F. Cardoso, P. Marcelino, and L. Bagulho, Acute liver failure: An up-to-date approach, Journal of Critical Care, vol.39, pp.25-30, 2017.
DOI : 10.1016/j.jcrc.2017.01.003

URL : http://repositorio.chlc.min-saude.pt/bitstream/10400.17/2791/1/J%20Crit%20Care%202017%2025.pdf

J. Bass, Circadian topology of metabolism, Nature, vol.22, issue.7424, pp.348-356, 2012.
DOI : 10.1016/j.cub.2012.03.038

A. Curtis, M. Bellet, and P. Sassone-corsi, Circadian Clock Proteins and Immunity, Immunity, vol.40, issue.2, pp.178-186, 2014.
DOI : 10.1016/j.immuni.2014.02.002

J. Early and A. Curtis, Immunometabolism: Is it under the eye of the clock? Semin Immunol, 2016.

K. Man, A. Loudon, and A. Chawla, Immunity around the clock, Science, vol.126, issue.6315, pp.999-1003, 2016.
DOI : 10.1016/j.cmet.2015.08.006

J. Gibbs, L. Ince, and L. Matthews, An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action, Nature Medicine, vol.49, issue.8, pp.919-926, 2014.
DOI : 10.1016/j.ab.2004.11.007

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4268501/pdf

S. Mendez-ferrer, D. Lucas, and M. Battista, Haematopoietic stem cell release is regulated by circadian oscillations, Nature, vol.91, issue.7186, pp.442-447, 2008.
DOI : 10.1172/JCI15543

K. Nguyen, S. Fentress, and Y. Qiu, Circadian Gene Bmal1 Regulates Diurnal Oscillations of Ly6Chi Inflammatory Monocytes, Science, vol.480, issue.7375, pp.1483-1491, 2013.
DOI : 10.1038/nature10653

C. Scheiermann, Y. Kunisaki, and D. Lucas, Adrenergic nerves govern circadian leukocyte recruitment to tissues Dysregulation of inflammatory responses by chronic circadian disruption, Immunity. J.Immunol, vol.37185, issue.10, pp.290-3015796, 2010.

A. Bugge, D. Feng, and L. Everett, Rev-erb?? and Rev-erb?? coordinately protect the circadian clock and normal metabolic function, Genes & Development, vol.26, issue.7, pp.657-667, 2012.
DOI : 10.1101/gad.186858.112

H. Cho, X. Zhao, and M. Hatori, Regulation of circadian behaviour and metabolism by REV-ERB-?? and REV-ERB-??, Nature, vol.6, issue.7396, pp.123-127, 2012.
DOI : 10.1021/cb1002575

H. Duez, J. Van-der-veen, and C. Duhem, Regulation of Bile Acid Synthesis by the Nuclear Receptor Rev-erb??, Gastroenterology, vol.135, issue.2, pp.689-698, 2008.
DOI : 10.1053/j.gastro.2008.05.035

E. Raspe, H. Duez, and A. Mansen, Identification of Rev-erb?? as a physiological repressor of apoC-III gene transcription, Journal of Lipid Research, vol.127, issue.12, pp.2172-2179, 2002.
DOI : 10.1161/01.CIR.98.24.2738

L. Solt, Y. Wang, and S. Banerjee, Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists, Nature, vol.8, issue.7396, pp.62-68, 2012.
DOI : 10.1210/me.2007-0519

L. Yin, N. Wu, and J. Curtin, Rev-erb??, a Heme Sensor That Coordinates Metabolic and Circadian Pathways, Science, vol.281, issue.28, pp.1786-1789, 2007.
DOI : 10.1074/jbc.M600050200

E. Woldt, Y. Sebti, and L. Solt, Rev-erb-?? modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy, Nature Medicine, vol.556, issue.8, pp.1039-1046, 2013.
DOI : 10.1007/978-1-60327-192-9_11

Z. Gerhart-hines, D. Feng, and M. Emmett, The nuclear receptor Rev-erbalpha controls circadian thermogenic plasticity Circadian clock regulates the host response to Salmonella, Nature Bellet MM Proc.Natl.Acad.Sci.U.S.A, vol.503110, pp.410-4139897, 2013.

J. Gibbs, J. Blaikley, and S. Beesley, The nuclear receptor REV-ERB?? mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines, Proceedings of the National Academy of Sciences, vol.8, issue.4, pp.582-587, 2012.
DOI : 10.1023/A:1008942828960

M. Keller, J. Mazuch, and U. Abraham, A circadian clock in macrophages controls inflammatory immune responses, Proceedings of the National Academy of Sciences, vol.22, issue.50, pp.21407-21412, 2009.
DOI : 10.1186/1471-2105-9-303

C. Fontaine, E. Rigamonti, and B. Pourcet, The Nuclear Receptor Rev-erb?? Is a Liver X Receptor (LXR) Target Gene Driving a Negative Feedback Loop on Select LXR-Induced Pathways in Human Macrophages, Molecular Endocrinology, vol.22, issue.8, pp.1797-1811, 2008.
DOI : 10.1210/me.2007-0439

M. Lam, H. Cho, and H. Lesch, Rev-Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription, Nature, vol.67, issue.7455, pp.511-515, 2013.
DOI : 10.1093/nass/nrn280

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3839578/pdf

M. Lamkanfi and V. Dixit, Mechanisms and Functions of Inflammasomes, Cell, vol.157, issue.5, pp.1013-1022, 2014.
DOI : 10.1016/j.cell.2014.04.007

K. Schroder, J. Tschopp, M. Ioannou, and Q. Wang, The inflammasomes Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis, Cell Inflammation Science, vol.140349, pp.821-832316, 2010.

E. Naik and V. Dixit, Modulation of Inflammasome Activity for the Treatment of Auto-inflammatory Disorders, Journal of Clinical Immunology, vol.30, issue.4, pp.485-490, 2010.
DOI : 10.4049/jimmunol.181.1.17

R. Munoz-planillo, P. Kuffa, and G. Martinez-colon, K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome, Immunity Nat Immunol, vol.3812, issue.29, pp.1142-53222, 2011.

N. Subramanian, K. Natarajan, and M. Clatworthy, The Adaptor MAVS Promotes NLRP3 Mitochondrial Localization and Inflammasome Activation, Cell, vol.153, issue.2, pp.348-61, 2013.
DOI : 10.1016/j.cell.2013.02.054

R. Zhou, A. Yazdi, and P. Menu, A role for mitochondria in NLRP3 inflammasome activation, Nature, vol.14, issue.7329, pp.221-226, 2011.
DOI : 10.1038/sj.cdd.4402142

E. Latz, T. Xiao, and A. Stutz, Activation and regulation of the inflammasomes, Nature Reviews Immunology, vol.285, issue.6, pp.397-411, 2013.
DOI : 10.1074/jbc.M109.082305

P. Duewell, H. Kono, and K. Rayner, NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals, The inflammasome in atherosclerosis and type 2 diabetes, pp.1357-1361, 2010.
DOI : 10.1038/nature08938

F. Sheedy, A. Grebe, and K. Rayner, CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation, Nature Immunology, vol.41, issue.8, pp.81-98, 2011.
DOI : 10.1083/jcb.200501113

L. Vande-walle, V. Opdenbosch, N. Jacques, and P. , Negative regulation of the NLRP3 inflammasome by A20 protects against arthritis, Nature, vol.159, issue.7512, pp.69-73, 2014.
DOI : 10.1124/jpet.104.074484

S. Guo, C. Yang, and B. Diao, The NLRP3 Inflammasome and IL-1beta Accelerate Immunologically Mediated Pathology in Experimental Viral Fulminant Hepatitis Genipin protects d-galactosamine and lipopolysaccharideinduced hepatic injury through suppression of the necroptosis-mediated inflammasome signaling, PLoS Pathog Eur J Pharmacol, vol.11812, pp.1005155-38128, 2015.

S. Furuya, H. Kono, and M. Hara, Interleukin 17A plays a role in lipopolysaccharide/d-galactosamine???induced fulminant hepatic injury in mice, Journal of Surgical Research, vol.199, issue.2, pp.487-93, 2015.
DOI : 10.1016/j.jss.2015.05.060

T. Nakama, S. Hirono, and A. Moriuchi, Etoposide prevents apoptosis in mouse liver with ?-galactosamine/lipopolysaccharide-induced fulminant hepatic failure resulting in reduction of lethality, Hepatology, vol.33, issue.6, pp.1441-50, 2001.
DOI : 10.1053/jhep.2001.24561

K. Shirozu, S. Hirai, and T. Tanaka, Farnesyltransferase Inhibitor, Tipifarnib, Prevents Galactosamine/Lipopolysaccharide-Induced Acute Liver Failure, Shock, vol.42, issue.6, pp.570-577, 2014.
DOI : 10.1097/SHK.0000000000000239

URL : http://pdfs.journals.lww.com/shockjournal/2014/12000/Farnesyltransferase_Inhibitor,_Tipifarnib,.12.pdf?token=method|ExpireAbsolute;source|Journals;ttl|1505847258511;payload|mY8D3u1TCCsNvP5E421JYK6N6XICDamxByyYpaNzk7FKjTaa1Yz22MivkHZqjGP4kdS2v0J76WGAnHACH69s21Csk0OpQi3YbjEMdSoz2UhVybFqQxA7lKwSUlA502zQZr96TQRwhVlocEp/sJ586aVbcBFlltKNKo+tbuMfL73hiPqJliudqs17cHeLcLbV/CqjlP3IO0jGHlHQtJWcICDdAyGJMnpi6RlbEJaRheGeh5z5uvqz3FLHgPKVXJzdpGRzmTA0M/Vx2WsqiQOgtaTmNg2Q6wn5iMzsqtQf/n5tgEjAG16c4vzLGe3tKBeR;hash|piPQxc1eJV5r0BpBPS7k4Q==

Z. Zhong, A. Umemura, and E. Sanchez-lopez, NF-??B Restricts Inflammasome Activation via Elimination of Damaged Mitochondria, Cell, vol.164, issue.5, pp.896-910, 2016.
DOI : 10.1016/j.cell.2015.12.057

T. Zhang, S. Cooper, and N. Brockdorff, The interplay of histone modifications - writers that read, EMBO reports, vol.16, issue.11, pp.1467-81, 2015.
DOI : 10.15252/embr.201540945

L. Broderick, D. Nardo, D. Franklin, and B. , The Inflammasomes and Autoinflammatory Syndromes, Annual Review of Pathology: Mechanisms of Disease, vol.10, issue.1, pp.395-424, 2015.
DOI : 10.1146/annurev-pathol-012414-040431

H. Guo, J. Callaway, and J. Ting, Inflammasomes: mechanism of action, role in disease and therapeutics, Nature Medicine, vol.34, issue.7, pp.677-687, 2015.
DOI : 10.1016/j.immuni.2011.02.006

R. Coll, A. Robertson, and J. Chae, A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases, Nature Medicine, vol.1040, issue.3, pp.248-255, 2015.
DOI : 10.1007/978-1-62703-523-1_2

G. Guarda, M. Braun, and F. Staehli, Type I Interferon Inhibits Interleukin-1 Production and Inflammasome Activation, Immunity, vol.34, issue.2, pp.213-236, 2011.
DOI : 10.1016/j.immuni.2011.02.006

URL : https://doi.org/10.1016/j.immuni.2011.02.006

N. Preitner, F. Damiola, and L. Lopez-molina, The Orphan Nuclear Receptor REV-ERB?? Controls Circadian Transcription within the Positive Limb of the Mammalian Circadian Oscillator, Cell, vol.110, issue.2, pp.251-60, 2002.
DOI : 10.1016/S0092-8674(02)00825-5

D. Kojetin and T. Burris, REV-ERB and ROR nuclear receptors as drug targets, Nature Reviews Drug Discovery, vol.43, issue.3, pp.197-216, 2014.
DOI : 10.1124/mol.112.078667

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4865262/pdf

H. Sherman, I. Frumin, and R. Gutman, Long-term restricted feeding alters circadian expression and reduces the level of inflammatory and disease markers, Journal of Cellular and Molecular Medicine, vol.59, issue.12, pp.2745-59, 2011.
DOI : 10.1002/jemt.10203

F. Bauernfeind, G. Horvath, and A. Stutz, Cutting Edge: NF-??B Activating Pattern Recognition and Cytokine Receptors License NLRP3 Inflammasome Activation by Regulating NLRP3 Expression, The Journal of Immunology, vol.183, issue.2, pp.787-91, 2009.
DOI : 10.4049/jimmunol.0901363

URL : http://www.jimmunol.org/content/jimmunol/183/2/787.full.pdf

L. Franchi, T. Eigenbrod, and G. Nunez, Cutting Edge: TNF-?? Mediates Sensitization to ATP and Silica via the NLRP3 Inflammasome in the Absence of Microbial Stimulation, The Journal of Immunology, vol.183, issue.2, pp.792-798, 2009.
DOI : 10.4049/jimmunol.0900173

C. Dinarello, A. Simon, and J. Van-der-meer, Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases, Nature Reviews Drug Discovery, vol.17, issue.8, pp.633-652, 2012.
DOI : 10.1016/j.ejogrb.2005.04.019

+. Nr1d1, Quantification of intracellular IL18 from 150 cells/slide. Data are means ± SD. ***p<0

. Saline, Data are means ± SEM (n=5-6) Data are means ± SEM. *p<0.05, **p<0.01, ***p<0.001 as determined by two-way ANOVA and Bonferroni post-hoc test