, Quelques mécanismes de protection cellulaires

. .. Le-stress-oxydant,

. .. Autophagie,

.. .. Stress,

. .. Apoptose,

. .. Micro-arns,

. .. Biosynthèse-des-miarns,

R. Dans-la-régulation-post-transcriptionnelle and .. .. ,

L. and .. .. ,

M. Matériels and . ????????????????????????,

. .. Conditions-d'élevage, , vol.87

. .. Gavage,

.. .. Abattage,

. .. Tests-de-pasteurisation,

. .. Analyses-biochimiques,

. .. Biologie-moléculaire,

, 7 RT-qPCR : méthode TaqMan (FRET)

, Analyse et traitements statistiques des résultats

1. Expérimentation, , p.105

, Article 1 : EXP1 cinétique??????????????????, p.107

, Article 2 : EXP1 microARNs??????????????????

T. Annabelle, R. Karine, B. Marie-dominique, D. Stéphane, and K. G. , Kinetics of expression of genes involved in glucose metabolism after the last meal in overfed mule ducks, Mol Cell Biochem, vol.430, pp.127-137, 2017.

J. Arroyo, C. Molette, F. Lavigne, C. Margetyal, O. Amador et al., Effects of dietary protein level during rearing period and age at overfeeding on magret and foie gras quality in male mule ducks, Anim Sci J, vol.89, pp.570-578, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02628855

A. Auvergne, H. Rémignon, R. Babilé, and C. Baudonnet-lenfant, Arch Für Geflügelkd, vol.59, pp.234-240, 1995.

S. Awde, N. Marty-gasset, K. Prahkarnkaeo, and H. Rémignon, Relationship between Proteolytic Activities and Cooking Loss Variability in Liver Issued from Force-Fed Mule Ducks, J Agric Food Chem, vol.62, pp.3262-3268, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02636930

R. Babile, A. Auvergne, J. P. Dubois, G. Benard, and H. Manse, Reversibilite de la steatose hepatique chez l'oie. 3emes Journees de la recherche sur les Palmipedes a Foie"gras, 1998.

E. Baéza, C. Marie-etancelin, S. Davail, and C. Diot, La stéatose hépatique chez les palmipèdes, INRA Prod Anim, vol.26, pp.403-414, 2013.

E. Baéza, N. Rideau, P. Chartrin, S. Davail, R. Hoo-paris et al., Canards de Barbarie, Pékin et leurs hybrides: aptitude à l'engraissement, INRA Prod Anim, vol.18, pp.131-141, 2005.

. Barbosa-tessmann and . Ip, Activation of the Human Asparagine Synthetase Gene by the Amino Acid Response and the Endoplasmic Reticulum Stress Response Pathways Occurs by Common Genomic Elements, J. Biol. Chem, 2000.

A. Barthel and D. Schmoll, Proteomic profile evolution during steatosis development in ducks, Am J Physiol-Endocrinol Metab, vol.285, pp.112-120, 2003.

G. Bénard, P. Bénard, D. Prehn, T. Bengone, J. Y. Jouglar et al., Démonstration de la réversibilité de la stéatose hépatique obtenue par gavage de canards mulards, Genet Mol Res, vol.12, pp.16353-16363, 1998.

C. Bonnefont, C. Molette, F. Lavigne, H. Manse, C. Bravo et al., Evolution of liver fattening and foie gras technological yield during the overfeeding period in mule duck, Metabolomic study of fatty livers in ducks: Identification by 1H-NMR of metabolic markers associated with technological quality, vol.93, p.128, 1999.
URL : https://hal.archives-ouvertes.fr/hal-02627243

P. Chartrin, M. Bernadet, G. Guy, J. Mourot, J. Hocquette et al., Does overfeeding enhance genotype effects on liver ability for lipogenesis and lipid secretion in ducks?, Comp Biochem Physiol A Mol Integr Physiol, vol.145, pp.390-396, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02659542

P. Chartrin, K. Meteau, H. Juin, M. D. Bernadet, G. Guy et al., Effects of Intramuscular Fat Levels on Sensory Characteristics of Duck Breast Meat, Poult Sci, vol.85, pp.914-922, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02657230

S. Chitturi, S. Abeygunasekera, G. C. Farrell, J. Holmes-walker, J. M. Hui et al., NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome, Hepatology, vol.35, pp.373-379, 2002.

P. Codogno and A. J. Meijer, Autophagy and signaling: their role in cell survival and cell death, Cell Death Differ, vol.12, pp.1509-1518, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00172272

T. Desvignes, C. Fauvel, and J. Bobe, The nme gene family in zebrafish oogenesis and early development, Naunyn Schmiedebergs Arch Pharmacol, vol.384, pp.439-449, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01205026

T. Desvignes, C. Fauvel, and J. Bobe, The nme gene family in zebrafish oogenesis and early development, Naunyn Schmiedebergs Arch Pharmacol, vol.384, pp.439-449, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01205026

J. Fantini, R. M. Epand, and F. J. Barrantes, Cholesterol-Recognition Motifs in Membrane Proteins, Direct Mechanisms in Cholesterol Modulation of Protein Function, pp.3-25
URL : https://hal.archives-ouvertes.fr/hal-02482669

Y. François, C. Marie-etancelin, A. Vignal, D. Viala, S. Davail et al., Foie Gras" Shows Different Metabolic States According to Its Quality Phenotype by Using a Proteomic Approach, J Agric Food Chem, vol.62, pp.7140-7150, 2014.

J. Gabarrou, M. Salichon, G. Guy, and J. Blum, Hybrid ducks overfed with boiled corn develop an acute hepatic steatosis with decreased choline and polyunsaturated fatty acid level in phospholipids, Reprod Nutr Dev, vol.36, pp.473-484, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02688463

K. Gontier, J. André, M. Bernadet, K. Ricaud, and S. Davail, Insulin effect on lipogenesis and fat distribution in three genotypes of ducks during overfeeding, Comp Biochem Physiol A Mol Integr Physiol, vol.164, pp.499-505, 2013.

G. Guy, D. Rousselot-pailley, and D. Gourichon, Comparaison des performances de l'oie, du canard mulard et du canard de Barbarie soumis au gavage, Annales de zootechnie, pp.297-305, 1995.
URL : https://hal.archives-ouvertes.fr/hal-00889185

J. Ha, K. Guan, and J. Kim, AMPK and autophagy in glucose/glycogen metabolism

, Mol Aspects Med, vol.46, pp.46-62, 2015.

E. V. Haar, S. Lee, S. Bandhakavi, T. J. Griffin, and D. Kim, Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40, Nat Cell Biol, vol.9, p.316, 2007.

H. P. Harding, Y. Zhang, H. Zeng, I. Novoa, P. D. Lu et al., An Integrated Stress Response Regulates Amino Acid Metabolism and Resistance to Oxidative Stress, He C, Klionsky DJ. Regulation Mechanisms and Signaling Pathways of Autophagy, vol.11, pp.619-633, 2003.

, Annu Rev Genet, vol.43, pp.67-93, 2009.

F. Hérault, C. Duby, E. Baéza, and C. Diot, Adipogenic genes expression in relation to hepatic steatosis in the liver of two duck species, Animal, vol.12, pp.2571-2577, 2018.

F. Hérault, G. Saez, R. E. Mohammad, A. A. Davail, S. Chartrin et al., Liver gene expression in relation to hepatic steatosis and lipid secretion in two duck species, Anim Genet, vol.41, pp.12-20, 2009.

D. Hermier, G. Guy, S. Guillaumin, S. Davail, J. André et al., Differential channelling of liver lipids in relation to susceptibility to hepatic steatosis in two species of ducks, Comp Biochem Physiol B Biochem Mol Biol, vol.135, pp.663-675, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01702602

T. Annabelle, R. Karine, B. Marie-dominique, D. Stéphane, K. et al., Kinetics of expression of genes involved in glucose metabolism after the last meal in overfed mule ducks, Mol. Cell. Biochem, vol.430, pp.127-137, 2017.

J. Arroyo, C. Molette, F. Lavigne, C. Margetyal, O. Amador et al., Effects of dietary protein level during rearing period and age at overfeeding on magret and foie gras quality in male mule ducks, Anim. Sci. J, vol.89, pp.570-578, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02628855

B. Aryal, A. K. Singh, N. Rotllan, N. Price, and C. Fernández-hernando, MicroRNAs and lipid metabolism, Curr. Opin. Lipidol, vol.28, pp.273-280, 2017.

A. Auvergne, H. Rémignon, R. Babilé, and C. Baudonnet-lenfant, Evolution corporelle au cours du gavage chez le canard de Barbarie, Arch. Für Geflügelkd, vol.59, pp.234-240, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02704024

R. Babile, A. Auvergne, J. P. Dubois, G. Benard, and H. Manse, Reversibilite de la steatose hepatique chez l'oie. 3emes Journees de la recherche sur les Palmipedes a Foie"gras, 1998.

D. P. Bartel, MicroRNAs: Genomics, Biogenesis, Mechanism, and Function, Cell, vol.116, pp.281-297, 2004.

S. Baulande, A. Criqui, and M. Duthieuw, Les microARN circulants, une nouvelle classe de biomarqueurs pour la médecine. médecine/sciences, vol.30, pp.289-296, 2014.

G. Bénard, P. Bénard, D. Prehn, T. Bengone, J. Y. Jouglar et al., , 1998.

, Démonstration de la réversibilité de la stéatose hépatique obtenue par gavage de canards mulards, Etude Réalis. Sur, vol.3, pp.49-52

C. M. Bonnefont, C. Molette, F. Lavigne, H. Manse, C. Bravo et al., Evolution of liver fattening and foie gras technological yield during the overfeeding period in mule duck, Poult. Sci, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02627243

J. L. Cazeils, M. Bouillier-oudot, A. Auvergne, M. Candau, and R. Babile, Lipid composition of hepatocyte plasma membranes from geese overfed with corn, Lipids, vol.34, pp.937-942, 1999.

S. Cermelli, A. Ruggieri, J. A. Marrero, G. N. Ioannou, and L. Beretta, Circulating MicroRNAs in Patients with Chronic Hepatitis C and Non-Alcoholic Fatty Liver Disease, PLoS ONE, vol.6, 2011.

P. Chartrin, M. Bernadet, G. Guy, J. Mourot, J. Hocquette et al., Does overfeeding enhance genotype effects on liver ability for lipogenesis and lipid secretion in ducks?, Comp. Biochem. Physiol. A. Mol. Integr. Physiol, vol.145, pp.390-396, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02659542

P. Chartrin, K. Meteau, H. Juin, M. D. Bernadet, G. Guy et al., Effects of Intramuscular Fat Levels on Sensory Characteristics of Duck Breast Meat, Poult. Sci, vol.85, pp.914-922, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02657230

F. Chen, H. Zhang, J. Li, Y. Tian, J. Xu et al., Identification of differentially expressed miRNAs in the fatty liver of Landes goose ( Anser anser ), Sci. Rep, vol.7, pp.1-9, 2017.

J. Elmén, M. Lindow, S. Schütz, M. Lawrence, A. Petri et al., LNA-mediated microRNA silencing in non-human primates, Nature, vol.452, pp.896-899, 2008.

C. Esau, S. Davis, S. F. Murray, X. X. Yu, S. K. Pandey et al., miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting, Cell Metab, vol.3, pp.87-98, 2006.

A. Esquela-kerscher and F. J. Slack, Oncomirs -microRNAs with a role in cancer, Nat. Rev. Cancer, vol.6, pp.259-269, 2006.

J. Fantini, R. M. Epand, and F. J. Barrantes, Cholesterol-Recognition Motifs in Membrane Proteins, Direct Mechanisms in Cholesterol Modulation of Protein Function, pp.3-25, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02482669

J. Gabarrou, M. Salichon, G. Guy, and J. Blum, Hybrid ducks overfed with boiled corn develop an acute hepatic steatosis with decreased choline and polyunsaturated fatty acid level in phospholipids, Reprod. Nutr. Dev, vol.36, pp.473-484, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02688463

M. Girard, E. Jacquemin, A. Munnich, S. Lyonnet, and A. Henrion-caude, miR-122, a paradigm for the role of microRNAs in the liver, J. Hepatol, vol.48, pp.648-656, 2008.

G. Guy, D. Rousselot-pailley, and D. Gourichon, Comparaison des performances de l'oie, du canard mulard et du canard de Barbarie soumis au gavage, Annales de Zootechnie, pp.297-305, 1995.
URL : https://hal.archives-ouvertes.fr/hal-00889185

C. N. Hayes and K. Chayama, MicroRNAs as Biomarkers for Liver Disease and Hepatocellular Carcinoma, Int. J. Mol. Sci, vol.17, p.280, 2016.

J. He, W. Wang, L. Lu, Y. Tian, D. Niu et al., Analysis of miRNAs and their target genes associated with lipid metabolism in duck liver, Sci. Rep, vol.6, pp.1-9, 2016.

F. Hérault, G. Saez, E. Robert, A. A. Mohammad, S. Davail et al., Liver gene expression in relation to hepatic steatosis and lipid secretion in two duck species, Anim. Genet, vol.41, pp.12-20, 2009.

D. Hermier, A. Saadoun, M. Salichon, N. Sellier, D. Rousselot-paillet et al., Plasma lipoproteins and liver lipids in two breeds of geese with different susceptibility to hepatic steatosis: Changes induced by development and force-feeding, Lipids, vol.26, pp.331-339, 1991.
URL : https://hal.archives-ouvertes.fr/hal-02713621

D. Hermier, M. R. Salichon, G. Guy, and R. Peresson, Differential channelling of liver lipids in relation to susceptibility to hepatic steatosis in the goose, Poult. Sci, vol.78, pp.1398-1406, 1999.
URL : https://hal.archives-ouvertes.fr/hal-02688967

D. Hermier, G. Guy, S. Guillaumin, S. Davail, J. André et al., Differential channelling of liver lipids in relation to susceptibility to hepatic steatosis in two species of ducks, Comp. Biochem. Physiol. B Biochem. Mol. Biol, vol.135, pp.663-675, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01702602

M. Inui, G. Martello, and S. Piccolo, MicroRNA control of signal transduction, Nat. Rev. Mol. Cell Biol, vol.11, pp.252-263, 2010.

K. N. Ivey and D. Srivastava, MicroRNAs as Regulators of Differentiation and Cell Fate Decisions, Cell Stem Cell, vol.7, pp.36-41, 2010.

J. Krützfeldt and M. Stoffel, MicroRNAs: A new class of regulatory genes affecting metabolism, Cell Metab, vol.4, pp.9-12, 2006.

J. Krützfeldt, N. Rajewsky, R. Braich, K. G. Rajeev, T. Tuschl et al., Silencing of microRNAs in vivo with 'antagomirs, Nature, vol.438, pp.685-689, 2005.

O. F. Laterza, M. G. Scott, P. W. Garrett-engele, K. M. Korenblat, and C. M. Lockwood, Circulating miR-122 as a potential biomarker of liver disease, Biomark. Med, vol.7, pp.205-210, 2013.

R. C. Lee, R. L. Feinbaum, A. , and V. , The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell, vol.75, pp.843-854, 1993.
URL : https://hal.archives-ouvertes.fr/in2p3-00597159

A. Meerson, M. Traurig, V. Ossowski, J. M. Fleming, M. Mullins et al., , 2013.

, Human adipose microRNA-221 is upregulated in obesity and affects fat metabolism downstream of leptin and TNF-?, Diabetologia, vol.56, pp.1971-1979

J. A. Mennigen, S. Skiba-cassy, and S. Panserat, Ontogenetic expression of metabolic genes and microRNAs in rainbow trout alevins during the transition from the endogenous to the exogenous feeding period, J. Exp. Biol, vol.216, pp.1597-1608, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02646393

H. Min, A. Kapoor, M. Fuchs, F. Mirshahi, H. Zhou et al., Increased Hepatic Synthesis and Dysregulation of Cholesterol Metabolism Is Associated with the Severity of Nonalcoholic Fatty Liver Disease, Cell Metab, vol.15, pp.665-674, 2012.

T. Annabelle, R. Karine, B. Marie-dominique, D. Stéphane, K. et al., Kinetics of expression of genes involved in glucose metabolism after the last meal in overfed mule ducks, Molecular and Cellular Biochemistry, vol.430, pp.127-137, 2017.

J. Arroyo, C. Molette, F. Lavigne, C. Margetyal, O. Amador et al., Effects of dietary protein level during rearing period and age at overfeeding on magret and foie gras quality in male mule ducks, Animal Science Journal, vol.89, pp.570-578, 2018.
URL : https://hal.archives-ouvertes.fr/hal-02628855

A. Auvergne, H. Rémignon, R. Babilé, and C. Baudonnet-lenfant, Evolution corporelle au cours du gavage chez le canard de Barbarie, Archiv Für Geflügelkunde, vol.59, pp.234-240, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02704024

S. Awde, N. Marty-gasset, K. Prahkarnkaeo, R. , and H. , Relationship between Proteolytic Activities and Cooking Loss Variability in Liver Issued from Force-Fed Mule Ducks, Journal of Agricultural and Food Chemistry, vol.62, pp.3262-3268, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02636930

R. Babile, A. Auvergne, J. P. Dubois, G. Benard, and H. Manse, Reversibilite de la steatose hepatique chez l'oie. 3emes Journees de la recherche sur les Palmipedes a Foie"gras, 1998.

E. Baéza, N. Rideau, P. Chartrin, S. Davail, R. Hoo-paris et al., Canards de Barbarie, Pékin et leurs hybrides: aptitude à l'engraissement, INRA Prod. Anim, vol.18, pp.131-141, 2005.

E. Baeza, C. Marie-etancelin, S. Davail, and C. Diot, La stéatose hépatique chez les palmipèdes, INRA Productions Animales, vol.26, pp.403-414, 2013.

I. P. Barbosa-tessmann, Activation of the Human Asparagine Synthetase Gene by the Amino Acid Response and the Endoplasmic Reticulum Stress Response Pathways Occurs by Common Genomic Elements, Journal of Biological Chemistry, 2000.

A. Barthel and D. Schmoll, Novel concepts in insulin regulation of hepatic gluconeogenesis, American Journal of Physiology-Endocrinology and Metabolism, vol.285, pp.685-692, 2003.

W. B'chir, A. Maurin, V. Carraro, J. Averous, C. Jousse et al., The eIF2?/ATF4 pathway is essential for stress-induced autophagy gene expression, Nucleic Acids Research, vol.41, pp.7683-7699, 2013.

I. Belghit, S. Skiba-cassy, I. Geurden, K. Dias, A. Surget et al., Dietary methionine availability affects the main factors involved in muscle protein turnover in rainbow trout ( Oncorhynchus mykiss ), British Journal of Nutrition, vol.112, pp.493-503, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02633816

G. Bénard, P. Bénard, D. Prehn, T. Bengone, J. Y. Jouglar et al., , 1998.

, Démonstration de la réversibilité de la stéatose hépatique obtenue par gavage de canards mulards, Etude Réalisée Sur, vol.3, pp.49-52

D. Benjamin, M. Colombi, C. Moroni, and M. N. Hall, Rapamycin passes the torch: a new generation of mTOR inhibitors, Nat Rev Drug Discov, vol.10, pp.868-880, 2011.

C. F. Bento, M. Renna, G. Ghislat, C. Puri, A. Ashkenazi et al., Mammalian Autophagy: How Does It Work?, Annual Review of Biochemistry, vol.85, pp.685-713, 2016.

C. M. Bonnefont, C. Molette, F. Lavigne, H. Manse, C. Bravo et al., Evolution of liver fattening and foie gras technological yield during the overfeeding period in mule duck, Poult. Sci, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02627243

C. J. Carlson, M. F. White, and C. M. Rondinone, Mammalian target of rapamycin regulates IRS-1 serine 307 phosphorylation, Biochemical and Biophysical Research Communications, vol.316, pp.533-539, 2004.

P. Chartrin, M. Bernadet, G. Guy, J. Mourot, J. Hocquette et al., Does overfeeding enhance genotype effects on liver ability for lipogenesis and lipid secretion in ducks?, Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, vol.145, pp.390-396, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02659542

P. Codogno and A. J. Meijer, Autophagy and signaling: their role in cell survival and cell death, Cell Death & Differentiation, vol.12, pp.1509-1518, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00172272

E. Coudert, S. Crochet, E. Cailleau-audouin, T. Bordeau, A. Collin et al., Expression and ontogenesis of glucose transporters SLC2A-1, -8 and -12 in different chicken muscles, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02624243

, Actes des 11èmes

, 25 et 26 mars, Journées de la Recherche Avicole et Palmipèdes à Foie Gras, pp.819-822, 2015.

W. Dai, S. Panserat, E. Plagnes-juan, I. Seiliez, and S. Skiba-cassy, Amino Acids Attenuate Insulin Action on Gluconeogenesis and Promote Fatty Acid Biosynthesis via mTORC1 Signaling Pathway in trout Hepatocytes, Cellular Physiology and Biochemistry, vol.36, pp.1084-1100, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01901412

T. Desvignes, C. Fauvel, and J. Bobe, The nme gene family in zebrafish oogenesis and early development, Naunyn-Schmiedeberg's Archives of Pharmacology, vol.384, pp.439-449, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01205026

A. Dufner, T. , and G. , Ribosomal S6 Kinase Signaling and the Control of Translation, Experimental Cell Research, vol.253, pp.100-109, 1999.

M. Fedorova, R. C. Bollineni, and R. Hoffmann, Protein carbonylation as a major hallmark of oxidative damage: Update of analytical strategies, Mass Spectrometry Reviews, vol.33, pp.79-97, 2014.

Y. François, C. Marie-etancelin, A. Vignal, D. Viala, S. Davail et al., , 2014.

, Shows Different Metabolic States According to Its Quality Phenotype by Using a Proteomic Approach, Journal of Agricultural and Food Chemistry, vol.62, pp.7140-7150

J. Gabarrou, M. Salichon, G. Guy, and J. Blum, Hybrid ducks overfed with boiled corn develop an acute hepatic steatosis with decreased choline and polyunsaturated fatty acid level in phospholipids, Reproduction Nutrition Development, vol.36, pp.473-484, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02688463

K. Gontier, J. André, M. Bernadet, K. Ricaud, and S. Davail, Insulin effect on lipogenesis and fat distribution in three genotypes of ducks during overfeeding, Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, vol.164, pp.499-505, 2013.

Y. Gueguen, L. Ferrari, M. Souidi, A. Batt, C. Lutton et al., , 2007.

, Compared Effect of Immunosuppressive Drugs Cyclosporine A and Rapamycin on Cholesterol Homeostasis Key Enzymes CYP27A1 and HMG-CoA Reductase, Basic & Clinical Pharmacology & Toxicology, vol.100, pp.392-397

G. Guy, D. Rousselot-pailley, and D. Gourichon, Comparaison des performances de l'oie, du canard mulard et du canard de Barbarie soumis au gavage, Annales de Zootechnie, pp.297-305, 1995.
URL : https://hal.archives-ouvertes.fr/hal-00889185

E. V. Haar, S. Lee, S. Bandhakavi, T. J. Griffin, and D. Kim, Insulin signalling to mTOR mediated by the Akt/PKB substrate PRAS40, Nature Cell Biology, vol.9, p.316, 2007.

H. P. Harding, Y. Zhang, H. Zeng, I. Novoa, P. D. Lu et al., An Integrated Stress Response Regulates Amino Acid Metabolism and Resistance to Oxidative Stress, Molecular Cell, vol.11, pp.619-633, 2003.

N. Hay and N. Sonenberg, Upstream and downstream of mTOR, Genes Dev, vol.18, pp.1926-1945, 2004.

F. Hérault, G. Saez, E. Robert, A. A. Mohammad, S. Davail et al., Liver gene expression in relation to hepatic steatosis and lipid secretion in two duck species, Animal Genetics, vol.41, pp.12-20, 2009.

D. Hermier, A. Saadoun, M. Salichon, N. Sellier, D. Rousselot-paillet et al., Plasma lipoproteins and liver lipids in two breeds of geese with different susceptibility to hepatic steatosis: Changes induced by development and force-feeding, Lipids, vol.26, pp.331-339, 1991.
URL : https://hal.archives-ouvertes.fr/hal-02713621

D. Hermier, G. Guy, S. Guillaumin, S. Davail, J. André et al., Differential channelling of liver lipids in relation to susceptibility to hepatic steatosis in two species of ducks, Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology, vol.135, pp.663-675, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01702602

M. S. Kilberg, M. Balasubramanian, L. Fu, and J. Shan, The Transcription Factor Network Associated With the Amino Acid Response in Mammalian Cells, Advances in Nutrition, vol.3, pp.295-306, 2012.

D. J. Klionsky, K. Abdelmohsen, A. Abe, M. J. Abedin, H. Abeliovich et al., Guidelines for the use and interpretation of assays for monitoring autophagy, Autophagy, vol.12, pp.1-222, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02641288

D. J. Kostyniuk, B. M. Culbert, J. A. Mennigen, and K. M. Gilmour, Social status affects lipid metabolism in rainbow trout, Oncorhynchus mykiss, American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, vol.315, pp.241-255, 2018.

V. J. Lavallard and P. Gual, Autophagy and Non-Alcoholic Fatty Liver Disease, BioMed Research International, vol.2014, pp.1-13, 2014.

G. A. Leveille, D. R. Romsos, Y. Yeh, and E. K. Hea, Lipid Biosynthesis in the Chick. A Consideration of Site of Synthesis, Influence of Diet and Possible Regulatory Mechanisms, Poult Sci, vol.54, pp.1075-1093, 1975.

L. Marandel, C. Labbe, J. Bobe, H. Jammes, J. Lareyre et al., Do not put all teleosts in one net: Focus on the sox2 and pou2 genes, Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology, vol.164, pp.69-79, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01000747

N. Mizushima and M. Komatsu, Autophagy: Renovation of Cells and Tissues, Cell, vol.147, pp.728-741, 2011.

M. R. Munday, D. G. Campbell, D. Carling, and D. G. Hardie, Identification by amino acid sequencing of three major regulatory phosphorylation sites on rat acetyl-CoA carboxylase, European Journal of Biochemistry, vol.175, pp.331-338, 1988.

M. E. Patti, E. Brambilla, L. Luzi, E. J. Landaker, and C. R. Kahn, Bidirectional modulation of insulin action by amino acids, 1998.

B. Pilo, G. , and J. C. , Diurnal and seasonal variation in liver glycogen and fat in relation to metabolic status of liver and m. pectoralis in the migratory starling, Sturnus roseus, wintering in India, Comparative Biochemistry and Physiology. A, Comparative Physiology, vol.74, pp.601-604, 1983.

T. Pioche, F. Skiba, M. Bernadet, I. Seiliez, W. Massimino et al., Kinetic study of the expression of genes related to hepatic steatosis, global intermediate metabolism and cellular stress during overfeeding in mule ducks, BioRxiv, vol.690156, 2019.

M. Proszkowiec-weglarz, M. P. Richards, B. D. Humphrey, R. W. Rosebrough, and J. P. Mcmurtry, AMP-activated protein kinase and carbohydrate response element binding protein: A study of two potential regulatory factors in the hepatic lipogenic program of broiler chickens, Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology, vol.154, pp.68-79, 2009.

A. Rashidpour, J. I. Silva-marrero, L. Seguí, I. V. Baanante, and I. Metón, Metformin counteracts glucose-dependent lipogenesis and impairs transdeamination in the liver of gilthead sea bream (Sparus aurata), American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, vol.316, pp.265-273, 2019.

C. N. Roybal, L. A. Hunsaker, O. Barbash, D. L. Jagt, and S. F. Abcouwer, The Oxidative Stressor Arsenite Activates Vascular Endothelial Growth Factor mRNA Transcription by an ATF4-dependent Mechanism, Journal of Biological Chemistry, vol.280, pp.20331-20339, 2005.

A. Saadoun and B. Leclercq, In Vivo Lipogenesis of Genetically Lean and Fat Chickens: Effects of Nutritional State and Dietary Fat, J Nutr, vol.117, pp.428-435, 1987.
URL : https://hal.archives-ouvertes.fr/hal-02720398

G. Saez, E. Baéza, S. Davail, D. Durand, D. Bauchart et al., Is the hepatic metabolism of glucose and linoleic acid influenced by species in overfed ducks?, Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology, vol.151, pp.576-581, 2008.
URL : https://hal.archives-ouvertes.fr/hal-02668078

H. Sano, S. Kane, E. Sano, C. P. M??inea, J. M. Asara et al., Insulin-stimulated Phosphorylation of a Rab GTPase-activating Protein Regulates GLUT4 Translocation, J. Biol. Chem, vol.278, pp.14599-14602, 2003.

Y. Seki, K. Sato, T. Kono, H. Abe, and Y. Akiba, Broiler chickens (Ross strain) lack insulin-responsive glucose transporter GLUT4 and have GLUT8 cDNA, General and Comparative Endocrinology, vol.133, pp.80-87, 2003.

Y. J. Suzuki, M. Carini, and D. A. Butterfield, Protein Carbonylation, Antioxid Redox Signal, vol.12, pp.323-325, 2010.

A. Tavernier, S. Davail, K. Ricaud, M. Bernadet, and K. Gontier, Genes involved in the establishment of hepatic steatosis in Muscovy, Pekin and mule ducks, Molecular and Cellular Biochemistry, vol.424, pp.147-161, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01606275

A. Tavernier, R. Karine, B. Marie-dominique, G. Karine, and D. Stéphane, Pre-and post-prandial expression of genes involved in lipid metabolism at the end of the overfeeding period of mule ducks, Molecular and Cellular Biochemistry, vol.438, pp.111-121, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01773149

L. A. Adams, Nonalcoholic fatty liver disease, Can. Med. Assoc. J, vol.172, pp.899-905, 2005.

T. Annabelle, R. Karine, B. Marie-dominique, D. Stéphane, K. et al., Kinetics of expression of genes involved in glucose metabolism after the last meal in overfed mule ducks, Mol. Cell. Biochem, vol.430, pp.127-137, 2017.

M. Arrese, A. Eguchi, and A. E. Feldstein, Circulating microRNAs: Emerging Biomarkers of Liver Disease, Semin. Liver Dis, vol.35, pp.43-54, 2015.

J. D. Arroyo, J. R. Chevillet, E. M. Kroh, I. K. Ruf, C. C. Pritchard et al., Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma, Proc. Natl. Acad. Sci, vol.108, pp.5003-5008, 2011.

A. Auvergne, H. Rémignon, R. Babilé, and C. Baudonnet-lenfant, Evolution corporelle au cours du gavage chez le canard de Barbarie, Arch. Für Geflügelkd, vol.59, pp.234-240, 1995.
URL : https://hal.archives-ouvertes.fr/hal-02704024

J. Averous, A. Bruhat, C. Jousse, V. Carraro, G. Thiel et al., Induction of CHOP Expression by Amino Acid Limitation Requires Both ATF4 Expression and ATF2 Phosphorylation, J. Biol. Chem, vol.279, pp.5288-5297, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02683570

J. Avruch, Insulin signal transduction through protein kinase cascades, pp.31-48, 1998.

R. Babile, A. Auvergne, J. P. Dubois, G. Benard, and H. Manse, Reversibilite de la steatose hepatique chez l'oie. 3emes Journees de la recherche sur les Palmipedes a Foie"gras, 1998.

E. Baéza, N. Rideau, P. Chartrin, S. Davail, R. Hoo-paris et al., Canards de Barbarie, Pékin et leurs hybrides: aptitude à l'engraissement, INRA Prod. Anim, vol.18, pp.131-141, 2005.

E. Baeza, C. Marie-etancelin, S. Davail, and C. Diot, La stéatose hépatique chez les palmipèdes, INRA Prod. Anim, vol.26, pp.403-414, 2013.

E. Baéza, C. Marie-etancelin, S. Davail, and C. Diot, La stéatose hépatique chez les palmipèdes, INRA Prod Anim, vol.26, pp.403-414, 2013.

X. Bai and Y. Jiang, Key factors in mTOR regulation, Cell. Mol. Life Sci, vol.67, pp.239-253, 2010.

J. Ban, M. Lee, W. Im, K. , and M. , Low pH increases the yield of exosome isolation, Biochem. Biophys. Res. Commun, vol.461, pp.76-79, 2015.

D. Banerjee and C. M. Redman, Biosynthesis of high density lipoprotein by chicken liver: nature of nascent intracellular high density lipoprotein, J. Cell Biol, vol.96, pp.651-660, 1983.

I. P. Barbosa-tessmann, Activation of the Human Asparagine Synthetase Gene by the Amino Acid Response and the Endoplasmic Reticulum Stress Response Pathways Occurs by Common Genomic Elements, J. Biol. Chem, 2000.

F. Bardag-gorce, T. Francis, L. Nan, J. Li, Y. He-lue et al., Modifications in P62 occur due to proteasome inhibition in alcoholic liver disease, Life Sci, vol.77, pp.2594-2602, 2005.

D. P. Bartel, MicroRNAs: Genomics, Biogenesis, Mechanism, and Function, Cell, vol.116, pp.281-297, 2004.

C. Baudonnet-lenfant, Facteurs de variation de la composition biochimique et de la qualité technologique des foies gras de canards, 1993.

S. Baulande, A. Criqui, and M. Duthieuw, Les microARN circulants, une nouvelle classe de biomarqueurs pour la médecine. médecine/sciences, vol.30, pp.289-296, 2014.

W. B'chir, A. Maurin, V. Carraro, J. Averous, C. Jousse et al., The eIF2?/ATF4 pathway is essential for stress-induced autophagy gene expression, Nucleic Acids Res, vol.41, pp.7683-7699, 2013.

G. Benard and C. Labie, Evolution histologique du foie des Palmipedes au cours du gavage. 3emes Journees de la``recherche sur les Palmipedes a Foie gras, Bordx. FranceOctober, pp.27-28, 1998.

G. Bénard, P. Bénard, D. Prehn, T. Bengone, J. Y. Jouglar et al., , 1998.

, Démonstration de la réversibilité de la stéatose hépatique obtenue par gavage de canards mulards, Etude Réalis. Sur, vol.3, pp.49-52

G. Benard, T. Bengone, D. Prehn, S. Durand, C. Labie et al., Contribution à l'étude de la physiologie du canard en gavage : étude de la stéatose hépatique, Bull. Académie Vét. Fr, p.43, 2006.

H. Berradi, G. Guy, and N. Rideau, A glucokinase-like enzyme induced in Mule duck livers by overfeeding, Poult. Sci, vol.83, pp.161-168, 2004.
URL : https://hal.archives-ouvertes.fr/hal-02669258

C. M. Bonnefont, C. Molette, F. Lavigne, H. Manse, C. Bravo et al., Evolution of liver fattening and foie gras technological yield during the overfeeding period in mule duck, Poult. Sci, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02627243

E. Bourneuf, F. Hérault, C. Chicault, W. Carré, S. Assaf et al., Microarray analysis of differential gene expression in the liver of lean and fat chickens, Gene, vol.372, pp.162-170, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00085700

P. Boya, F. Reggiori, and P. Codogno, Emerging regulation and functions of autophagy, Nat. Cell Biol, vol.15, pp.713-720, 2013.

J. Brennecke, A. Stark, R. B. Russell, and S. M. Cohen, Principles of MicroRNA-Target Recognition, PLOS Biol, vol.3, p.85, 2005.

J. D. Browning and J. D. Horton, Molecular mediators of hepatic steatosis and liver injury, 2004.

A. Bruhat, C. Jousse, V. Carraro, A. Maurin, C. Chaveroux et al., L'activation de la voie eIF2?-ATF4, vol.31, pp.1057-1060, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01595102

J. Brun, M. M. Richard, C. Marie-etancelin, R. Rouvier, and C. Larzul, Le canard mulard: déterminisme génétique d'un hybride intergénérique, Prod. Anim.-PARIS-Inst. Natl. Rech. Agron, vol.18, p.295, 2005.

E. M. Brunt, Pathology of nonalcoholic fatty liver disease, Nat. Rev. Gastroenterol. Hepatol, vol.7, pp.195-203, 2010.

C. D. Byrne and G. Targher, NAFLD: A multisystem disease, J. Hepatol, vol.62, pp.47-64, 2015.

L. Cabon, A. Martinez-torres, and S. A. Susin, La mort cellulaire programmée ne manque pas de vocabulaire. médecine/sciences, vol.29, pp.1117-1124, 2013.

S. H. Caldwell, R. H. Swerdlow, E. M. Khan, J. C. Iezzoni, E. E. Hespenheide et al., Mitochondrial abnormalities in non-alcoholic steatohepatitis, J. Hepatol, vol.31, pp.430-434, 1999.

J. Capeau, Voies de signalisation de l'insuline : mécanismes affectés dans l'insulinorésistance. médecine/sciences 19, pp.834-839, 2003.

C. J. Carlson, M. F. White, and C. M. Rondinone, Mammalian target of rapamycin regulates IRS-1 serine 307 phosphorylation, Biochem. Biophys. Res. Commun, vol.316, pp.533-539, 2004.

J. L. Cazeils, M. Bouillier-oudot, A. Auvergne, M. Candau, and R. Babile, Lipid composition of hepatocyte plasma membranes from geese overfed with corn, Lipids, vol.34, pp.937-942, 1999.

N. Chalasani, M. Deeg, and D. Crabb, Systemic Levels of Lipid Peroxidation and Its Metabolic and Dietary Correlates in Patients with Nonalcoholic Steatohepatitis, Am. J. Gastroenterol, vol.99, pp.1497-1502, 2004.

P. Chartrin, M. Bernadet, G. Guy, J. Mourot, J. Hocquette et al., Does overfeeding enhance genotype effects on liver ability for lipogenesis and lipid secretion in ducks?, Comp. Biochem. Physiol. A. Mol. Integr. Physiol, vol.145, pp.390-396, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02659542

P. Chartrin, M. Bernadet, G. Guy, J. Mourot, J. Hocquette et al., Does overfeeding enhance genotype effects on energy metabolism and lipid deposition in breast muscle of ducks?, Comp. Biochem. Physiol. A. Mol. Integr. Physiol, vol.145, pp.413-418, 2006.
URL : https://hal.archives-ouvertes.fr/hal-02657325

F. Chen, H. Zhang, J. Li, Y. Tian, J. Xu et al., Identification of differentially expressed miRNAs in the fatty liver of Landes goose ( Anser anser ), Sci. Rep, vol.7, pp.1-9, 2017.

X. Chen, Y. Ba, L. Ma, X. Cai, Y. Yin et al., Characterization of microRNAs in serum: a novel class of biomarkers for diagnosis of cancer and other diseases, Cell Res, vol.18, pp.997-1006, 2008.

X. Chen, H. Liang, J. Zhang, K. Zen, and C. Zhang, Secreted microRNAs: a new form of intercellular communication, Trends Cell Biol, vol.22, pp.125-132, 2012.

Y. Chen, D. Gao, and L. Huang, In vivo delivery of miRNAs for cancer therapy: Challenges and strategies, Adv. Drug Deliv. Rev, vol.81, pp.128-141, 2015.

S. W. Cheng, L. G. Fryer, D. Carling, and P. R. Shepherd, Thr2446 Is a Novel Mammalian Target of Rapamycin (mTOR) Phosphorylation Site Regulated by Nutrient Status, J. Biol. Chem, vol.279, pp.15719-15722, 2004.

O. Cheung, P. Puri, C. Eicken, M. J. Contos, F. Mirshahi et al., Nonalcoholic steatohepatitis is associated with altered hepatic MicroRNA expression, Hepatology, vol.48, pp.1810-1820, 2008.

J. R. Chevillet, Q. Kang, I. K. Ruf, H. A. Briggs, L. N. Vojtech et al., Quantitative and stoichiometric analysis of the microRNA content of exosomes, Proc. Natl. Acad. Sci, vol.111, pp.14888-14893, 2014.

S. S. Chim, T. K. Shing, E. C. Hung, T. Leung, T. Lau et al., Detection and Characterization of Placental MicroRNAs in Maternal Plasma, Clin. Chem, vol.54, pp.482-490, 2008.

S. Chitturi, S. Abeygunasekera, G. C. Farrell, J. Holmes-walker, J. M. Hui et al., NASH and insulin resistance: Insulin hypersecretion and specific association with the insulin resistance syndrome, Hepatology, vol.35, pp.373-379, 2002.

P. Codogno and A. J. Meijer, Autophagy and signaling: their role in cell survival and cell death, Cell Death Differ, vol.12, pp.1509-1518, 2005.
URL : https://hal.archives-ouvertes.fr/inserm-00172272

J. Copp, G. Manning, and T. Hunter, TORC-Specific Phosphorylation of Mammalian Target of Rapamycin (mTOR): Phospho-Ser 2481 Is a Marker for Intact mTOR Signaling Complex 2, Cancer Res, vol.69, pp.1821-1827, 2009.

F. Corpet, Multiple sequence alignment with hierarchical clustering, Nucleic Acids Res, vol.16, pp.10881-10890, 1988.
URL : https://hal.archives-ouvertes.fr/hal-02728178

E. Coudert, S. Crochet, E. Cailleau-audouin, T. Bordeau, A. Collin et al., Expression and ontogenesis of glucose transporters SLC2A-1, -8 and -12 in different chicken muscles, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02624243

, Actes 11èmes Journ. Rech. Avic. Palmipèdes À Foie Gras Tours Fr. 25 26 Mars, pp.819-822, 2015.

E. Coudert, G. Pascal, J. Dupont, J. Simon, E. Cailleau-audouin et al., Phylogenesis and Biological Characterization of a New Glucose Transporter in the Chicken (Gallus gallus, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01594233

A. Couzinet, Z. Hérincs, and A. Hueber, Régulation de la mort cellulaire programmée : vers une conception plus dynamique. médecine/sciences, vol.18, pp.841-852, 2002.

S. J. Crozier, S. R. Kimball, S. W. Emmert, J. C. Anthony, J. et al., Oral Leucine Administration Stimulates Protein Synthesis in Rat Skeletal Muscle, J. Nutr, vol.135, pp.376-382, 2005.

A. M. Cuervo, Autophagy: Many paths to the same end, Mol. Cell. Biochem, vol.263, pp.55-72, 2004.

A. M. Cuervo, E. Bergamini, U. T. Brunk, W. Dröge, M. Ffrench et al., , 2005.

, Autophagy and Aging: The Importance of Maintaining, Clean" Cells. Autophagy, vol.1, pp.131-140

W. Dai, S. Panserat, E. Plagnes-juan, I. Seiliez, and S. Skiba-cassy, Amino Acids Attenuate Insulin Action on Gluconeogenesis and Promote Fatty Acid Biosynthesis via mTORC1 Signaling Pathway in trout Hepatocytes, Cell. Physiol. Biochem, vol.36, pp.1084-1100, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01901412

S. Davail, N. Rideau, G. Guy, J. André, D. Hermier et al., Hormonal and metabolic responses to overfeeding in three genotypes of ducks, Comp. Biochem. Physiol. A. Mol. Integr. Physiol, vol.134, pp.707-715, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01702605

S. Davail, N. Rideau, G. Guy, J. André, D. Hermier et al., Hormonal and metabolic responses to overfeeding in three genotypes of ducks, Comp. Biochem. Physiol. A. Mol. Integr. Physiol, vol.134, pp.707-715, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01702605

R. Dentin, F. Benhamed, J. Pégorier, F. Foufelle, B. Viollet et al., Polyunsaturated fatty acids suppress glycolytic and lipogenic genes through the inhibition of ChREBP nuclear protein translocation, 2005.

S. Desagher and J. Martinou, Mitochondria as the central control point of apoptosis, Trends Cell Biol, vol.10, pp.369-377, 2000.

T. Desvignes, C. Fauvel, and J. Bobe, The nme gene family in zebrafish oogenesis and early development, Naunyn. Schmiedebergs Arch. Pharmacol, vol.384, pp.439-449, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01205026

J. M. Dietschy, S. D. Turley, and D. K. Spady, Role of liver in the maintenance of cholesterol and low density lipoprotein homeostasis in different animal species, including humans, J. Lipid Res, vol.34, pp.1637-1659, 1993.

V. R. Dolnik and T. I. Blyumental, Autumnal Premigratory and Migratory Periods in the Chaffinch (Fringilla coelebs coelebs) and Some Other Temperate-Zone Passerine Birds, The Condor, vol.69, pp.435-468, 1967.

J. Dong, H. Qiu, M. Garcia-barrio, J. Anderson, and A. G. Hinnebusch, Uncharged tRNA Activates GCN2 by Displacing the Protein Kinase Moiety from a Bipartite tRNA-Binding Domain, Mol. Cell, vol.6, pp.269-279, 2000.

A. Dufner, T. , and G. , Ribosomal S6 Kinase Signaling and the Control of Translation, Exp. Cell Res, vol.253, pp.100-109, 1999.

C. Duve and R. Wattiaux, Functions of lysosomes, Ann Rev Physiol, vol.28, pp.435-492, 1966.

S. Eaton, Control of mitochondrial ?-oxidation flux, Prog. Lipid Res, vol.41, pp.197-239, 2002.

J. Elmén, M. Lindow, A. Silahtaroglu, M. Bak, M. Christensen et al., Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver, Nucleic Acids Res, vol.36, pp.1153-1162, 2008.

C. Esau, S. Davis, S. F. Murray, X. X. Yu, S. K. Pandey et al., miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting, Cell Metab, vol.3, pp.87-98, 2006.

A. Esquela-kerscher and F. J. Slack, Oncomirs -microRNAs with a role in cancer, Nat. Rev. Cancer, vol.6, pp.259-269, 2006.

J. Estaquier, F. Vallette, J. Vayssiere, and B. Mignotte, The Mitochondrial Pathways of Apoptosis, Advances in Mitochondrial Medicine, pp.157-183, 2012.

J. Fantini, R. M. Epand, and F. J. Barrantes, Cholesterol-Recognition Motifs in Membrane Proteins, Direct Mechanisms in Cholesterol Modulation of Protein Function, pp.3-25, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02482669

A. Farhat and E. R. Chavez, Comparative performance, blood chemistry, and carcass composition of two lines of Pekin ducks reared mixed or separated by sex, Poult. Sci, vol.79, pp.460-465, 2000.

X. Fernandez, M. Bouillier-oudot, C. Molette, M. D. Bernadet, and H. Manse, Duration of transport and holding in lairage at constant postprandial delay to slaughter--Effects on fatty liver and breast muscle quality in mule ducks, Poult. Sci, vol.90, pp.2360-2369, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02652778

X. Fernandez, G. Guy, J. B. Laverze, C. Bonnefont, C. Knudsen et al., A kinetic study of the natural induction of liver steatosis in Greylag Landaise geese: the role of hyperphagia 1, vol.10, pp.1288-1295, 2016.

P. F. Finn and J. F. Dice, Proteolytic and lipolytic responses to starvation, Nutrition, vol.22, pp.830-844, 2006.

M. Flamment and F. Foufelle, Le stress du réticulum endoplasmique : de la physiologie à la pathogenèse du diabète de type 2. médecine/sciences, vol.29, pp.756-764, 2013.

G. B. Fleming, Carbohydrate metabolism in ducks, J. Physiol, vol.53, pp.236-246, 1919.

E. S. Ford, W. H. Giles, and W. H. Dietz, Prevalence of the Metabolic Syndrome Among US Adults: Findings From the Third National Health and Nutrition Examination Survey, JAMA, vol.287, pp.356-359, 2002.

F. Foufelle and P. Ferré, La réponse UPR: Son rôle physiologique et physiopathologique. médecine/sciences 23, pp.291-296, 2007.

Y. François, C. Marie-etancelin, A. Vignal, D. Viala, S. Davail et al., , 2014.

, Shows Different Metabolic States According to Its Quality Phenotype by Using a Proteomic Approach, J. Agric. Food Chem, vol.62, pp.7140-7150

Y. François, C. Marie-etancelin, A. Vignal, D. Viala, S. Davail et al., , 2014.

, Shows Different Metabolic States According to Its Quality Phenotype by Using a Proteomic Approach, J. Agric. Food Chem, vol.62, pp.7140-7150

R. C. Friedman, K. K. Farh, .. Burge, C. B. Bartel, and D. P. , Most mammalian mRNAs are conserved targets of microRNAs, Genome Res, vol.19, pp.92-105, 2009.

J. Gabarrou, M. Salichon, G. Guy, and J. Blum, Hybrid ducks overfed with boiled corn develop an acute hepatic steatosis with decreased choline and polyunsaturated fatty acid level in phospholipids, Reprod. Nutr. Dev, vol.36, pp.473-484, 1996.
URL : https://hal.archives-ouvertes.fr/hal-02688463

J. Geng and D. J. Klionsky, The Golgi as a potential membrane source for autophagy, Autophagy, vol.6, pp.950-951, 2010.

M. Girard, E. Jacquemin, A. Munnich, S. Lyonnet, and A. Henrion-caude, miR-122, a paradigm for the role of microRNAs in the liver, J. Hepatol, vol.48, pp.648-656, 2008.

L. Goedeke and C. Fernández-hernando, Regulation of cholesterol homeostasis, Cell. Mol. Life Sci, vol.69, pp.915-930, 2012.

I. J. Goldberg, Lipoprotein lipase and lipolysis: central roles in lipoprotein metabolism and atherogenesis, J. Lipid Res, vol.37, pp.693-707, 1996.

J. L. Goldstein and M. S. Brown, The LDL Receptor, Arterioscler. Thromb. Vasc. Biol, vol.29, pp.431-438, 2009.

K. Gontier, J. André, M. Bernadet, K. Ricaud, and S. Davail, Insulin effect on lipogenesis and fat distribution in three genotypes of ducks during overfeeding, Comp. Biochem. Physiol. A. Mol. Integr. Physiol, vol.164, pp.499-505, 2013.

J. Gonzalez, H. Düttmann, and M. Wink, Phylogenetic relationships based on two mitochondrial genes and hybridization patterns in Anatidae, J. Zool, vol.279, pp.310-318, 2009.

. González-rodríguez, R. Mayoral, N. Agra, M. P. Valdecantos, V. Pardo et al., Impaired autophagic flux is associated with increased endoplasmic reticulum stress during the development of NAFLD, Cell Death Dis, vol.5, pp.1179-1179, 2014.

A. G. Goodridge, D. W. Back, S. B. Wilson, and M. J. Goldman, Regulation of Genes for Enzymes Involved in Fatty Acid Synthesisa, Ann. N. Y. Acad. Sci, vol.478, pp.46-62, 1986.

Y. Gosmain, N. Dif, V. Berbe, E. Loizon, J. Rieusset et al., Regulation of SREBP-1 expression and transcriptional action on HKII and FAS genes during fasting and refeeding in rat tissues, J. Lipid Res, vol.46, pp.697-705, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02048999

D. R. Green, The End and After: How Dying Cells Impact the Living Organism, Immunity, vol.35, pp.441-444, 2011.

Y. Gueguen, L. Ferrari, M. Souidi, A. Batt, C. Lutton et al., , 2007.

, Compared Effect of Immunosuppressive Drugs Cyclosporine A and Rapamycin on Cholesterol Homeostasis Key Enzymes CYP27A1 and HMG-CoA Reductase, Basic Clin. Pharmacol. Toxicol, vol.100, pp.392-397

F. Guo and D. R. Cavener, The GCN2 eIF2? Kinase Regulates Fatty-Acid Homeostasis in the Liver during Deprivation of an Essential Amino Acid, Cell Metab, vol.5, pp.103-114, 2007.

J. Guo, G. Shu, L. Zhou, X. Zhu, W. Liao et al., Selective transport of long-chain fatty acids by FAT/CD36 in skeletal muscle of broilers, vol.7, pp.422-429, 2013.

G. Guy, D. Rousselot-pailley, and D. Gourichon, Comparaison des performances de l'oie, du canard mulard et du canard de Barbarie soumis au gavage, Ann Zootech, vol.44, pp.297-305, 1995.
URL : https://hal.archives-ouvertes.fr/hal-00889185

G. Guy, L. Fortun-lamothe, G. Bénard, and X. Fernandez, Natural induction of spontaneous liver steatosis in Greylag Landaise geese (Anser anser), J. Anim. Sci, vol.91, pp.455-464, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02650256

J. Ha, K. Guan, K. , and J. , AMPK and autophagy in glucose/glycogen metabolism, Mol. Aspects Med, vol.46, pp.46-62, 2015.

M. E. Haas, A. D. Attie, and S. B. Biddinger, The regulation of ApoB metabolism by insulin, Trends Endocrinol. Metab, vol.24, pp.391-397, 2013.

T. Hajri and N. A. Abumrad, FATTY ACID TRANSPORT ACROSS MEMBRANES: Relevance to Nutrition and Metabolic Pathology, Annu. Rev. Nutr, vol.22, pp.383-415, 2002.

B. E. Halliwell, Lipid peroxidation: a radical chain reaction. Free Radic, Biol. Med, 1989.

J. Han and R. J. Kaufman, The role of ER stress in lipid metabolism and lipotoxicity, 2016.

, Lipid Res, vol.57, pp.1329-1338

H. P. Harding, Y. Zhang, H. Zeng, I. Novoa, P. D. Lu et al., An Integrated Stress Response Regulates Amino Acid Metabolism and Resistance to Oxidative Stress, Mol. Cell, vol.11, pp.619-633, 2003.

N. Hay and N. Sonenberg, Upstream and downstream of mTOR, Genes Dev, vol.18, pp.1926-1945, 2004.

C. N. Hayes and K. Chayama, MicroRNAs as Biomarkers for Liver Disease and Hepatocellular Carcinoma, Int. J. Mol. Sci, vol.17, p.280, 2016.

J. Hayes, P. P. Peruzzi, and S. Lawler, MicroRNAs in cancer: biomarkers, functions and therapy, Trends Mol. Med, vol.20, pp.460-469, 2014.

R. L. Hazelwood and F. W. Lorenz, Effects of fasting and insulin on carbohydrate metabolism of the domestic fowl, Am. J. Physiol.-Leg. Content, 1959.

C. He and D. J. Klionsky, Regulation Mechanisms and Signaling Pathways of Autophagy, Annu. Rev. Genet, vol.43, pp.67-93, 2009.

J. He, Y. Tian, J. Li, J. Shen, Z. Tao et al., Expression pattern of L-FABP gene in different tissues and its regulation of fat metabolism-related genes in duck, Mol. Biol. Rep, vol.40, pp.189-195, 2013.

J. He, W. Wang, L. Lu, Y. Tian, D. Niu et al., Analysis of miRNAs and their target genes associated with lipid metabolism in duck liver, Sci. Rep, vol.6, pp.1-9, 2016.

J. He, Y. Tian, Y. Zhao, Y. Liu, Z. Tao et al., MiR-144 affects fatty acid composition by regulating ELOVL6 expression in duck hepatocytes, Cell Biol. Int, vol.41, pp.691-696, 2017.

F. Hérault, G. Saez, E. Robert, A. A. Mohammad, S. Davail et al., Liver gene expression in relation to hepatic steatosis and lipid secretion in two duck species, Anim. Genet, vol.41, pp.12-20, 2009.

D. Hermier, A. Saadoun, M. Salichon, N. Sellier, D. Rousselot-paillet et al., Plasma lipoproteins and liver lipids in two breeds of geese with different susceptibility to hepatic steatosis: Changes induced by development and force-feeding, Lipids, vol.26, pp.331-339, 1991.
URL : https://hal.archives-ouvertes.fr/hal-02713621

D. Hermier, M. R. Salichon, G. Guy, R. Peresson, J. Mourot et al., La stéatose hépatique des palmipèdes gavés : bases métaboliques et sensibilité génétique, 1999.

D. Hermier, G. Guy, S. Guillaumin, S. Davail, J. André et al., Differential channelling of liver lipids in relation to susceptibility to hepatic steatosis in two species of ducks, Comp. Biochem. Physiol. B Biochem. Mol. Biol, vol.135, pp.663-675, 2003.
URL : https://hal.archives-ouvertes.fr/hal-01702602

J. Hu, Y. Xu, J. Hao, S. Wang, C. Li et al., MiR-122 in hepatic function and liver diseases, Protein Cell, vol.3, pp.364-371, 2012.

B. D. Humphrey, C. B. Stephensen, C. C. Calvert, and K. C. Klasing, Glucose and cationic amino acid transporter expression in growing chickens, 2004.

. Comp, Biochem. Physiol. A. Mol. Integr. Physiol, vol.138, pp.515-525

M. M. Hussain, P. Rava, M. Walsh, M. Rana, and J. Iqbal, , 2012.

K. Iizuka, R. K. Bruick, G. Liang, J. D. Horton, and K. Uyeda, Deficiency of carbohydrate response element-binding protein (ChREBP) reduces lipogenesis as well as glycolysis, Proc. Natl. Acad. Sci, vol.101, pp.7281-7286, 2004.

D. Iliopoulos, K. Drosatos, Y. Hiyama, I. J. Goldberg, and V. I. Zannis, MicroRNA-370 controls the expression of MicroRNA-122 and Cpt1? and affects lipid metabolism, J. Lipid Res, vol.51, pp.1513-1523, 2010.

M. Inui, G. Martello, and S. Piccolo, MicroRNA control of signal transduction, Nat. Rev. Mol. Cell Biol, vol.11, pp.252-263, 2010.

S. Ishii, K. Iizuka, B. C. Miller, and K. Uyeda, Carbohydrate response element binding protein directly promotes lipogenic enzyme gene transcription, Proc. Natl. Acad. Sci, vol.101, pp.15597-15602, 2004.

R. Iurlaro and C. Muñoz-pinedo, Cell death induced by endoplasmic reticulum stress, 2016.

, FEBS J, vol.283, pp.2640-2652

K. N. Ivey and D. Srivastava, MicroRNAs as Regulators of Differentiation and Cell Fate Decisions, Cell Stem Cell, vol.7, pp.36-41, 2010.

J. Iwata, J. Ezaki, M. Komatsu, S. Yokota, T. Ueno et al., Excess Peroxisomes Are Degraded by Autophagic Machinery in Mammals, J. Biol. Chem, vol.281, pp.4035-4041, 2006.

H. B. Jefferies, S. Fumagalli, P. B. Dennis, C. Reinhard, R. B. Pearson et al., Rapamycin suppresses 5?TOP mRNA translation through inhibition of p70s6k, EMBO J, vol.16, pp.3693-3704, 1997.

S. D. Jordan, M. Krüger, D. M. Willmes, N. Redemann, F. T. Wunderlich et al., Obesity-induced overexpression of miRNA-143 inhibits insulin-stimulated AKT activation and impairs glucose metabolism, Nat. Cell Biol, vol.13, pp.434-446, 2011.

M. S. Kilberg, J. Shan, and N. Su, ATF4-dependent transcription mediates signaling of amino acid limitation, Trends Endocrinol. Metab, vol.20, pp.436-443, 2009.

M. S. Kilberg, M. Balasubramanian, L. Fu, and J. Shan, The Transcription Factor Network Associated With the Amino Acid Response in Mammalian Cells, Adv. Nutr, vol.3, pp.295-306, 2012.

I. Kim, S. Rodriguez-enriquez, and J. J. Lemasters, Selective degradation of mitochondria by mitophagy, Arch. Biochem. Biophys, vol.462, pp.245-253, 2007.

J. Kim, M. Kundu, B. Viollet, and K. Guan, AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1, Nat. Cell Biol, vol.13, pp.132-141, 2011.

S. Kimball, J. , and L. , Control of protein synthesis by amino acid availability, Curr. Opin. Clin. Nutr. Metab. Care, vol.5, pp.63-67, 2002.

D. J. Klionsky, The molecular machinery of autophagy: unanswered questions, J. Cell Sci, vol.118, pp.7-18, 2005.

D. J. Klionsky, K. Abdelmohsen, A. Abe, M. J. Abedin, H. Abeliovich et al., Guidelines for the use and interpretation of assays for monitoring autophagy, Autophagy, vol.12, pp.1-222, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02641288

D. J. Klionsky, K. Abdelmohsen, A. Abe, M. J. Abedin, H. Abeliovich et al., Guidelines for the use and interpretation of assays for monitoring autophagy, Autophagy, vol.12, pp.1-222, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02641288

C. Knudsen, C. Bonnefont, L. Fortun-lamothe, K. Ricaud, and X. Fernandez, , 2018.

, L'engraissement spontané du foie ches les palmipèdes : état des lieux et perspectives de recherche. INRA Prod, vol.31, pp.117-130

H. Koga, S. Kaushik, and A. M. Cuervo, Altered lipid content inhibits autophagic vesicular fusion, FASEB J, vol.24, pp.3052-3065, 2010.

T. Kono, M. Nishida, Y. Nishiki, Y. Seki, K. Sato et al., Characterisation of glucose transporter (GLUT) gene expression in broiler chickens, Br. Poult. Sci, vol.46, pp.510-515, 2005.

T. Koressaar and M. Remm, Enhancements and modifications of primer design program Primer3, Bioinformatics, vol.23, pp.1289-1291, 2007.

D. J. Kostyniuk, B. M. Culbert, J. A. Mennigen, and K. M. Gilmour, Social status affects lipid metabolism in rainbow trout, Oncorhynchus mykiss, Am. J. Physiol.-Regul. Integr. Comp. Physiol, vol.315, pp.241-255, 2018.

J. Krützfeldt and M. Stoffel, MicroRNAs: A new class of regulatory genes affecting metabolism, Cell Metab, vol.4, pp.9-12, 2006.

J. Krützfeldt, N. Rajewsky, R. Braich, K. G. Rajeev, T. Tuschl et al., Silencing of microRNAs in vivo with 'antagomirs, Nature, vol.438, pp.685-689, 2005.

K. A. Kumar, A. Lalitha, D. Pavithra, I. J. Padmavathi, M. Ganeshan et al., Maternal dietary folate and/or vitamin B12 restrictions alter body composition (adiposity) and lipid metabolism in Wistar rat offspring, J. Nutr. Biochem, vol.24, pp.25-31, 2013.

C. Labie and J. Tournut, Investigations on the histological and biochemical modifications in geese subject to cramming, Cah. Med. Veterinaire, 1970.

M. Laplante and D. M. Sabatini, mTOR signaling at a glance, J. Cell Sci, vol.122, pp.3589-3594, 2009.

C. Larzul, B. Imbert, M. Bernadet, G. Guy, R. et al., Meat quality in an intergeneric factorial crossbreeding between muscovy ( Cairina moschata ) and Pekin ( Anas platyrhynchos ) ducks, Anim. Res, vol.55, pp.219-229, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00890062

I. Laudadio, I. Manfroid, Y. Achouri, D. Schmidt, M. D. Wilson et al., A Feedback Loop Between the Liver-Enriched Transcription Factor Network and Mir-122 Controls Hepatocyte Differentiation, Gastroenterology, vol.142, pp.119-129, 2012.

B. Leclercq, J. C. Blum, M. R. Salichon, and ;. Canard, Ann. Biol. Anim. Biochim. Biophys, vol.15, pp.559-568, 1975.

J. Lee, R. Mendez, H. H. Heng, Z. Yang, and K. Zhang, Pharmacological ER stress promotes hepatic lipogenesis and lipid droplet formation, p.12

R. C. Lee, R. L. Feinbaum, A. , and V. , The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell, vol.75, pp.843-854, 1993.
URL : https://hal.archives-ouvertes.fr/in2p3-00597159

J. J. Lemasters, Selective Mitochondrial Autophagy, or Mitophagy, as a Targeted Defense Against Oxidative Stress, Mitochondrial Dysfunction, and Aging, Rejuvenation Res, vol.8, pp.3-5, 2005.

G. I. Lepesheva, R. D. Ott, T. Y. Hargrove, Y. Y. Kleshchenko, I. Schuster et al., Sterol 14?-Demethylase as a Potential Target for Antitrypanosomal Therapy: Enzyme Inhibition and Parasite Cell Growth, Chem. Biol, vol.14, pp.1283-1293, 2007.

A. P. Lewis and C. L. Jopling, Regulation and biological function of the liver-specific miR-122, Biochem. Soc. Trans, vol.38, pp.1553-1557, 2010.

H. Liu, J. Han, S. Y. Cao, T. Hong, D. Zhuo et al., Hepatic Autophagy Is Suppressed in the Presence of Insulin Resistance and Hyperinsulinemia INHIBITION OF FoxO1-DEPENDENT EXPRESSION OF KEY AUTOPHAGY GENES BY INSULIN, J. Biol. Chem, vol.284, pp.31484-31492, 2009.

K. J. Livak and T. D. Schmittgen, Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2???CT Method, Methods, vol.25, pp.402-408, 2001.

L. Locsmándi, R. Romvári, F. Bogenfürst, A. Szabó, M. Molnár et al., In vivo studies on goose liver development by means of computer tomography, Anim. Res, vol.54, pp.135-145, 2005.

L. Locsmándi, G. Hegedüs, G. Andrássy-baka, F. Bogenfürst, and R. Romvári, Following the goose liver development by means of cross-sectional digital imaging, liver histology and blood biochemical parameters, Acta Biol. Hung, 2007.

C. Loguercio, V. De-girolamo, I. De-sio, C. Tuccillo, A. Ascione et al., Non-alcoholic fatty liver disease in an area of southern Italy: main clinical, histological, and pathophysiological aspects, J. Hepatol, vol.35, pp.568-574, 2001.

M. C. Lorenz and J. Heitman, TOR Mutations Confer Rapamycin Resistance by Preventing Interaction with FKBP12-Rapamycin, J. Biol. Chem, vol.270, pp.27531-27537, 1995.

P. D. Lu, H. P. Harding, R. , and D. , Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response, J. Cell Biol, vol.167, pp.27-33, 2004.

D. Lutter, C. Marr, J. Krumsiek, E. W. Lang, and F. J. Theis, Intronic microRNAs support their host genes by mediating synergistic and antagonistic regulatory effects, BMC Genomics, vol.11, p.224, 2010.

L. Marandel, C. Labbe, J. Bobe, H. Jammes, J. Lareyre et al., Do not put all teleosts in one net: Focus on the sox2 and pou2 genes, Comp. Biochem. Physiol. B Biochem. Mol. Biol, vol.164, pp.69-79, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01000747

G. Marchesini, M. Brizi, A. M. Morselli-labate, G. Bianchi, E. Bugianesi et al., Association of nonalcoholic fatty liver disease with insulin resistance, Am. J. Med, vol.107, pp.450-455, 1999.

G. Mariño, F. Pietrocola, T. Eisenberg, Y. Kong, S. A. Malik et al., Regulation of Autophagy by Cytosolic Acetyl-Coenzyme A. Mol. Cell, vol.53, pp.710-725, 2014.

H. C. Masuoka and N. Chalasani, Nonalcoholic fatty liver disease: an emerging threat to obese and diabetic individuals, Ann. N. Y. Acad. Sci, vol.1281, pp.106-122, 2013.

J. D. Mcgarry and N. F. Brown, The Mitochondrial Carnitine Palmitoyltransferase System -From Concept to Molecular Analysis, Eur. J. Biochem, vol.244, pp.1-14, 1997.

C. J. Mcneil, S. M. Hay, G. J. Rucklidge, M. Reid, G. Duncan et al., Disruption of lipid metabolism in the liver of the pregnant rat fed folate-deficient and methyl donor-deficient diets, Br. J. Nutr, vol.99, pp.262-271, 2008.

C. J. Mcneil, S. M. Hay, G. J. Rucklidge, M. D. Reid, G. J. Duncan et al., Maternal diets deficient in folic acid and related methyl donors modify mechanisms associated with lipid metabolism in the fetal liver of the rat, Br. J. Nutr, vol.102, pp.1445-1452, 2009.

A. Meerson, M. Traurig, V. Ossowski, J. M. Fleming, M. Mullins et al., , 2013.

, Human adipose microRNA-221 is upregulated in obesity and affects fat metabolism downstream of leptin and TNF-?, Diabetologia, vol.56, pp.1971-1979

C. Migdal and M. Serres, Espèces réactives de l'oxygène et stress oxydant. médecine/sciences, vol.27, pp.405-412, 2011.

H. Min, A. Kapoor, M. Fuchs, F. Mirshahi, H. Zhou et al., Increased Hepatic Synthesis and Dysregulation of Cholesterol Metabolism Is Associated with the Severity of Nonalcoholic Fatty Liver Disease, Cell Metab, vol.15, pp.665-674, 2012.

P. S. Mitchell, R. K. Parkin, E. M. Kroh, B. R. Fritz, S. K. Wyman et al., Circulating microRNAs as stable blood-based markers for cancer detection, Proc. Natl. Acad. Sci, vol.105, pp.10513-10518, 2008.

N. Mizushima and M. Komatsu, Autophagy: Renovation of Cells and Tissues, Cell, vol.147, pp.728-741, 2011.

W. Molee, M. Bouillier-oudot, A. Auvergne, and R. Babilé, Changes in lipid composition of hepatocyte plasma membrane induced by overfeeding in duck, Comp. Biochem. Physiol. B Biochem. Mol. Biol, vol.141, pp.437-444, 2005.
URL : https://hal.archives-ouvertes.fr/hal-02671890

J. Mourot, N. Institut, R. A. De-la, and M. Kouba, Development of intra-and intermuscular adipose tissue in growing Large White and Meishan pigs, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00900287

, Reprod. Nutr. Dev. Fr

B. T. Nave, D. M. Ouwens, D. J. Withers, D. R. Alessi, and P. R. Shepherd, Mammalian target of rapamycin is a direct target for protein kinase B : identification of a convergence point for opposing effects of insulin and amino-acid deficiency on protein translation, vol.5, 1999.

H. Niemeyer, T. Ureta, and L. Clark-turri, Adaptive character of liver glucokinase, Mol. Cell. Biochem, vol.6, pp.109-126, 1975.

J. Nofer, B. Kehrel, M. Fobker, B. Levkau, G. Assmann et al., , 2002.

, HDL and arteriosclerosis: beyond reverse cholesterol transport, Atherosclerosis, vol.161, pp.1-16

F. J. Ortega, J. M. Mercader, V. Catalán, J. M. Moreno-navarrete, N. Pueyo et al., Targeting the Circulating MicroRNA Signature of Obesity, Clin. Chem, vol.59, pp.781-792, 2013.

S. Ottosen, T. B. Parsley, L. Yang, K. Zeh, L. Doorn et al., In Vitro Antiviral Activity and Preclinical and Clinical Resistance Profile of Miravirsen, a Novel Anti-Hepatitis C Virus Therapeutic Targeting the Human Factor miR-122, Antimicrob. Agents Chemother, vol.59, pp.599-608, 2015.

M. Pan, V. Maitin, S. Parathath, U. Andreo, S. X. Lin et al., Presecretory oxidation, aggregation, and autophagic destruction of apoprotein-B: A pathway for late-stage quality control, Proc. Natl. Acad. Sci, vol.105, pp.5862-5867, 2008.

K. R. Parzych and D. J. Klionsky, An Overview of Autophagy: Morphology, Mechanism, and Regulation, Antioxid. Redox Signal, vol.20, pp.460-473, 2013.

M. E. Patti, E. Brambilla, L. Luzi, E. J. Landaker, and C. R. Kahn, Bidirectional modulation of insulin action by amino acids, 1998.

S. Pattingre, A. Tassa, X. Qu, R. Garuti, X. H. Liang et al., Bcl-2 Antiapoptotic Proteins Inhibit Beclin 1-Dependent Autophagy, Cell, vol.122, pp.927-939, 2005.

T. Pioche, F. Skiba, M. Bernadet, I. Seiliez, W. Massimino et al., Kinetic study of the expression of genes related to hepatic steatosis, global intermediate metabolism and cellular stress during overfeeding in mule ducks, BioRxiv, vol.690156, 2019.

R. L. Printz, S. Koch, L. R. Potter, R. M. O'doherty, J. J. Tiesinga et al., Hexokinase II mRNA and gene structure, regulation by insulin, and evolution, J. Biol. Chem, vol.268, pp.5209-5219, 1993.

C. C. Pritchard, E. Kroh, B. Wood, J. D. Arroyo, K. J. Dougherty et al., Blood Cell Origin of Circulating MicroRNAs: A Cautionary Note for Cancer Biomarker Studies, Cancer Prev. Res. (Phila. Pa.), vol.5, pp.492-497, 2012.

A. Rashidpour, J. I. Silva-marrero, L. Seguí, I. V. Baanante, and I. Metón, Metformin counteracts glucose-dependent lipogenesis and impairs transdeamination in the liver of gilthead sea bream (Sparus aurata), Am. J. Physiol.-Regul. Integr. Comp. Physiol, vol.316, pp.265-273, 2019.

K. Ray, NAFLD-the next global epidemic, Nat. Rev. Gastroenterol. Hepatol, vol.10, p.621, 2013.

K. J. Rayner, Y. Suárez, A. Dávalos, S. Parathath, M. L. Fitzgerald et al., MiR-33 contributes to the regulation of cholesterol homeostasis, Science, vol.328, pp.1570-1573, 2010.

I. Regulustherapeutics, RG-125 (AZD4076), a microRNA therapeutic targeting microRNA 103/107 for the treatment of NASH in patients with type 2 diabetes/Pre-Diabetes, selected as clinical candidate by AstraZeneca, 2015.

H. Rémignon, R. B. Yahia, N. Marty-gasset, and J. Wilkesman, Apoptosis during the development of the hepatic steatosis in force-fed ducks and cooking yield implications, Poult. Sci, vol.97, pp.2211-2217, 2018.

T. C. Roberts, The microRNA machinery, MicroRNA: Basic Science, pp.15-30, 2015.

X. Roblin, J. Pofelski, and J. Zarski, Role de l'homocysteine au cours de la steatose hepatique et de l'hepatite chronique C, Gastroentérologie Clin. Biol, vol.31, pp.415-420, 2007.

A. Rodriguez, S. Griffiths-jones, J. L. Ashurst, and A. Bradley, Identification of Mammalian microRNA Host Genes and Transcription Units, Genome Res, vol.14, pp.1902-1910, 2004.

M. A. Rogers, J. Liu, B. Song, B. Li, C. C. Chang et al., Acyl-CoA:cholesterol acyltransferases (ACATs/SOATs): Enzymes with multiple sterols as substrates and as activators, J. Steroid Biochem. Mol. Biol, vol.151, pp.102-107, 2015.

M. Rong, G. Chen, and Y. Dang, Increased MiR-221 expression in hepatocellular carcinoma tissues and its role in enhancing cell growth and inhibiting apoptosis in vitro, BMC Cancer, vol.13, p.21, 2013.

A. A. Rowland and G. K. Voeltz, Endoplasmic reticulum-mitochondria contacts: function of the junction, Nat. Rev. Mol. Cell Biol, vol.13, pp.607-615, 2012.

C. N. Roybal, L. A. Hunsaker, O. Barbash, D. L. Jagt, and S. F. Abcouwer, The Oxidative Stressor Arsenite Activates Vascular Endothelial Growth Factor mRNA Transcription by an ATF4-dependent Mechanism, J. Biol. Chem, vol.280, pp.20331-20339, 2005.

S. Rozen and H. Skaletsky, Primer3 on the WWW for General Users and for Biologist Programmers, Bioinformatics Methods and Protocols, pp.365-386, 1999.

A. Saadoun and B. Leclercq, In Vivo Lipogenesis of Genetically Lean and Fat Chickens: Effects of Nutritional State and Dietary Fat, J. Nutr, vol.117, pp.428-435, 1987.
URL : https://hal.archives-ouvertes.fr/hal-02720398

G. Saez, S. Davail, G. Gentes, J. F. Hocquette, T. Jourdan et al., , 2009.

, Gene expression and protein content in relation to intramuscular fat content in Muscovy and Pekin ducks, Poult. Sci, vol.88, pp.2382-2391

M. Saitoh, N. Pullen, P. Brennan, D. Cantrell, P. B. Dennis et al., Regulation of an Activated S6 Kinase 1 Variant Reveals a Novel Mammalian Target of Rapamycin Phosphorylation Site, J. Biol. Chem, vol.277, pp.20104-20112, 2002.

M. R. Salichon, G. Guy, D. Rousselot, and J. C. Blum, Composition des 3 types de foie gras: oie, canard mulard et canard de Barbarie, Annales de Zootechnie, pp.213-220, 1994.
URL : https://hal.archives-ouvertes.fr/hal-00888972

A. R. Saltiel and C. R. Kahn, Insulin signalling and the regulation of glucose and lipid metabolism, Nature, vol.414, p.799, 2001.

H. Sano, S. Kane, E. Sano, C. P. M??inea, J. M. Asara et al., Insulin-stimulated Phosphorylation of a Rab GTPase-activating Protein Regulates GLUT4 Translocation, J. Biol. Chem, vol.278, pp.14599-14602, 2003.

A. J. Sanyal, C. Campbell-sargent, F. Mirshahi, W. B. Rizzo, M. J. Contos et al., Nonalcoholic steatohepatitis: Association of insulin resistance and mitochondrial abnormalities, Gastroenterology, vol.120, pp.1183-1192, 2001.

R. J. Schulze, A. Sathyanarayan, and D. G. Mashek, Breaking fat: The regulation and mechanisms of lipophagy, Biochim. Biophys. Acta BBA -Mol. Cell Biol. Lipids, vol.1862, pp.1178-1187, 2017.

S. Séité, T. Pioche, N. Ory, E. Plagnes-juan, S. Panserat et al., The Autophagic Flux Inhibitor Bafilomycine A1 Affects the Expression of Intermediary Metabolism-Related Genes in Trout Hepatocytes, Front. Physiol, vol.10, 2019.

Y. Seki, K. Sato, T. Kono, H. Abe, and Y. Akiba, Broiler chickens (Ross strain) lack insulin-responsive glucose transporter GLUT4 and have GLUT8 cDNA, Gen. Comp. Endocrinol, vol.133, pp.80-87, 2003.

D. Senft and Z. A. Ronai, UPR, autophagy, and mitochondria crosstalk underlies the ER stress response, Trends Biochem. Sci, vol.40, pp.141-148, 2015.

S. Shefer, G. Salen, A. Honda, A. K. Batta, L. B. Nguyen et al., Regulation of rat hepatic 3?-hydroxysterol ?7-reductase: substrate specificity, competitive and non-competitive inhibition, and phosphorylation/dephosphorylation, J. Lipid Res, vol.39, pp.2471-2476, 1998.

Y. Shen and E. White, p53-Dependent apoptosis pathways, Advances in Cancer Research, pp.55-84, 2001.

W. Shen, S. Azhar, and F. B. Kraemer, SR-B1: A Unique Multifunctional Receptor for Cholesterol Influx and Efflux, Annu. Rev. Physiol, vol.80, pp.95-116, 2018.

P. Simonen, A. Kotronen, M. Hallikainen, K. Sevastianova, J. Makkonen et al., Cholesterol synthesis is increased and absorption decreased in non-alcoholic fatty liver disease independent of obesity, J. Hepatol, vol.54, pp.153-159, 2011.

R. Singh and A. M. Cuervo, Autophagy in the Cellular Energetic Balance, Cell Metab, vol.13, pp.495-504, 2011.

R. Singh and A. M. Cuervo, Lipophagy: Connecting Autophagy and Lipid Metabolism, Int. J. Cell Biol, vol.2012, pp.1-12, 2012.

R. Singh, S. Kaushik, Y. Wang, Y. Xiang, I. Novak et al., Autophagy regulates lipid metabolism, Nature, vol.458, pp.1131-1135, 2009.

R. Singh, Y. Wang, Y. Xiang, K. E. Tanaka, W. A. Gaarde et al., , 2009.

, Differential effects of JNK1 and JNK2 inhibition on murine steatohepatitis and insulin resistance, Hepatology, vol.49, pp.87-96

P. Siuta-mangano, D. R. Janero, and M. D. Lane, Association and assembly of triglyceride and phospholipid with glycosylated and unglycosylated apoproteins of very low density lipoprotein in the intact liver cell, J. Biol. Chem, vol.257, pp.11463-11467, 1982.

S. Skiba-cassy, A. Collin, P. Chartrin, F. Médale, J. Simon et al., Chicken liver and muscle carnitine palmitoyltransferase 1: Nutritional regulation of messengers, Comp. Biochem. Physiol. B Biochem. Mol. Biol, vol.147, pp.278-287, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02667451

B. W. Smith, A. , and L. A. , Non-alcoholic fatty liver disease, Crit. Rev. Clin. Lab. Sci, vol.48, pp.97-113, 2011.

H. S. Soifer, J. J. Rossi, and P. Saetrom, MicroRNAs in Disease and Potential Therapeutic Applications, Mol. Ther, vol.15, pp.2070-2079, 2007.

E. Solsona-vilarrasa, R. Fucho, S. Torres, S. Nuñez, N. Nuño-lámbarri et al., Cholesterol enrichment in liver mitochondria impairs oxidative phosphorylation and disrupts the assembly of respiratory supercomplexes, Redox Biol, vol.24, p.101214, 2019.

A. Stahl, A current review of fatty acid transport proteins (SLC27), Pflüg. Arch, vol.447, pp.722-727, 2004.

J. Stenvang, A. Petri, M. Lindow, S. Obad, and S. Kauppinen, Inhibition of microRNA function by antimiR oligonucleotides, Silence, vol.3, p.1, 2012.

J. J. Strandholm, J. M. Cardenas, and R. D. Dyson, Pyruvate kinase isoenzymes in adult and fetal tissues of chicken, Biochemistry, vol.14, pp.2242-2246, 1975.

Z. Su, Z. Yang, Y. Xu, Y. Chen, Y. et al., MicroRNAs in apoptosis, autophagy and necroptosis, vol.6, pp.8474-8490, 2015.

N. Sud, J. Taher, and Q. Su, MicroRNAs and Noncoding RNAs in Hepatic Lipid and Lipoprotein Metabolism: Potential Therapeutic Targets of Metabolic Disorders: NONCODING RNAS AND MICRORNAS IN ANTIMETABOLIC SYNDROME, vol.76, p.318, 2015.

K. Takane, K. Fujishima, Y. Watanabe, A. Sato, N. Saito et al., Computational prediction and experimental validation of evolutionarily conserved microRNA target genes in bilaterian animals, BMC Genomics, vol.11, p.101, 2010.

C. M. Taniguchi, B. Emanuelli, and C. R. Kahn, Critical nodes in signalling pathways: insights into insulin action, Nat. Rev. Mol. Cell Biol, vol.7, pp.85-96, 2006.

A. Tavernier, S. Davail, K. Ricaud, M. Bernadet, and K. Gontier, Genes involved in the establishment of hepatic steatosis in Muscovy, Pekin and mule ducks, Mol. Cell. Biochem, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01606275

A. Tavernier, S. Davail, K. Ricaud, M. Bernadet, and K. Gontier, Genes involved in the establishment of hepatic steatosis in Muscovy, Pekin and mule ducks, Mol. Cell. Biochem, vol.424, pp.147-161, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01606275

A. Tavernier, R. Karine, B. Marie-dominique, G. Karine, and D. Stéphane, Pre-and post-prandial expression of genes involved in lipid metabolism at the end of the overfeeding period of mule ducks, Mol. Cell. Biochem, vol.438, pp.111-121, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01773149

L. Theron, X. Fernandez, N. Marty-gasset, C. Pichereaux, M. Rossignol et al., Identification by Proteomic Analysis of Early Post-mortem Markers Involved in the Variability in Fat Loss during Cooking of Mule Duck "Foie Gras, J. Agric. Food Chem, vol.59, pp.12617-12628, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02647532

L. Théron, T. Astruc, M. Bouillier-oudot, C. Molette, A. Vénien et al., The fusion of lipid droplets is involved in fat loss during cooking of duck "foie gras, Meat Sci, vol.89, pp.377-383, 2011.

M. Trajkovski, J. Hausser, J. Soutschek, B. Bhat, A. Akin et al., MicroRNAs 103 and 107 regulate insulin sensitivity, Nature, vol.474, pp.649-653, 2011.

F. Tremblay and A. Marette, Amino Acid and Insulin Signaling via the mTOR/p70 S6 Kinase Pathway A NEGATIVE FEEDBACK MECHANISM LEADING TO INSULIN RESISTANCE IN SKELETAL MUSCLE CELLS, J. Biol. Chem, vol.276, pp.38052-38060, 2001.

W. Tsai, S. Hsu, C. Hsu, T. Lai, S. Chen et al., MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis, J. Clin. Invest, vol.122, pp.2884-2897, 2012.

A. Untergasser, I. Cutcutache, T. Koressaar, J. Ye, B. C. Faircloth et al., Primer3-new capabilities and interfaces, vol.40, pp.115-115, 2012.

K. Uyeda and J. J. Repa, Carbohydrate response element binding protein, ChREBP, a transcription factor coupling hepatic glucose utilization and lipid synthesis, Cell Metab, vol.4, pp.107-110, 2006.

B. Vanhaesebroeck, A. , and D. R. , The PI3K-PDK1 connection : more than just a road to PKB, p.16, 2000.

L. A. Videla, R. Rodrigo, M. Orellana, V. Fernandez, G. Tapia et al., Oxidative stress-related parameters in the liver of non-alcoholic fatty liver disease patients, Clin. Sci, vol.106, pp.261-268, 2004.

S. Walrand, C. Guillet, J. Salles, N. Tardif, A. Maurin et al., Acides aminés et signalisation cellulaire, Nutr. Clin. Métabolisme, vol.22, pp.161-167, 2008.

H. Wang and R. H. Eckel, Lipoprotein lipase: from gene to obesity, Am. J. Physiol.-Endocrinol. Metab, vol.297, pp.271-288, 2009.

H. R. Waterham and R. J. Wanders, Biochemical and genetic aspects of 7-dehydrocholesterol reductase and Smith-Lemli-Opitz syndrome, Biochim. Biophys. Acta BBA -Mol. Cell Biol. Lipids, vol.1529, pp.340-356, 2000.

J. Weber, The physiology of long-distance migration: extending the limits of endurance metabolism, J. Exp. Biol, vol.212, pp.593-597, 2009.

J. A. Weber, D. H. Baxter, S. Zhang, D. Y. Huang, K. Huang et al., The MicroRNA Spectrum in 12 Body Fluids, Clin. Chem, vol.56, pp.1733-1741, 2010.

M. D. Weltman, G. C. Farrell, P. Hall, M. Ingelman-sundberg, and C. Liddle, Hepatic cytochrome P450 2E1 is increased in patients with nonalcoholic steatohepatitis, Hepatology, vol.27, pp.128-133, 1998.

M. F. White, The insulin signalling system and the IRS proteins, Diabetologia, vol.40, pp.2-17, 1997.

M. F. White, IRS proteins and the common path to diabetes, Am. J. Physiol.-Endocrinol, 2002.

. Metab, , vol.283, pp.413-422

E. Wienholds and R. H. Plasterk, MicroRNA function in animal development, FEBS Lett, vol.579, pp.5911-5922, 2005.

J. E. Wilson, Isozymes of mammalian hexokinase: structure, subcellular localization and metabolic function, J. Exp. Biol, vol.206, pp.2049-2057, 2003.

D. Wu, X. Wang, R. Zhou, and A. Cederbaum, CYP2E1 enhances ethanol-induced lipid accumulation but impairs autophaghy in HepG2 E47 cells, Biochem. Biophys. Res. Commun, vol.402, pp.116-122, 2010.

Z. Yang and D. J. Klionsky, Eaten alive: a history of macroautophagy, Nat. Cell Biol, vol.12, pp.814-822, 2010.

J. Yang, M. Fernández-galilea, L. Martínez-fernández, P. González-muniesa, A. Pérez-chávez et al., Oxidative Stress and Non-Alcoholic Fatty Liver Disease: Effects of Omega-3 Fatty Acid Supplementation, Nutrients, vol.11, p.872, 2019.

Z. Yang, H. Chen, H. Si, X. Li, X. Ding et al., Serum miR-23a, a potential biomarker for diagnosis of pre-diabetes and type 2 diabetes, Acta Diabetol, vol.51, pp.823-831, 2014.

S. Yousefi, R. Perozzo, I. Schmid, A. Ziemiecki, T. Schaffner et al., Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis, Nat. Cell Biol, vol.8, pp.1124-1132, 2006.

C. Zhang and A. M. Cuervo, Restoration of chaperone-mediated autophagy in aging liver improves cellular maintenance and hepatic function, Nat. Med, vol.14, pp.959-965, 2008.

P. Zhang, B. C. Mcgrath, J. Reinert, D. S. Olsen, L. Lei et al., The GCN2 eIF2 Kinase Is Required for Adaptation to Amino Acid Deprivation in Mice, Mol. Cell. Biol, vol.22, pp.6681-6688, 2002.

R. Zhao, J. Feng, and G. He, miR-613 regulates cholesterol efflux by targeting LXR? and ABCA1 in PPAR? activated THP-1 macrophages, Biochem. Biophys. Res. Commun, vol.448, pp.329-334, 2014.

L. H. Zhu, H. Meng, X. J. Duan, G. Q. Xu, J. Zhang et al., Gene expression profile in the liver tissue of geese after overfeeding, Poult. Sci, vol.90, pp.107-117, 2011.

X. Zhuang, K. P. Chung, Y. Cui, W. Lin, C. Gao et al., ATG9 regulates autophagosome progression from the endoplasmic reticulum in Arabidopsis, Proc. Natl. Acad. Sci, vol.114, pp.426-435, 2017.

, Liste des annexes Article pour les Treizièmes Journées de la Recherche Avicole et Palmipèdes à foie gras, 2019.

, Recherche de biomarqueurs de l'établissement de la stéatose hépatique chez le canard mulard : étude cinétique de l'expression de gènes en lien avec le métabolisme du cholestérol et l'autophagie

, Interest in food proteins to stimulate the synthesis of foie gras in overfed mule ducks, 2018.

, Interest of amino acids to stimulate hepatic steatosis in mule ducks, vol.2, pp.107-110, 1996.

E. Baeza, INRA Productions Animales, vol.26, pp.403-414, 2013.

G. Bénard, , vol.3, pp.49-52, 1998.

A. Bruhat, /sciences, vol.31, pp.1057-1060, 2015.

P. Chartrin, Comparative Biochemistry and Physiology Part A, vol.145, issue.3, pp.390-396, 2006.

A. M. Cuervo, Mol. Cell. Biochem, vol.263, pp.55-72, 2004.

S. Davail, Comparative Biochemistry and Physiology Part A, vol.134, issue.4, pp.707-715, 2003.

D. Hermier, Poultry Science, vol.78, pp.1398-1406, 1999.

D. Hermier, Comparative Biochemistry and Physiology Part B, vol.135, issue.4, pp.663-675, 2003.

H. Min, Cell Metabolism, vol.15, issue.5, pp.665-674, 2012.

N. Mizushima and M. Komatsu, Cell, vol.147, issue.4, pp.728-741, 2011.

A. Tavernier, Molecular and Cellular Biochemistry, vol.424, issue.1-2, pp.147-161, 2017.

S. Walrand, Nutrition Clinique et Métabolisme, vol.22, issue.4, pp.161-167, 2008.

L. H. Zhu, Poultry Science, vol.90, issue.1, pp.107-117, 2011.

X. Zhuang, Proceedings of the National Academy of Sciences, vol.114, issue.3, pp.426-435, 2017.