P. Brissot, M. Ropert, L. Lan, C. Loréal, and O. , Non-transferrin bound iron: A key role in iron overload and iron toxicity, Biochimica et Biophysica Acta (BBA) - General Subjects, vol.1820, issue.3, pp.403-413, 2012.
DOI : 10.1016/j.bbagen.2011.07.014

URL : https://hal.archives-ouvertes.fr/hal-00739430

B. Esposito, W. Breuer, P. Sirankapracha, P. Pootrakul, C. Hershko et al., Labile plasma iron in iron overload: redox activity and susceptibility to chelation, Blood, vol.102, issue.7, pp.2670-2677, 2003.
DOI : 10.1182/blood-2003-03-0807

N. Andrews, Disorders of iron metabolism, N Engl J Med Dec, vol.23341, issue.26, pp.1986-95, 1999.

J. Przybyszewska and E. Zekanowska, The role of hepcidin, ferroportin, HCP1, and DMT1 protein in iron absorption in the human digestive tract, Gastroenterology Review, vol.4, issue.4, pp.208-221, 2014.
DOI : 10.5114/pg.2014.45102

N. Andrews, The iron transporter DMT1, The International Journal of Biochemistry & Cell Biology, vol.31, issue.10, 1999.
DOI : 10.1016/S1357-2725(99)00065-5

M. Simovich, L. Hainsworth, P. Fields, J. Umbreit, and M. Conrad, Localization of the iron transport proteins mobilferrin and DMT-1 in the duodenum: The surprising role of mucin, American Journal of Hematology, vol.37, issue.1, pp.32-45, 2003.
DOI : 10.1002/ajh.10383

A. Pietrangelo, The ferroportin disease, Blood Cells, Molecules, and Diseases, vol.32, issue.1, pp.131-139, 2004.
DOI : 10.1016/j.bcmd.2003.08.003

C. Vulpe, Y. Kuo, T. Murphy, L. Cowley, C. Askwith et al., Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse, Nat Genet. Feb, vol.21, issue.2, pp.195-204, 1999.

F. Canonne-hergaux, A. Zhang, P. Ponka, and P. Gros, Characterization of the iron transporter DMT1 (NRAMP2/DCT1) in red blood cells of normal and anemic mk/mk mice, Blood, vol.98, issue.13, pp.3823-3853, 2001.
DOI : 10.1182/blood.V98.13.3823

T. Lambe, R. Simpson, S. Dawson, T. Bouriez-jones, T. Crockford et al., Identification of a Steap3 endosomal targeting motif essential for normal iron metabolism, Blood, vol.113, issue.8, pp.1805-1813, 2009.
DOI : 10.1182/blood-2007-11-120402

P. Aisen and I. Listowsky, Iron Transport and Storage Proteins, Annual Review of Biochemistry, vol.49, issue.1, pp.357-93, 1980.
DOI : 10.1146/annurev.bi.49.070180.002041

C. Thomas and L. Thomas, Biochemical markers and hematologic indices in the diagnosis of functional iron deficiency, Clin Chem, vol.48, issue.7, pp.1066-76, 2002.

T. Lambe, R. Simpson, S. Dawson, T. Bouriez-jones, T. Crockford et al., Identification of a Steap3 endosomal targeting motif essential for normal iron metabolism, Blood, vol.113, issue.8, pp.1805-1813, 2009.
DOI : 10.1182/blood-2007-11-120402

M. Knutson, M. Oukka, L. Koss, F. Aydemir, and M. Wessling-resnick, Iron release from macrophages after erythrophagocytosis is up-regulated by ferroportin 1 overexpression and down-regulated by hepcidin, Proceedings of the National Academy of Sciences, vol.102, issue.5, pp.1324-1332, 2005.
DOI : 10.1073/pnas.0409409102

S. Osaki, D. Johnson, and E. Frieden, The possible significance of the ferrous oxidase activity of ceruloplasmin in normal human serum, J Biol Chem, vol.241, issue.12, pp.2746-51, 1966.

J. Goforth, S. Anderson, C. Nizzi, and R. Eisenstein, Multiple determinants within iron-responsive elements dictate iron regulatory protein binding and regulatory hierarchy, RNA, vol.16, issue.1, pp.154-69, 2010.
DOI : 10.1261/rna.1857210

F. Samaniego, J. Chin, K. Iwai, T. Rouault, and R. Klausner, Molecular characterization of a second iron-responsive element binding protein, iron regulatory protein 2. Structure, function, and post-translational regulation, J Biol Chem, vol.269, issue.49, pp.30904-30914, 1994.

S. Marro, D. Chiabrando, E. Messana, J. Stolte, E. Turco et al., Heme controls ferroportin1 (FPN1) transcription involving Bach1, Nrf2 and a MARE/ARE sequence motif at position -7007 of the FPN1 promoter, Haematologica, vol.95, issue.8, pp.1261-1269, 2010.
DOI : 10.3324/haematol.2009.020123

E. Theil and . Ferritin, The Protein Nanocage and Iron Biomineral in Health and in Disease Available from, Inorg Chem, vol.52, issue.21, 2013.

A. Krause, R. Sillard, B. Kleemeier, E. Klüver, E. Maronde et al., Isolation and biochemical characterization of LEAP-2, a novel blood peptide expressed in the liver, Protein Science, vol.12, issue.1, pp.143-52, 2003.
DOI : 10.1110/ps.0213603

G. Nicolas, M. Bennoun, I. Devaux, C. Beaumont, B. Grandchamp et al., Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice, Proceedings of the National Academy of Sciences, vol.98, issue.15, pp.8780-8785, 2001.
DOI : 10.1073/pnas.151179498

URL : https://hal.archives-ouvertes.fr/inserm-00331349

G. Nicolas, M. Bennoun, A. Porteu, S. Mativet, C. Beaumont et al., Severe iron deficiency anemia in transgenic mice expressing liver hepcidin, Proceedings of the National Academy of Sciences, vol.99, issue.7, pp.4596-601, 2002.
DOI : 10.1073/pnas.072632499

C. Pigeon, G. Ilyin, B. Courselaud, P. Leroyer, B. Turlin et al., A New Mouse Liver-specific Gene, Encoding a Protein Homologous to Human Antimicrobial Peptide Hepcidin, Is Overexpressed during Iron Overload, Journal of Biological Chemistry, vol.276, issue.11, pp.7811-7820, 2001.
DOI : 10.1074/jbc.M008923200

H. Kulaksiz, F. Theilig, S. Bachmann, S. Gehrke, D. Rost et al., The iron-regulatory peptide hormone hepcidin: expression and cellular localization in the mammalian kidney, Journal of Endocrinology, vol.184, issue.2, pp.361-70, 2005.
DOI : 10.1677/joe.1.05729

G. Maisetta, R. Petruzzelli, F. Brancatisano, S. Esin, A. Vitali et al., Antimicrobial activity of human hepcidin 20 and 25 against clinically relevant bacterial strains: Effect of copper and acidic pH, Peptides, vol.31, issue.11, pp.1995-2002, 2010.
DOI : 10.1016/j.peptides.2010.08.007

C. Laarakkers, E. Wiegerinck, S. Klaver, M. Kolodziejczyk, H. Gille et al., Improved Mass Spectrometry Assay For Plasma Hepcidin: Detection and ! ! 94

P. Schwarz, J. Kübler, P. Strnad, K. Müller, T. Barth et al., infection, Gut, vol.61, issue.2, pp.193-201, 2012.
DOI : 10.1136/gut.2011.241208

URL : https://hal.archives-ouvertes.fr/hal-00710042

O. Itkonen, U. Stenman, J. Parkkinen, R. Soliymani, M. Baumann et al., Binding of Hepcidin to Plasma Proteins, Clinical Chemistry, vol.58, issue.7, pp.1158-60
DOI : 10.1373/clinchem.2012.186916

F. Wolff, M. Deleers, C. Melot, B. Gulbis, and F. Cotton, Hepcidin-25: Measurement by LC???MS/MS in serum and urine, reference ranges and urinary fractional excretion, Clinica Chimica Acta, vol.423, pp.99-104, 2013.
DOI : 10.1016/j.cca.2013.04.021

E. Nemeth, M. Tuttle, J. Powelson, M. Vaughn, A. Donovan et al., Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science, pp.2090-2093, 2004.

C. Delaby, N. Pilard, A. Gonçalves, C. Beaumont, and F. Canonne-hergaux, Presence of the iron exporter ferroportin at the plasma membrane of macrophages is enhanced by iron loading and down-regulated by hepcidin, Blood, vol.106, issue.12, pp.3979-84, 2005.
DOI : 10.1182/blood-2005-06-2398

J. Kaplan, D. Ward, D. Domenico, and I. , The molecular basis of iron overload disorders and iron-linked anemias, International Journal of Hematology, vol.26, issue.1, pp.14-20, 2011.
DOI : 10.1007/s12185-010-0760-0

R. Wang, C. Li, X. Xu, Y. Zheng, C. Xiao et al., 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

P. Lee, Role of Matriptase-2 (TMPRSS6) in Iron Metabolism, Acta Haematologica, vol.122, issue.2-3, pp.87-96, 2009.
DOI : 10.1159/000243792

L. Silvestri, A. Pagani, A. Nai, D. Domenico, I. Kaplan et al., The Serine Protease Matriptase-2 (TMPRSS6) Inhibits Hepcidin Activation by Cleaving Membrane Hemojuvelin, Cell Metabolism, vol.8, issue.6, pp.502-513, 2008.
DOI : 10.1016/j.cmet.2008.09.012

F. Guillem, S. Lawson, C. Kannengiesser, M. Westerman, C. Beaumont et al., Two nonsense mutations in the TMPRSS6 gene in a patient with microcytic anemia and iron deficiency, Blood, vol.112, issue.5, pp.2089-91, 2008.
DOI : 10.1182/blood-2008-05-154740

A. Pietrangelo, Hereditary Hemochromatosis: Pathogenesis, Diagnosis, and Treatment, Gastroenterology, vol.139, issue.2, pp.393-408, 2010.
DOI : 10.1053/j.gastro.2010.06.013

G. Nicolas, C. Chauvet, L. Viatte, J. Danan, X. Bigard et al., The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation, Journal of Clinical Investigation, vol.110, issue.7, pp.1037-1081, 2002.
DOI : 10.1172/JCI0215686

C. Besson-fournier, C. Latour, L. Kautz, J. Bertrand, T. Ganz et al., Induction of activin B by inflammatory stimuli up-regulates expression of the iron-regulatory peptide hepcidin through Smad1/5/8 signaling. Blood, pp.431-440, 2012.

L. Kautz and E. Nemeth, Molecular liaisons between erythropoiesis and iron metabolism. Blood, Jul, vol.24124, issue.4, pp.479-82, 2014.

L. Kautz, Erythroferrone, an erythroid regulator of iron metabolism]. Médecine Sci MS, pp.834-840, 2014.

R. Wenger, Cellular adaptation to hypoxia: O2-sensing protein hydroxylases, hypoxia-inducible transcription factors, and O2-regulated gene expression, The FASEB Journal, vol.16, issue.10, pp.1151-62, 2002.
DOI : 10.1096/fj.01-0944rev

C. Peyssonnaux, A. Zinkernagel, R. Schuepbach, E. Rankin, S. Vaulont et al., Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs), Journal of Clinical Investigation, vol.117, issue.7
DOI : 10.1172/JCI31370

A. Pietrangelo, Hereditary Hemochromatosis: Pathogenesis, Diagnosis, and Treatment, Gastroenterology, vol.139, issue.2, pp.393-408, 2010.
DOI : 10.1053/j.gastro.2010.06.013

G. Bergamaschi and L. Villani, Serum hepcidin: a novel diagnostic tool in disorders of iron metabolism, Haematologica, vol.94, issue.12, pp.1631-1634, 2009.
DOI : 10.3324/haematol.2009.013615

P. Brissot, M. Troadec, E. Bardou-jacquet, L. Lan, C. Jouanolle et al., Current approach to hemochromatosis, Blood Reviews, vol.22, issue.4, pp.195-210, 2008.
DOI : 10.1016/j.blre.2008.03.001

A. Roetto, A. Totaro, A. Piperno, A. Piga, F. Longo et al., New mutations inactivating transferrin receptor 2 in hemochromatosis type 3, Blood, vol.97, issue.9, pp.2555-60, 2001.
DOI : 10.1182/blood.V97.9.2555

P. Santos, L. Dinardo, C. Cançado, R. Schettert, I. Krieger et al., Non-HFE hemochromatosis, Revista Brasileira de Hematologia e Hemoterapia, vol.34, issue.4, pp.311-317, 2012.
DOI : 10.5581/1516-8484.20120079

M. Malekzadeh, A. Radmard, A. Nouroozi, M. Akbari, M. Amini et al., Juvenile Hemochromatosis, Genetic Study and Long-term Follow up after Therapy, Middle East J Dig Dis. 2014, vol.6, issue.2, pp.87-92

S. Bottomley and M. Fleming, Sideroblastic Anemia, Hematology/Oncology Clinics of North America, vol.28, issue.4, pp.653-70, 2014.
DOI : 10.1016/j.hoc.2014.04.008

E. Nemeth, Hepcidin in ??-thalassemia, Annals of the New York Academy of Sciences, vol.23, issue.1, pp.31-36, 2010.
DOI : 10.1111/j.1749-6632.2010.05585.x

J. Arezes and E. Nemeth, Hepcidin and iron disorders: new biology and clinical approaches, International Journal of Laboratory Hematology, vol.35, issue.8, pp.92-100
DOI : 10.1111/ijlh.12358

J. Zaritsky, Y. B. Wang, H. Westerman, M. Olbina, G. Nemeth et al., Hepcidin--A Potential Novel Biomarker for Iron Status in Chronic Kidney Disease, Clinical Journal of the American Society of Nephrology, vol.4, issue.6, pp.1051-1057, 2009.
DOI : 10.2215/CJN.05931108

R. Mogadam, A. Nemati, F. Amani, A. Ghorbanihaghjo, H. Argani et al., Association between hepcidin, haemoglobin level and iron status in stage 4 chronic kidney disease patients with anaemia, JPMA J Pak Med Assoc. 2015, vol.65, issue.4, pp.354-361

S. Lasocki, G. Baron, F. Driss, M. Westerman, H. Puy et al., Diagnostic accuracy of serum hepcidin for iron deficiency in critically ill patients with anemia, Intensive Care Medicine, vol.369, issue.Suppl 1, pp.1044-1052, 2010.
DOI : 10.1007/s00134-010-1794-8

M. Busbridge, C. Griffiths, D. Ashby, D. Gale, A. Jayantha et al., Development of a novel immunoassay for the iron regulatory peptide hepcidin, British Journal of Biomedical Science, vol.66, issue.3, pp.150-157, 2009.
DOI : 10.1080/09674845.2009.11730263

J. Kroot, E. Kemna, S. Bansal, M. Busbridge, N. Campostrini et al., Results of the first international round robin for the quantification of urinary and plasma hepcidin assays: need for standardization, Haematologica, vol.94, issue.12, pp.1748-52, 2009.
DOI : 10.3324/haematol.2009.010322

T. Ganz, G. Olbina, D. Girelli, E. Nemeth, and M. Westerman, Immunoassay for human serum hepcidin. Blood, Nov, vol.15112, issue.10, pp.4292-4299, 2008.

N. Grebenchtchikov, A. Geurts-moespot, J. Kroot, M. Den-heijer, H. Tjalsma et al., High-sensitive radioimmunoassay for human serum hepcidin, British Journal of Haematology, vol.26, issue.3, pp.317-342, 2009.
DOI : 10.1111/j.1365-2141.2009.07758.x

H. Peters, C. Laarakkers, D. Swinkels, and J. Wetzels, Serum hepcidin-25 levels in patients with chronic kidney disease are independent of glomerular filtration rate. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc -Eur Ren Assoc, pp.848-53, 2010.

H. Li, M. Rose, L. Tran, J. Zhang, L. Miranda et al., Development of a method for the sensitive and quantitative determination of hepcidin in human serum using LC-MS/MS, Journal of Pharmacological and Toxicological Methods, vol.59, issue.3, pp.171-80, 2009.
DOI : 10.1016/j.vascn.2009.02.004

C. Delaby, J. Vialaret, P. Bros, A. Gabelle, T. Lefebvre et al., Clinical measurement of Hepcidin-25 in human serum: Is quantitative mass spectrometry up to the job? Available from: https://www.mysciencework.com/publication/show/8062330/clinical-measurement-of- hepcidin-25-in-human-serum-is-quantitative-mass-spectrometry-up-to-the-job, 2015.

C. Schaap, J. Hendriks, G. Kortman, S. Klaver, J. Kroot et al., Diurnal Rhythm rather than Dietary Iron Mediates Daily Hepcidin Variations, Clinical Chemistry, vol.59, issue.3, pp.527-562, 2013.
DOI : 10.1373/clinchem.2012.194977

O. Itkonen, J. Parkkinen, U. Stenman, and E. Hämäläinen, Preanalytical factors and reference intervals for serum hepcidin LC???MS/MS method, Clinica Chimica Acta, vol.413, issue.7-8, pp.7-8696, 2012.
DOI : 10.1016/j.cca.2011.12.015

J. Kroot, J. Hendriks, C. Laarakkers, S. Klaver, E. Kemna et al., (Pre)analytical imprecision, between-subject variability, and daily variations in serum and urine hepcidin: Implications for clinical studies, Analytical Biochemistry, vol.389, issue.2, pp.124-133, 2009.
DOI : 10.1016/j.ab.2009.03.039

M. Koenig, L. Tussing-humphreys, J. Day, B. Cadwell, and E. Nemeth, Hepcidin and Iron Homeostasis during Pregnancy. Nutrients, pp.3062-83, 2014.

N. Van-der-weerd, M. Grooteman, M. Bots, M. Van-den-dorpel, C. Hoedt et al., Hepcidin-25 in Chronic Hemodialysis Patients Is Related to Residual Kidney Function and Not to Treatment with Erythropoiesis Stimulating Agents Available from, PLoS ONE [Internet], vol.7, issue.7, 2012.

S. Song, M. Iwahashi, N. Tomosugi, K. Uno, J. Yamana et al., Comparative evaluation of the effects of treatment with tocilizumab and TNF-?? inhibitors on serum hepcidin, anemia response and disease activity in rheumatoid arthritis patients, Arthritis Research & Therapy, vol.15, issue.5, p.141, 2013.
DOI : 10.1371/journal.pone.0038136

A. Pietrangelo, Hepcidin in human iron disorders: Therapeutic implications, Journal of Hepatology, vol.54, issue.1, pp.173-81, 2011.
DOI : 10.1016/j.jhep.2010.08.004

G. Briand, S. Lemaire-ewing, F. Parente, and R. Garnotel, Mass spectrometry and inherited metabolic diseases diagnosis] 2015 Feb, Ann Biol Clin, vol.73, issue.1, pp.93-106

M. Jemal, L. High-throughput-quantitative-bioanalysis-by, . Ms, and . Ms, High-throughput quantitative bioanalysis by LC/MS/MS, Biomedical Chromatography, vol.71, issue.6, pp.422-431, 2000.
DOI : 10.1002/1099-0801(200010)14:6<422::AID-BMC25>3.0.CO;2-I

G. Hopfgartner, E. Varesio, V. Tschäppät, C. Grivet, E. Bourgogne et al., Triple quadrupole linear ion trap mass spectrometer for the analysis of small molecules and macromolecules, Journal of Mass Spectrometry, vol.39, issue.8, pp.845-55, 2004.
DOI : 10.1002/jms.659

V. Laiko, M. Baldwin, and A. Burlingame, Atmospheric Pressure Matrix-Assisted Laser Desorption/Ionization Mass Spectrometry, Analytical Chemistry, vol.72, issue.4, pp.652-659, 2000.
DOI : 10.1021/ac990998k

H. Lam, E. Deutsch, J. Eddes, J. Eng, N. King et al., Development and validation of a spectral library searching method for peptide identification from MS/MS, PROTEOMICS, vol.5, issue.5, pp.655-67, 2007.
DOI : 10.1002/pmic.200600625

R. Craig, J. Cortens, and R. Beavis, The use of proteotypic peptide libraries for protein identification, Rapid Communications in Mass Spectrometry, vol.20, issue.13, pp.1844-50, 2005.
DOI : 10.1002/rcm.1992

. Méthode, Two Sample t-test; Alternative :two.sided ! Statistique observée Qobs : 1

. Moyenne, Groupe 1: 12.014615384615, Groupe, vol.2, pp.9-5670588235294