M. Simon, M. Bourel, B. Genetet, and R. Fauchet, Idiopathic Hemochromatosis, New England Journal of Medicine, vol.297, issue.19, pp.1017-1038, 1977.
DOI : 10.1056/NEJM197711102971901

J. Feder, A. Gnirke, and W. Thomas, A novel MHC class I???like gene is mutated in patients with hereditary haemochromatosis, Nature Genetics, vol.88, issue.4, pp.399-408, 1996.
DOI : 10.1016/0888-7543(95)80219-C

A. Krause, S. Neitz, and H. Magert, LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity, FEBS Letters, vol.93, issue.2-3, pp.147-50, 2000.
DOI : 10.1016/S0014-5793(00)01920-7

C. Pigeon, G. Ilyin, and B. Courselaud, A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice Hepcidin, a urinary antimicrobial peptide synthesized in the liver, J Biol Chem Proc Natl Acad Sci U S A J Biol Chem, vol.27698276, issue.5, pp.7811-98780, 2001.

T. Ganz, Hepcidin and iron regulation, 10 years later, Blood, vol.117, issue.17, pp.4425-4458, 2011.
DOI : 10.1182/blood-2011-01-258467

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3099567

E. Nemeth, M. Tuttle, and J. Powelson, Hepcidin Regulates Cellular Iron Efflux by Binding to Ferroportin and Inducing Its Internalization, Science, vol.306, issue.5704, pp.2090-2093, 2004.
DOI : 10.1126/science.1104742

J. Gao, J. Chen, M. Kramer, H. Tsukamoto, A. Zhang et al., Interaction of the Hereditary Hemochromatosis Protein HFE with Transferrin Receptor 2 Is Required for Transferrin-Induced Hepcidin Expression, Cell Metabolism, vol.9, issue.3, pp.217-244, 2009.
DOI : 10.1016/j.cmet.2009.01.010

E. Corradini, M. Rozier, and D. Meynard, Iron regulation of hepcidin despite attenuated Smad1

B. Andriopoulos, J. Corradini, E. Xia, and Y. , BMP6 is a key endogenous regulator of hepcidin expression and iron metabolism Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression Modulation of bone morphogenetic protein signaling in vivo regulates systemic iron balance, Nat Genet Nat Genet Babitt JL J Clin Invest, vol.4138117, issue.13, pp.482-7531, 2006.

M. Shayeghi, G. Latunde-dada, and J. Oakhill, Identification of an Intestinal Heme Transporter, Cell, vol.122, issue.5, pp.789-801, 2005.
DOI : 10.1016/j.cell.2005.06.025

H. Gunshin, Y. Fujiwara, A. Custodio, C. Direnzo, S. Robine et al., Slc11a2 is required for intestinal iron absorption and erythropoiesis but dispensable in placenta and liver, Journal of Clinical Investigation, vol.115, issue.5, pp.1258-66, 2005.
DOI : 10.1172/JCI24356

S. Abboud and D. Haile, A Novel Mammalian Iron-regulated Protein Involved in Intracellular Iron Metabolism, Journal of Biological Chemistry, vol.275, issue.26, pp.19906-19918, 2000.
DOI : 10.1074/jbc.M000713200

A. Mckie, P. Marciani, and A. Rolfs, A Novel Duodenal Iron-Regulated Transporter, IREG1, Implicated in the Basolateral Transfer of Iron to the Circulation, Molecular Cell, vol.5, issue.2, pp.299-309, 2000.
DOI : 10.1016/S1097-2765(00)80425-6

A. Donovan, A. Brownlie, and Y. Zhou, Positional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter, Nature, vol.403, pp.776-81, 2000.

C. Vulpe, Y. Kuo, and T. Murphy, Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse, Nat Genet, vol.21, pp.195-204, 1999.

S. Bekri, P. Gual, and R. Anty, Increased Adipose Tissue Expression of Hepcidin in Severe Obesity Is Independent From Diabetes and NASH, Gastroenterology, vol.131, issue.3, pp.788-96, 2006.
DOI : 10.1053/j.gastro.2006.07.007

C. Peyssonnaux, A. Zinkernagel, V. Datta, X. Lauth, R. Johnson et al., TLR4-dependent hepcidin expression by myeloid cells in response to bacterial pathogens Hepcidin, a putative mediator of anemia of inflammation, is a type II acute-phase protein Hepcidin, a new iron regulatory peptide, Blood Blood Blood Cells Mol Dis, vol.10710129, issue.23, pp.3727-322461, 2002.

T. Ganz, Molecular Control of Iron Transport, Journal of the American Society of Nephrology, vol.18, issue.2, pp.394-400, 2007.
DOI : 10.1681/ASN.2006070802

S. Rivera, E. Nemeth, V. Gabayan, M. Lopez, D. Farshidi et al., Synthetic hepcidin causes rapid dose-dependent hypoferremia and is concentrated in ferroportin-containing organs, Blood, vol.106, issue.6, pp.2196-2205, 2005.
DOI : 10.1182/blood-2005-04-1766

B. Courselaud, C. Pigeon, and Y. Inoue, EBPalpha regulates hepatic transcription of hepcidin, an antimicrobial peptide and regulator of iron metabolism. Cross-talk between C/EBP pathway and iron metabolism Natural and synthetic STAT3 inhibitors reduce hepcidin expression in differentiated mouse hepatocytes expressing the active phosphorylated STAT3 form STAT3 is required for IL-6-gp130-dependent activation of hepcidin in vivo STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation, 30. Wrighting DM, Andrews NC. Interleukin-6 induces hepcidin expression through STAT3, pp.41163-70477, 2002.

J. Truksa, H. Peng, P. Lee, and E. Beutler, Bone morphogenetic proteins 2, 4, and 9 stimulate murine hepcidin 1 expression independently of Hfe, transferrin receptor 2 (Tfr2), and IL-6, Proceedings of the National Academy of Sciences, vol.103, issue.27, pp.10289-93, 2006.
DOI : 10.1073/pnas.0603124103

R. Wang, C. Li, and X. Xu, A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression, Cell Metabolism, vol.2, issue.6, pp.399-409, 2005.
DOI : 10.1016/j.cmet.2005.10.010

A. Steinbicker, T. Bartnikas, and L. Lohmeyer, Perturbation of hepcidin expression by BMP type I receptor deletion induces iron overload in mice, Blood, vol.118, issue.15, pp.4224-4254, 2011.
DOI : 10.1182/blood-2011-03-339952

L. Kautz, D. Meynard, and A. Monnier, Iron regulates phosphorylation of Smad1/5/8 and gene expression of Bmp6, Smad7, Id1, and Atoh8 in the mouse liver, Blood, vol.112, issue.4, pp.1503-1512, 2008.
DOI : 10.1182/blood-2008-03-143354

URL : https://hal.archives-ouvertes.fr/hal-00298517

D. Meynard, L. Kautz, V. Darnaud, F. Canonne-hergaux, H. Coppin et al., Lack of the bone morphogenetic protein BMP6 induces massive iron overload, Nature Genetics, vol.41, issue.4, pp.478-81, 2009.
DOI : 10.1093/nar/29.9.e45

URL : https://hal.archives-ouvertes.fr/hal-00385005

E. Ramos, L. Kautz, and R. Rodriguez, Evidence for distinct pathways of hepcidin regulation by acute and chronic iron loading in mice The hemochromatosis proteins HFE, TfR2, and HJV form a membrane-associated protein complex for hepcidin regulation, Hepatology J Hepatol, vol.5357, issue.37, pp.1333-411052, 2011.

D. Wallace, L. Summerville, E. Crampton, D. Frazer, G. Anderson et al., Combined deletion of Hfe and transferrin receptor 2 in mice leads to marked dysregulation of hepcidin and iron overload, Hepatology, vol.94, issue.6, pp.1992-2000, 2009.
DOI : 10.1002/hep.23198

P. Schmidt and M. Fleming, Transgenic HFE-dependent induction of hepcidin in mice does not require transferrin receptor-2, American Journal of Hematology, vol.115, issue.6, pp.588-95, 2012.
DOI : 10.1002/ajh.23173

T. Goswami and N. Andrews, Hereditary Hemochromatosis Protein, HFE, Interaction with Transferrin Receptor 2 Suggests a Molecular Mechanism for Mammalian Iron Sensing, Journal of Biological Chemistry, vol.281, issue.39, pp.28494-28502, 2006.
DOI : 10.1074/jbc.C600197200

M. Johnson and C. Enns, Diferric transferrin regulates transferrin receptor 2 protein stability, Blood, vol.104, issue.13, pp.4287-93, 2004.
DOI : 10.1182/blood-2004-06-2477

A. Calzolari, C. Raggi, and S. Deaglio, TfR2 localizes in lipid raft domains and is released in exosomes to activate signal transduction along the MAPK pathway, Journal of Cell Science, vol.119, issue.21, pp.4486-98, 2006.
DOI : 10.1242/jcs.03228

E. Valore and T. Ganz, Posttranslational processing of hepcidin in human hepatocytes is mediated by the prohormone convertase furin, Blood Cells, Molecules, and Diseases, vol.40, issue.1, pp.132-140, 2008.
DOI : 10.1016/j.bcmd.2007.07.009

L. Silvestri, A. Pagani, and C. Camaschella, Furin-mediated release of soluble hemojuvelin: a new link between hypoxia and iron homeostasis, Blood, vol.111, issue.2, pp.924-955, 2008.
DOI : 10.1182/blood-2007-07-100677

C. Garuti, Y. Tian, and G. Montosi, Hepcidin Expression Does Not Rescue the Iron-Poor Phenotype of Kupffer Cells in Hfe-Null Mice After Liver Transplantation, Gastroenterology, vol.139, issue.1, pp.315-337, 2010.
DOI : 10.1053/j.gastro.2010.03.043

E. Bardou-jacquet, J. Philip, and R. Lorho, Liver transplantation normalizes serum hepcidin level and cures iron metabolism alterations in HFE hemochromatosis. Hepatology 2013. 47 Hepcidin in iron overload disorders Non-specific serum iron in thalassaemia: an abnormal serum iron fraction of potential toxicity, Blood Br J Haematol, vol.10540, pp.255-63, 1978.
URL : https://hal.archives-ouvertes.fr/hal-00876626

R. Hider, Nature of nontransferrin-bound iron, European Journal of Clinical Investigation, vol.95, issue.s1, pp.50-54, 2002.
DOI : 10.1182/blood.V97.3.792

P. Brissot, T. Wright, W. Ma, R. Weisiger, B. Esposito et al., Efficient clearance of non-transferrin-bound iron by rat liver. Implications for hepatic iron loading in iron overload states Labile plasma iron in iron overload: redox activity and susceptibility to chelation, J Clin Invest Blood, vol.76102, issue.51, pp.1463-702670, 1985.

L. Lan, C. Loreal, O. Cohen, and T. , Redox active plasma iron in C282Y/C282Y hemochromatosis, Blood, vol.105, issue.11, pp.4527-4558, 2005.
DOI : 10.1182/blood-2004-09-3468

D. Domenico, I. Ward, D. Musci, G. Kaplan, J. Drakesmith et al., Iron overload due to mutations in ferroportin Resistance to hepcidin is conferred by hemochromatosis-associated mutations of ferroportin, Haematologica Blood, vol.91106, issue.54, pp.92-51092, 2005.

D. Domenico, I. Ward, D. Di-patti, and M. , Ferroxidase activity is required for the stability of cell surface ferroportin in cells expressing GPI-ceruloplasmin Hemochromatosis: an endocrine liver disease, Embo J Hepatology, vol.2646, pp.2823-311291, 2007.

A. Piperno, D. Girelli, and E. Nemeth, Blunted hepcidin response to oral iron challenge in HFE-related hemochromatosis, Blood, vol.110, issue.12, pp.4096-100, 2007.
DOI : 10.1182/blood-2007-06-096503

O. Loreal, C. Haziza-pigeon, and M. Troadec, Hepcidin in iron metabolism Hepcidin levels in humans are correlated with hepatic iron stores, hemoglobin levels, and hepatic function The molecular pathogenesis of hereditary hemochromatosis Quantitation of hepcidin from human and mouse serum using liquid chromatography tandem mass spectrometry, Curr Protein Pept Sci Blood Semin Liver Dis Blood, vol.610631110, issue.61, pp.279-91746, 2005.

A. Jouanolle, P. Fergelot, G. Gandon, J. Yaouanq, L. Gall et al., A candidate gene for hemochromatosis: frequency of the C282Y and H63D mutations, Human Genetics, vol.100, issue.5-6, pp.544-551, 1997.
DOI : 10.1007/s004390050549

P. Adams, D. Reboussin, and J. Barton, Hemochromatosis and iron-overload screening in a racially diverse population Gender-specific phenotypic expression and screening strategies in C282Y-linked haemochromatosis: a study of 9396 French people Global prevalence of putative haemochromatosis mutations, N Engl J Med Br J Haematol J Med Genet, vol.35211834, issue.65, pp.1769-781170, 1997.

P. Gochee, L. Powell, D. Cullen, D. Sart, D. Rossi et al., A population-based study of the biochemical and clinical expression of the H63D hemochromatosis mutation The clinical relevance of compound heterozygosity for the C282Y and H63D substitutions in hemochromatosis1403-10. 68. EASL clinical practice guidelines for HFE hemochromatosis Diagnosis and management of hemochromatosis, Gastroenterology Clin Gastroenterol Hepatol J Hepatol Bacon BR Hepatology, vol.12245354, issue.69, pp.646-513, 2002.

P. Aguilar-martinez, B. Grandchamp, and S. Cunat, Iron overload in HFE C282Y heterozygotes at first genetic testing: a strategy for identifying rare HFE variants Penetrance of 845G--> A (C282Y) HFE hereditary haemochromatosis mutation in the USA, Haematologica Lancet, vol.96359, pp.507-14211, 2002.

J. Olynyk, D. Cullen, S. Aquilia, E. Rossi, L. Summerville et al., A Population-Based Study of the Clinical Expression of the Hemochromatosis Gene, New England Journal of Medicine, vol.341, issue.10, pp.718-742, 1999.
DOI : 10.1056/NEJM199909023411002

J. Waalen, B. Nordestgaard, and E. Beutler, The penetrance of hereditary hemochromatosis, Best Practice & Research Clinical Haematology, vol.18, issue.2, pp.203-223, 2005.
DOI : 10.1016/j.beha.2004.08.023

K. Allen, L. Gurrin, and C. Constantine, Iron-overload-related disease in HFE hereditary hemochromatosis HFE genotypes and dietary heme iron: no evidence of strong gene-nutrient interaction on serum ferritin concentrations in middleaged women, N Engl J Med Nutr Metab Cardiovasc Dis, vol.35816, issue.75, pp.221-3060, 2006.

J. Kaltwasser, E. Werner, K. Schalk, C. Hansen, R. Gottschalk et al., Clinical trial on the effect of regular tea drinking on iron accumulation in genetic haemochromatosis, Gut, vol.43, issue.5, pp.699-704, 1998.
DOI : 10.1136/gut.43.5.699

O. Loreal, Y. Deugnier, and R. Moirand, Liver fibrosis in genetic hemochromatosis, Journal of Hepatology, vol.16, issue.1-2, pp.122-129, 1992.
DOI : 10.1016/S0168-8278(05)80104-7

K. Bridle, T. Cheung, and T. Murphy, Hepcidin is down-regulated in alcoholic liver injury: implications for the pathogenesis of alcoholic liver disease Decreased iron burden in overweight C282Y homozygous women: Putative role of increased hepcidin production, Alcohol Clin Exp Res Hepatology, vol.3057, pp.106-121784, 2006.

F. Laine, A. Jouannolle, and J. Morcet, Phenotypic expression in detected C282Y homozygous women depends on body mass index, Journal of Hepatology, vol.43, issue.6, pp.1055-1064, 2005.
DOI : 10.1016/j.jhep.2005.05.027

R. Moirand, P. Adams, V. Bicheler, P. Brissot, and Y. Deugnier, Clinical features of genetic hemochromatosis in women compared with men Genes that modify the hemochromatosis phenotype in mice, Ann Intern Med J Clin Invest, vol.127105, pp.105-101209, 1997.

M. Bensaid, S. Fruchon, C. Mazeres, S. Bahram, M. Roth et al., Multigenic control of hepatic iron loading in a murine model of hemochromatosis Haptoglobin modifies the hemochromatosis phenotype in mice, Gastroenterology Blood, vol.126105, pp.3353-3358, 2004.

S. Jacolot, L. Gac, G. Scotet, V. Quere, I. Mura et al., HAMP as a modifier gene that increases the phenotypic expression of the HFE pC282Y homozygous genotype Digenic inheritance of mutations in HAMP and HFE results in different types of haemochromatosis, Blood Hum Mol Genet, vol.10312, issue.86, pp.2835-402241, 2003.

L. Gac, G. Scotet, V. Ka, and C. , The recently identified type 2A juvenile haemochromatosis gene (HJV), a second candidate modifier of the C282Y homozygous phenotype, Human Molecular Genetics, vol.13, issue.17, pp.1913-1921, 2004.
DOI : 10.1093/hmg/ddh206

J. Milet, V. Dehais, and C. Bourgain, Common Variants in the BMP2, BMP4, and HJV Genes of the Hepcidin Regulation Pathway Modulate HFE Hemochromatosis Penetrance, The American Journal of Human Genetics, vol.81, issue.4, pp.799-807, 2007.
DOI : 10.1086/520001

P. Lee, T. Gelbart, C. West, C. Halloran, E. Beutler et al., Seeking candidate mutations that affect iron homeostasis Idiopathic hemochromatosis in a young female. A case study and review of the syndrome in young people, Blood Cells Mol Dis Gastroenterology, vol.2976, issue.90, pp.471-87178, 1979.

G. Papanikolaou, M. Samuels, and E. Ludwig, Mutations in HFE2 cause iron overload in chromosome 1q-linked juvenile hemochromatosis Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis Conditional disruption of mouse HFE2 gene: maintenance of systemic iron homeostasis requires hepatic but not skeletal muscle hemojuvelin, Nat Genet Nat Genet Hepatology Island ML Jouanolle AM, Mosser A Haematologica, vol.36335494, issue.94, pp.77-8221, 2003.

P. Hsiao, K. Tsai, and S. Shin, A novel mutation of transferrin receptor 2 in a Taiwanese woman with type 3 hemochromatosis, Journal of Hepatology, vol.47, issue.2, pp.303-309, 2007.
DOI : 10.1016/j.jhep.2007.04.014

L. Gac, G. Mons, F. Jacolot, S. Scotet, V. Ferec et al., Early onset hereditary hemochromatosis resulting from a novel TFR2 gene nonsense mutation (R105X) in two siblings of north French descent, British Journal of Haematology, vol.73, issue.5, pp.674-682, 2004.
DOI : 10.1182/blood-2003-07-2378

S. Majore, F. Milano, and F. Binni, Homozygous p.M172K mutation of the TFR2 gene in an Italian family with type 3 hereditary hemochromatosis and early onset iron overload, Haematologica, vol.91, pp.33-98, 2006.

A. Piperno, A. Roetto, and R. Mariani, Homozygosity for transferrin receptor-2 Y250X mutation induces early iron overload New mutations inactivating transferrin receptor 2 in hemochromatosis type 3, Haematologica Blood, vol.8997, pp.359-602555, 2001.

E. Bardou-jacquet, S. Cunat, and M. Beaumont-epinette, Variable age of onset and clinical severity in transferrin receptor 2 related haemochromatosis: novel observations, British Journal of Haematology, vol.54, issue.Pt 1, pp.278-81, 2013.
DOI : 10.1111/bjh.12350

URL : https://hal.archives-ouvertes.fr/inserm-00836894

D. Girelli, C. Bozzini, and A. Roetto, Clinical and pathologic findings in hemochromatosis type 3 due to a novel mutation in transferrin receptor 2 gene, Gastroenterology, vol.122, issue.5, pp.1295-302, 2002.
DOI : 10.1053/gast.2002.32984

V. Gerolami, L. Gac, G. Mercier, L. Nezri, M. Berge-lefranc et al., Early-onset haemochromatosis caused by a novel combination of TFR2 mutations(p.R396X/c.1538-2 A>G) in a woman of Italian descent, Haematologica, vol.93, issue.5, pp.45-51, 2008.
DOI : 10.3324/haematol.12884

G. Montosi, A. Donovan, and A. Totaro, Autosomal-dominant hemochromatosis is associated with a mutation in the ferroportin (SLC11A3) gene A mutation in SLC11A3 is associated with autosomal dominant hemochromatosis, J Clin Invest Nat Genet, vol.10828, issue.104, pp.619-23213, 2001.

J. Barton, R. Acton, P. Lee, and C. West, SLC40A1 Q248H allele frequencies and Q248H-associated risk of non-HFE iron overload in persons of sub-Saharan African descent, Blood Cells, Molecules, and Diseases, vol.39, issue.2, pp.206-217, 2007.
DOI : 10.1016/j.bcmd.2007.03.008

E. Beutler, J. Barton, and V. Felitti, Ferroportin 1 (SCL40A1) variant associated with iron overload in African-Americans, Blood Cells, Molecules, and Diseases, vol.31, issue.3, pp.305-314, 2003.
DOI : 10.1016/S1079-9796(03)00165-7

V. Subramaniam, D. Wallace, J. Dixon, L. Fletcher, and D. Crawford, Ferroportin disease due to the A77D mutation in Australia, Gut, vol.54, issue.7, pp.1048-1057, 2005.
DOI : 10.1136/gut.2005.069021

L. Lan, C. Mosser, A. Ropert, and M. , Sex and Acquired Cofactors Determine Phenotypes of Ferroportin Disease, Gastroenterology, vol.140, issue.4, pp.1199-207, 2011.
DOI : 10.1053/j.gastro.2010.12.049

URL : https://hal.archives-ouvertes.fr/inserm-00554693

D. Wallace, R. Clark, H. Harley, V. Subramaniam, P. Phatak et al., Autosomal dominant iron overload due to a novel mutation of ferroportin1 associated with parenchymal iron loading and cirrhosis Autosomal dominant hereditary hemochromatosis associated with a novel ferroportin mutation and unique clinical features Aceruloplasminemia, an inherited disorder of iron metabolism, J Hepatol Blood Cells Mol Dis Biometals, vol.403416, issue.111, pp.710-3157, 2003.

K. Yoshida, K. Furihata, and S. Takeda, A mutation in the ceruloplasmin gene is associated with systemic hemosiderosis in humans, Nature Genetics, vol.224, issue.3, pp.267-72, 1995.
DOI : 10.1006/bbrc.1993.2596

S. Kono, H. Suzuki, and K. Takahashi, Hepatic iron overload associated with a decreased serum ceruloplasmin level in a novel clinical type of aceruloplasminemia Molecular characterization of a third case of human atransferrinemia Identification of a human mutation of DMT1 in a patient with microcytic anemia and iron overload, Gastroenterology Blood Blood, vol.131104105, issue.115, pp.240-526071337, 2004.

C. Beaumont, J. Delaunay, G. Hetet, B. Grandchamp, M. De-montalembert et al., Two new human DMT1 gene mutations in a patient with microcytic anemia, low ferritinemia, and liver iron overload Iolascon A Microcytic anemia and hepatic iron overload in a child with compound heterozygous mutations in DMT1 (SCL11A2) A novel N491S mutation in the human SLC11A2 gene impairs protein trafficking and in association with the G212V mutation leads to microcytic anemia and liver iron overload, Blood Blood Bardou-Jacquet E Blood Cells Mol Dis, vol.10710747, issue.118, pp.4168-70349, 2006.

T. Cox, S. Bottomley, J. Wiley, M. Bawden, C. Matthews et al., -to-Ser Substitution in Erythroid 5-Aminolevulinate Synthase, New England Journal of Medicine, vol.330, issue.10, pp.675-684, 1994.
DOI : 10.1056/NEJM199403103301004

URL : https://hal.archives-ouvertes.fr/in2p3-00656167

R. Allikmets, W. Raskind, A. Hutchinson, N. Schueck, M. Dean et al., Mutation of a putative mitochondrial iron transporter gene (ABC7) in X-linked sideroblastic anemia and ataxia (XLSA/A) The human counterpart of zebrafish shiraz shows sideroblastic-like microcytic anemia and iron overload Regulation of ferritin expression by alcohol in a human hepatoblastoma cell line and in rat hepatocyte cultures The metabolic syndrome, Hum Mol Genet Blood J Hepatol Lancet, vol.811023375, issue.121, pp.743-9431, 1995.

M. Mendler, B. Turlin, and R. Moirand, Insulin resistance???associated hepatic iron overload, Gastroenterology, vol.117, issue.5, pp.1155-63, 1999.
DOI : 10.1016/S0016-5085(99)70401-4

M. Ruivard, F. Laine, and T. Ganz, Iron absorption in dysmetabolic iron overload syndrome is decreased and correlates with increased plasma hepcidin, Journal of Hepatology, vol.50, issue.6, pp.1219-1244, 2009.
DOI : 10.1016/j.jhep.2009.01.029

M. Ferrante, A. Geubel, J. Fevery, G. Marogy, Y. Horsmans et al., Hereditary hyperferritinaemia???cataract syndrome: a challenging diagnosis for the hepatogastroenterologist, European Journal of Gastroenterology & Hepatology, vol.17, issue.11, pp.1247-53, 2005.
DOI : 10.1097/00042737-200511000-00016

C. Kannengiesser, A. Jouanolle, and G. Hetet, A new missense mutation in the L ferritin coding sequence associated with elevated levels of glycosylated ferritin in serum and absence of iron overload, Haematologica, vol.94, issue.3, pp.335-344, 2009.
DOI : 10.3324/haematol.2008.000125

D. Girelli, C. Bozzini, and G. Zecchina, Clinical, biochemical and molecular findings in a series of families with hereditary hyperferritinaemia-cataract syndrome, British Journal of Haematology, vol.304, issue.2, pp.334-374, 2001.
DOI : 10.1006/jmbi.1994.1325

Y. Gandon, D. Olivie, and D. Guyader, Non-invasive assessment of hepatic iron stores by MRI, The Lancet, vol.363, issue.9406, pp.357-62, 2004.
DOI : 10.1016/S0140-6736(04)15436-6

S. Pierre, T. Clark, P. Chua-anusorn, and W. , Noninvasive measurement and imaging of liver iron concentrations using proton magnetic resonance Magnetic resonance imaging to identify classic and nonclassic forms of ferroportin disease, Blood Blood Cells Mol Dis, vol.10537, pp.855-61192, 2005.

J. Wood, C. Enriquez, and N. Ghugre, MRI R2 and R2* mapping accurately estimates hepatic iron concentration in transfusion-dependent thalassemia and sickle cell disease patients Deugnier Y, Turlin B. Pathology of hepatic iron overload, Blood World J Gastroenterol, vol.10613, issue.133, pp.1460-54755, 2005.

Y. Deugnier and B. Turlin, Pathology of hepatic iron overload Stauber RE, Lackner C. Noninvasive diagnosis of hepatic fibrosis in chronic hepatitis C, Semin Liver Dis World J Gastroenterol, vol.3113, issue.135, pp.260-714287, 2007.

D. Crawford, T. Murphy, and L. Ramm, Serum hyaluronic acid with serum ferritin accurately predicts cirrhosis and reduces the need for liver biopsy in C282Y hemochromatosis, Hepatology, vol.122, issue.2, pp.418-443, 2009.
DOI : 10.1002/hep.22650

J. Barton, P. Lee, C. West, and S. Bottomley, Iron overload and prolonged ingestion of iron supplements: Clinical features and mutation analysis of hemochromatosis-associated genes in four cases, American Journal of Hematology, vol.326, issue.10, pp.760-767, 2006.
DOI : 10.1002/ajh.20714

Y. Deugnier, O. Loreal, and F. Carre, Increased body iron stores in elite road cyclists, Medicine & Science in Sports & Exercise, vol.34, issue.5, pp.876-80, 2002.
DOI : 10.1097/00005768-200205000-00023

T. Tanno, N. Bhanu, and P. Oneal, High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin, Nature Medicine, vol.33, issue.9, pp.1096-101, 2007.
DOI : 10.1038/nm1629

K. Musallam, S. Rivella, E. Vichinsky, and E. Rachmilewitz, Non-transfusion-dependent thalassemias, Haematologica, vol.98, issue.6, pp.833-877, 2013.
DOI : 10.3324/haematol.2012.066845

URL : http://doi.org/10.3324/haematol.2012.066845

J. Dale, M. Burritt, and A. Zinsmeister, Diurnal Variation of Serum Iron, Iron-Binding Capacity, Transferrin Saturation, and Ferritin Levels, American Journal of Clinical Pathology, vol.117, issue.5, pp.802-810, 2002.
DOI : 10.1309/2YT4-CMP3-KYW7-9RK1

P. Adams, D. Reboussin, and R. Press, Biological variability of transferrin saturation and unsaturated iron-binding capacity e1-7. 143. EASL. EASL clinical practice guidelines for HFE hemochromatosis, Am J Med J Hepatol, vol.12053, pp.9993-10015, 2007.

P. Phatak, P. Brissot, and M. Wurster, A phase 1/2, dose-escalation trial of deferasirox for the treatment of iron overload in HFE-related hereditary hemochromatosis, Hepatology, vol.122, issue.Suppl 2, pp.1671-779, 2010.
DOI : 10.1002/hep.23879

. Hemojuvelin, TFR1 : Transferrin Receptor 1. TFR2 : Transferrin Receptor 2. BMP Receptor : Bone Morphegenic Protein receptor