogenous protective cytokine against Accepted Article . ethanol-induced apoptosis in the liver: involvement of induction of Bcl-2 and Bcl-xL proteins, Oncogene, vol.21, issue.1, pp.32-43, 2002.

C. M. Cahill and J. T. Rogers, Interleukin (IL) 1? Induction of IL-6 Is Mediated by a Novel Phosphatidylinositol 3-Kinase-dependent AKT/I?B Kinase ? Pathway Targeting Activator Protein-1, J Biol Chem, vol.283, issue.38, pp.25900-25912, 2008.

A. Fettelschoss, M. Kistowska, S. Leibundgut-landmann, H. Beer, P. Johansen et al., Inflammasome activation and IL-1 target IL-1 for secretion as opposed to surface expression, Proc Natl Acad Sci, vol.108, issue.44, pp.18055-60, 2011.

K. O. Lindros, A. Stowell, P. Pikkarainen, and M. Salaspuro, Elevated blood acetaldehyde in alcoholics with accelerated ethanol elimination, Pharmacol Biochem Behav, vol.13, pp.119-143, 1980.
DOI : 10.1016/s0091-3057(80)80019-0

C. J. Eriksson, Human blood acetaldehyde concentration during ethanol oxidation (update 1982), Pharmacol Biochem Behav, vol.18, issue.1, pp.141-50, 1983.
DOI : 10.1016/0091-3057(83)90162-4

D. Padova, C. Alderman, J. Lieber, and C. S. , Improved methods for the measurement of acetaldehyde concentrations in plasma and red blood cells, Alcohol Clin Exp Res, vol.10, issue.1, pp.86-95, 1986.

A. I. Cederbaum, Alcohol Metabolism. Clin Liver Dis, vol.16, issue.4, pp.667-85, 2012.

C. Eriksson, Measurement of acetaldehyde: what levels occur naturally and in response to alcohol?, Novartis Found Symp, vol.285, pp.247-55, 2007.

R. Hernandez-munoz, X. L. Ma, E. Baraona, and C. S. Lieber, Method of acetaldehyde measurement with minimal artifactual formation in red blood cells and plasma of actively drinking subjects with alcoholism, J Lab Clin Med, vol.120, issue.1, pp.35-41, 1992.

S. Seeland, H. Kettiger, M. Murphy, A. Treiber, J. Giller et al., ATP-induced cellular stress and mitochondrial toxicity in cells expressing purinergic P2X7 receptor, Pharmacol Res Perspect, vol.3, issue.2, p.123, 2015.

C. M. Cruz, A. Rinna, H. J. Forman, A. Ventura, P. M. Persechini et al., ATP Activates a Reactive Oxygen Species-dependent Oxidative Stress Response and Secretion of Proinflammatory Cytokines in Macrophages, J Biol Chem, vol.282, issue.5, pp.2871-2880, 2007.

A. Abderrazak, T. Syrovets, D. Couchie, E. Hadri, K. Friguet et al., NLRP3 inflammasome: From a danger signal sensor to a regulatory node of oxidative stress and inflammatory diseases, Redox Biol, vol.4, pp.296-307, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01112142

Y. Zhang, J. Q. Li, X. Jiang, M. Cui, B. Xia et al., Amelioration of Alcoholic Liver Steatosis by Dihydroquercetin through the Modulation of AMPK-Dependent Lipogenesis Mediated by P2X7R-NLRP3-Inflammasome Activation, J Agric Food Chem, vol.66, pp.4862-71, 2018.

M. Lombardi, M. E. Mantione, D. Baccellieri, D. Ferrara, R. Castellano et al., P2X7 receptor antagonism modulates IL-1? and MMP9 in human atherosclerotic vessels, vol.7, 2017.
DOI : 10.1038/s41598-017-05137-y

URL : https://www.nature.com/articles/s41598-017-05137-y.pdf

O. Karoutzou, S. Kwak, S. Lee, D. Martínez-falguera, F. Sureda et al., Towards a Novel Class of Multitarget-Directed Ligands: Dual P2X7-NMDA Receptor Antagonists, Molecules, vol.23, issue.1, p.230, 2018.