Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-zzh7m Total loading time: 0 Render date: 2024-04-29T01:01:04.799Z Has data issue: false hasContentIssue false

2 - Normal iron absorption and metabolism

Published online by Cambridge University Press:  01 June 2011

James C. Barton
Affiliation:
University of Alabama, Birmingham
Corwin Q. Edwards
Affiliation:
University of Utah Medical Center
Pradyumna D. Phatak
Affiliation:
University of Rochester Medical Center, New York
Robert S. Britton
Affiliation:
St Louis University, Missouri
Bruce R. Bacon
Affiliation:
St Louis University, Missouri
James C. Barton
Affiliation:
University of Alabama, Birmingham
Corwin Q. Edwards
Affiliation:
University of Utah School of Medicine, Salt Lake City
Pradyumna D. Phatak
Affiliation:
University of Rochester Medical Center, New York
Robert S. Britton
Affiliation:
St Louis University, Missouri
Bruce R. Bacon
Affiliation:
St Louis University, Missouri
Get access

Summary

Iron is essential to life because it is the central oxygen ligand in the heme proteins hemoglobin and myoglobin. Accordingly, there are many interactions between iron homeostasis and oxygen regulation and delivery. Iron is also required for cytochrome P-450 enzyme oxidative metabolism and DNA synthesis. In health, body iron content is controlled by absorption that responds to iron losses and the rate of erythropoiesis. Multiple mechanisms provide functional feedback control of iron homeostasis, tissue oxygen sensing and delivery, and the tempo of red blood cell production. The physiologic capacity to excrete iron is very limited. Thus, body iron content is regulated almost entirely by controlled absorption. This chapter reviews the basic physiologic and molecular characteristics of iron metabolism and homeostasis, and their pertinence to iron overload disorders.

Iron physiology

Normal iron homeostasis is maintained by absorption of iron from the diet that precisely balances iron loss, and by controlled iron distribution in the body. Normal healthy adults have 4000–5000 mg of iron (Table 2.1). Daily iron loss occurs due to perspiration, desquamation from skin, and minor injuries, and from the gastrointestinal tract. The rate of this unavoidable iron loss is proportional to body iron stores. Women lose additional iron due to menstruation, pregnancy and childbirth, and lactation. Overall, daily iron losses in adult men and post-menopausal women are approximately 1.0 mg and in menstruating women approximately 1.5 mg. The median iron loss ascribable to pregnancy is 500 mg.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Finch, CA, Deubelbeiss, K, Cook, JD, et al. Ferrokinetics in man. Medicine (Baltimore) 1970; 49: 17–53.CrossRefGoogle ScholarPubMed
Conrad, ME, Barton, JC. Factors affecting iron balance. Am J Hematol 1981; 10: 199–225.CrossRefGoogle ScholarPubMed
Chung, J, Wessling-Resnick, M. Molecular mechanisms and regulation of iron transport. Crit Rev Clin Lab Sci 2003; 40: 151–82.CrossRefGoogle ScholarPubMed
Bothwell, TH, Charlton, RW, Cook, JD, Finch, CA.Iron loss. Iron Metabolism in Man. Oxford, Blackwell Scientific Publications. 1979; 245–55.Google Scholar
Harrison, PM, Fischbach, FA, Hoy, TG, Haggis, GH. Ferric oxyhydroxide core of ferritin. Nature 1967; 216: 1188–90.CrossRefGoogle ScholarPubMed
Theil, EC. Ferritin: structure, gene regulation, and cellular function in animals, plants, and microorganisms. Annu Rev Biochem 1987; 56: 289–315.CrossRefGoogle ScholarPubMed
Levi, S, Yewdall, SJ, Harrison, PM, et al. Evidence of H-and L-chains have co-operative roles in the iron-uptake mechanism of human ferritin. Biochem J 1992; 288 (Pt 2): 591–6.CrossRefGoogle ScholarPubMed
Harrison, PM, Arosio, P. The ferritins: molecular properties, iron storage function and cellular regulation. Biochim Biophys Acta 1996; 1275: 161–203.CrossRefGoogle ScholarPubMed
Wixom, RL, Prutkin, L, Munro, HN. Hemosiderin: nature, formation, and significance. Int Rev Exp Pathol 1980; 22: 193–225.Google ScholarPubMed
Domenico, I, Vaughn, MB, Li, L, et al. Ferroportin-mediated mobilization of ferritin iron precedes ferritin degradation by the proteasome. EMBO J 2006; 25: 5396–404.CrossRefGoogle ScholarPubMed
Levy, JE, Jin, O, Fujiwara, Y, Kuo, F, Andrews, NC. Transferrin receptor is necessary for development of erythrocytes and the nervous system. Nat Genet 1999; 21: 396–9.Google Scholar
Ned, RM, Swat, W, Andrews, NC. Transferrin receptor 1 is differentially required in lymphocyte development. Blood 2003; 102: 3711–18.CrossRefGoogle ScholarPubMed
Ohgami, RS, Campagna, DR, Greer, EL, et al. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat Genet 2005; 37: 1264–9.CrossRefGoogle ScholarPubMed
Fleming, MD, Trenor, CC, Su, MA, et al. Microcytic anaemia mice have a mutation in Nramp2, a candidate iron transporter gene. Nat Genet 1997; 16: 383–6.CrossRefGoogle ScholarPubMed
Gunshin, H, Mackenzie, B, Berger, UV, et al. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature 1997; 388: 482–8.CrossRefGoogle ScholarPubMed
Gunshin, H, Fujiwara, Y, Custodio, AO, Direnzo, C, Robine, S, Andrews, NC. Slc11a2 is required for intestinal iron absorption and erythropoiesis but dispensable in placenta and liver. J Clin Invest 2005; 115: 1258–66.CrossRefGoogle ScholarPubMed
Shindo, M, Torimoto, Y, Saito, H, et al. Functional role of DMT1 in transferrin-independent iron uptake by human hepatocyte and hepatocellular carcinoma cell, HLF. Hepatol Res 2006; 35: 152–62.Google ScholarPubMed
Davies, PS, Zhang, AS, Anderson, EL, et al. Evidence for the interaction of the hereditary haemochromatosis protein, HFE, with the transferrin receptor in endocytic compartments. Biochem J 2003; 373: 145–53.CrossRefGoogle ScholarPubMed
Aziz, N, Munro, HN. Iron regulates ferritin mRNA translation through a segment of its 5' untranslated region. Proc Natl Acad Sci USA 1987; 84: 8478–82.CrossRefGoogle ScholarPubMed
Hentze, MW, Rouault, TA, Caughman, SW, Dancis, A, Harford, JB, Klausner, RD. A cis-acting element is necessary and sufficient for translational regulation of human ferritin expression in response to iron. Proc Natl Acad Sci USA 1987; 84: 6730–4.CrossRefGoogle Scholar
Casey, JL, Koeller, DM, Ramin, VC, Klausner, RD, Harford, JB. Iron regulation of transferrin receptor mRNA levels requires iron-responsive elements and a rapid turnover determinant in the 3' untranslated region of the mRNA. EMBO J 1989; 8: 3693–9.Google Scholar
Abboud, S, Haile, DJ. A novel mammalian iron-regulated protein involved in intracellular iron metabolism. J Biol Chem 2000; 275: 19 906–12.CrossRefGoogle ScholarPubMed
McKie, AT, Marciani, P, Rolfs, A, et al. A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation. Mol Cell 2000; 5: 299–309.CrossRefGoogle Scholar
Hubert, N, Hentze, MW. Previously uncharacterized isoforms of divalent metal transporter (DMT)-1: implications for regulation and cellular function. Proc Natl Acad Sci USA 2002; 99: 12 345–50.CrossRefGoogle ScholarPubMed
Lymboussaki, A, Pignatti, E, Montosi, G, Garuti, C, Haile, DJ, Pietrangelo, A. The role of the iron-responsive element in the control of ferroportin 1/IREG1/MTP1 gene expression. J Hepatol 2003; 39: 710–15.CrossRefGoogle ScholarPubMed
Leipuviene, R, Theil, EC. The family of iron-responsive RNA structures regulated by changes in cellular iron and oxygen. Cell Mol Life Sci 2007; 64: 2945–55.CrossRefGoogle ScholarPubMed
Casey, JL, Hentze, MW, Koeller, DM, et al. Iron-responsive elements: regulatory RNA sequences that control mRNA levels and translation. Science 1988; 240: 924–8.CrossRefGoogle Scholar
Hentze, MW, Caughman, SW, Rouault, TA, et al. Identification of the iron-responsive element for the translational regulation of human ferritin mRNA. Science 1987; 238: 1570–3.CrossRefGoogle ScholarPubMed
Caughman, SW, Hentze, MW, Rouault, TA, Harford, JB, Klausner, RD.The iron-responsive element is the single element responsible for iron-dependent translational regulation of ferritin biosynthesis. Evidence for function as the binding site for a translational repressor. J Biol Chem 1988; 263: 19,048–52.Google ScholarPubMed
Leibold, EA, Munro, HN. Cytoplasmic protein binds in vitro to a highly conserved sequence in the 5′ untranslated region of ferritin heavy- and light-subunit mRNAs. Proc Natl Acad Sci USA 1988; 85: 2171–5.CrossRefGoogle ScholarPubMed
Rouault, TA, Hentze, MW, Caughman, SW, Harford, JB, Klausner, RD. Binding of a cytosolic protein to the iron-responsive element of human ferritin messenger RNA. Science 1988; 241: 1207–10.CrossRefGoogle ScholarPubMed
Chen, Y, Qian, ZM, Du, J, et al. Iron loading inhibits ferroportin 1 expression in PC12 cells. Neurochem Int 2005; 47: 507–13.CrossRefGoogle ScholarPubMed
Hentze, MW, Argos, P.Homology between IRE-BP, a regulatory RNA-binding protein, aconitase, and isopropylmalate isomerase. Nucleic Acids Res 1991; 19: 1739–40.CrossRefGoogle ScholarPubMed
Kaptain, S, Downey, WE, Tang, C, et al. A regulated RNA-binding protein also possesses aconitase activity. Proc Natl Acad Sci USA 1991; 88: 10 109–13.CrossRefGoogle ScholarPubMed
Eisenstein, RS. Iron regulatory proteins and the molecular control of mammalian iron metabolism. Annu Rev Nutr 2000; 20: 627–62.CrossRefGoogle ScholarPubMed
Walden, WE, Selezneva, AI, Dupuy, J, et al. Structure of dual function iron regulatory protein 1 complexed with ferritin IRE-RNA. Science 2006; 314: 1903–8.CrossRefGoogle ScholarPubMed
Haile, DJ, Hentze, MW, Rouault, TA, Harford, JB, Klausner, RD. Regulation of interaction of the iron-responsive element-binding protein with iron-responsive RNA elements. Mol Cell Biol 1989; 9: 5055–61.CrossRefGoogle ScholarPubMed
Meyron-Holtz, EG, Ghosh, MC, Iwai, K, et al. Genetic ablations of iron regulatory proteins 1 and 2 reveal why iron regulatory protein 2 dominates iron homeostasis. EMBO J 2004; 23: 386–95.CrossRefGoogle ScholarPubMed
Muckenthaler, M, Gray, NK, Hentze, MW. IRP-1 binding to ferritin mRNA prevents the recruitment of the small ribosomal subunit by the cap-binding complex eIF4F. Mol Cell 1998; 2: 383–8.CrossRefGoogle ScholarPubMed
Ganz, T. Hepcidin and its role in regulating systemic iron metabolism. Hematology Am Soc Hematol Educ Program 2006; 29–35.Google ScholarPubMed
Krause, A, Neitz, S, Magert, HJ, et al. LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity. FEBS Lett 2000; 480: 147–50.CrossRefGoogle ScholarPubMed
Park, CH, Valore, EV, Waring, AJ, Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J Biol Chem 2001; 276: 7806–10.CrossRefGoogle ScholarPubMed
Pigeon, C, Ilyin, G, Courselaud, B, et al. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem 2001; 276: 7811–19.CrossRefGoogle Scholar
Scamuffa, N, Basak, A, Lalou, C, et al. Regulation of prohepcidin processing and activity by the subtilisin-like pro-protein convertases Furin, PC5, PACE4, and PC7. Gut 2008; 57: 1573–82.CrossRefGoogle Scholar
Andrews, NC. Forging a field: the golden age of iron biology. Blood 2008; 112: 219–30.CrossRefGoogle ScholarPubMed
Gehrke, SG, Kulaksiz, H, Herrmann, T, et al. Expression of hepcidin in hereditary hemochromatosis: evidence for a regulation in response to the serum transferrin saturation and to non-transferrin-bound iron. Blood 2003; 102: 371–6.CrossRefGoogle ScholarPubMed
Papanikolaou, G, Samuels, ME, Ludwig, EH, et al. Mutations in HFE2 cause iron overload in chromosome 1q-linked juvenile hemochromatosis. Nat Genet 2004; 36: 77–82.CrossRefGoogle ScholarPubMed
Nemeth, E, Roetto, A, Garozzo, G, Ganz, T, Camaschella, C. Hepcidin is decreased in TFR2 hemochromatosis. Blood 2005; 105: 1803–6.CrossRefGoogle ScholarPubMed
Piperno, A, Girelli, D, Nemeth, E, et al. Blunted hepcidin response to oral iron challenge in HFE-related hemochromatosis. Blood 2007; 110: 4096–100.CrossRefGoogle ScholarPubMed
Wrighting, DM, Andrews, NC. Interleukin-6 induces hepcidin expression through STAT3. Blood 2006; 108: 3204–9.CrossRefGoogle ScholarPubMed
Truksa, J, Lee, P, Beutler, E.Two BMP responsive elements, STAT, and bZIP/HNF4/COUP motifs of the hepcidin promoter are critical for BMP, SMAD1, and HJV responsiveness. Blood 2009; 113: 688–95.CrossRefGoogle ScholarPubMed
Casanovas, G, Mleczko-Sanecka, K, Altamura, S, Hentze, MW, Muckenthaler, MU. Bone morphogenetic protein (BMP)-responsive elements located in the proximal and distal hepcidin promoter are critical for its response to HJV/BMP/SMAD. J Mol Med 2009; 87: 471–80.CrossRefGoogle ScholarPubMed
Merryweather-Clarke, AT, Cadet, E, Bomford, A, et al. Digenic inheritance of mutations in HAMP and HFE results in different types of haemochromatosis. Hum Mol Genet 2003; 12: 2241–7.CrossRefGoogle ScholarPubMed
Jacolot, S, Gac, G, Scotet, V, Quere, I, Mura, C, Ferec, C. HAMP as a modifier gene that increases the phenotypic expression of the HFE pC282Y homozygous genotype. Blood 2004; 103: 2835–40.CrossRefGoogle ScholarPubMed
Porto, G, Roetto, A, Daraio, F, et al. A Portuguese patient homozygous for the -25G>A mutation of the HAMP promoter shows evidence of steady-state transcription but fails to up-regulate hepcidin levels by iron. Blood 2005; 106: 2922–3.CrossRefGoogle ScholarPubMed
Nicolas, G, Bennoun, M, Devaux, I, et al. Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice. Proc Natl Acad Sci USA 2001; 98: 8780–5.CrossRefGoogle ScholarPubMed
Lesbordes-Brion, JC, Viatte, L, Bennoun, M, et al. Targeted disruption of the hepcidin 1 gene results in severe hemochromatosis. Blood 2006; 108: 1402–5.CrossRefGoogle ScholarPubMed
Nicolas, G, Bennoun, M, Porteu, A, et al. Severe iron deficiency anemia in transgenic mice expressing liver hepcidin. Proc Natl Acad Sci USA 2002; 99: 4596–601.CrossRefGoogle ScholarPubMed
Roy, CN, Mak, HH, Akpan, I, Losyev, G, Zurakowski, D, Andrews, NC. Hepcidin antimicrobial peptide transgenic mice exhibit features of the anemia of inflammation. Blood 2007; 109: 4038–44.CrossRefGoogle ScholarPubMed
Nicolas, G, Chauvet, C, Viatte, L, et al. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Invest 2002; 110: 1037–44.CrossRefGoogle Scholar
Weinstein, DA, Roy, CN, Fleming, MD, Loda, MF, Wolfsdorf, JI, Andrews, NC. Inappropriate expression of hepcidin is associated with iron refractory anemia: implications for the anemia of chronic disease. Blood 2002; 100: 3776–81.CrossRefGoogle ScholarPubMed
Adamsky, K, Weizer, O, Amariglio, N, et al. Decreased hepcidin mRNA expression in thalassemic mice. Br J Haematol 2004; 124: 123–4.CrossRefGoogle ScholarPubMed
Ashrafian, H. Hepcidin: the missing link between hemochromatosis and infections. Infect Immun 2003; 71: 6693–700.CrossRefGoogle ScholarPubMed
Ganz, T. Hepcidin–a peptide hormone at the interface of innate immunity and iron metabolism. Curr Top Microbiol Immunol 2006; 306: 183–98.Google ScholarPubMed
Paradkar, PN, Domenico, I, Durchfort, N, Zohn, I, Kaplan, J, Ward, DM. Iron depletion limits intracellular bacterial growth in macrophages. Blood 2008; 112: 866–74.CrossRefGoogle ScholarPubMed
Babitt, JL, Huang, FW, Xia, Y, Sidis, Y, Andrews, NC, Lin, HY. Modulation of bone morphogenetic protein signaling in vivo regulates systemic iron balance. J Clin Invest 2007; 117: 1933–9.CrossRefGoogle ScholarPubMed
Babitt, JL, Huang, FW, Wrighting, DM, et al. Bone morphogenetic protein signaling by hemojuvelin regulates hepcidin expression. Nat Genet 2006; 38: 531–9.CrossRefGoogle ScholarPubMed
Wang, RH, Li, C, Xu, X, et al. A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression. Cell Metab 2005; 2: 399–409.CrossRefGoogle ScholarPubMed
Anderson, GJ, Frazer, DM. Iron metabolism meets signal transduction. Nat Genet 2006; 38: 503–4.CrossRefGoogle ScholarPubMed
Kautz, L, Meynard, D, Monnier, A, et al. Iron regulates phosphorylation of Smad1/5/8 and gene expression of Bmp6, Smad7, Id1, and Atoh8 in the mouse liver. Blood 2008; 112: 1503–9.CrossRefGoogle ScholarPubMed
McReynolds, LJ, Gupta, S, Figueroa, ME, Mullins, MC, Evans, T. Smad1 and Smad5 differentially regulate embryonic hematopoiesis. Blood 2007; 110: 3881–90.CrossRefGoogle ScholarPubMed
Truksa, J, Lee, P, Peng, H, Flanagan, J, Beutler, E. The distal location of the iron-responsive region of the hepcidin promoter. Blood 2007; 110: 3436–7.CrossRefGoogle ScholarPubMed
Verga Falzacappa, MV, Vujic, SM, Kessler, R, Stolte, J, Hentze, MW, Muckenthaler, MU. STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation. Blood 2007; 109: 353–8.CrossRefGoogle ScholarPubMed
Peyssonnaux, C, Zinkernagel, AS, Schuepbach, RA, et al. Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs). J Clin Invest 2007; 117: 1926–32.CrossRefGoogle Scholar
Domenico, I, Nemeth, E, Nelson, JM, et al. The hepcidin-binding site on ferroportin is evolutionarily conserved. Cell Metab 2008; 8: 146–56.CrossRefGoogle ScholarPubMed
Domenico, I, Ward, DM, Musci, G, Kaplan, J. Evidence for the multimeric structure of ferroportin. Blood 2007; 109: 2205–9.CrossRefGoogle ScholarPubMed
Schimanski, LM, Drakesmith, H, Talbott, C, et al. Ferroportin: lack of evidence for multimers. Blood Cells Mol Dis 2008; 40: 360–9.CrossRefGoogle ScholarPubMed
Nemeth, E, Tuttle, MS, Powelson, J, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004; 306: 2090–3.CrossRefGoogle ScholarPubMed
Domenico, I, Ward, DM, Langelier, C, et al. The molecular mechanism of hepcidin-mediated ferroportin down-regulation. Mol Biol Cell 2007; 18: 2569–78.CrossRefGoogle ScholarPubMed
Parmley, RT, Barton, JC, Conrad, ME, Austin, RL. Ultrastructural cytochemistry of iron absorption. Am J Pathol 1978; 93: 707–27.Google ScholarPubMed
McKie, AT, Barrow, D, Latunde-Dada, GO, et al. An iron-regulated ferric reductase associated with the absorption of dietary iron. Science 2001; 291: 1755–9.CrossRefGoogle ScholarPubMed
Ohgami, RS, Campagna, DR, McDonald, A, Fleming, MD. The Steap proteins are metalloreductases. Blood 2006; 108: 1388–94.CrossRefGoogle ScholarPubMed
Canonne-Hergaux, F, Gruenheid, S, Ponka, P, Gros, P. Cellular and subcellular localization of the Nramp2 iron transporter in the intestinal brush border and regulation by dietary iron. Blood 1999; 93: 4406–17.Google ScholarPubMed
Lam-Yuk-Tseung, S, Touret, N, Grinstein, S, Gros, P. Carboxyl-terminus determinants of the iron transporter DMT1/SLC11A2 isoform II (-IRE/1B) mediate internalization from the plasma membrane into recycling endosomes. Biochemistry 2005; 44: 12 149–59.CrossRefGoogle ScholarPubMed
Andrews, NC. The iron transporter DMT1. Int J Biochem Cell Biol 1999; 31: 991–4.CrossRefGoogle ScholarPubMed
Frazer, DM, Wilkins, SJ, Becker, EM, et al. A rapid decrease in the expression of DMT1 and Dcytb but not Ireg1 or hephaestin explains the mucosal block phenomenon of iron absorption. Gut 2003; 52: 340–6.CrossRefGoogle ScholarPubMed
Priwitzerova, M, Pospisilova, D, Prchal, JT, et al. Severe hypochromic microcytic anemia caused by a congenital defect of the iron transport pathway in erythroid cells. Blood 2004; 103: 3991–2.CrossRefGoogle ScholarPubMed
Conrad, ME, Umbreit, JN, Moore, EG. Rat duodenal iron-binding protein mobilferrin is a homologue of calreticulin. Gastroenterology 1993; 104: 1700–4.CrossRefGoogle ScholarPubMed
Conrad, ME, Umbreit, JN, Peterson, RD, Moore, EG, Harper, KP. Function of integrin in duodenal mucosal uptake of iron. Blood 1993; 81: 517–21.Google Scholar
Grasbeck, R, Kouvonen, I, Lundberg, M, Tenhunen, R. An intestinal receptor for heme. Scand J Haematol 1979; 23: 5–9.CrossRefGoogle ScholarPubMed
Parmley, RT, Barton, JC, Conrad, ME, Austin, RL, Holland, RM. Ultrastructural cytochemistry and radioautography of hemoglobin–iron absorption. Exp Mol Pathol 1981; 34: 131–44.CrossRefGoogle ScholarPubMed
Grasbeck, R, Majuri, R, Kouvonen, I, Tenhunen, R. Spectral and other studies on the intestinal haem receptor of the pig. Biochim Biophys Acta 1982; 700: 137–42.CrossRefGoogle ScholarPubMed
Uc, A, Stokes, JB, Britigan, BE. Heme transport exhibits polarity in Caco-2 cells: evidence for an active and membrane protein-mediated process. Am J Physiol Gastrointest Liver Physiol 2004; 287: G1150–7.CrossRefGoogle ScholarPubMed
Shayeghi, M, Latunde-Dada, GO, Oakhill, JS, et al. Identification of an intestinal heme transporter. Cell 2005; 122: 789–801.CrossRefGoogle ScholarPubMed
Qiu, A, Jansen, M, Sakaris, A, et al. Identification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorption. Cell 2006; 127: 917–28.CrossRefGoogle ScholarPubMed
Donovan, A, Brownlie, A, Zhou, Y, et al. Positional cloning of zebrafish ferroportin 1 identifies a conserved vertebrate iron exporter. Nature 2000; 403: 776–81.CrossRefGoogle ScholarPubMed
Zoller, H, Theurl, I, Koch, R, Kaser, A, Weiss, G. Mechanisms of iron mediated regulation of the duodenal iron transporters divalent metal transporter 1 and ferroportin 1. Blood Cells Mol Dis 2002; 29: 488–97.CrossRefGoogle ScholarPubMed
Graham, RM, Chua, AC, Herbison, CE, Olynyk, JK, Trinder, D. Liver iron transport. World J Gastroenterol 2007; 13: 4725–36.CrossRefGoogle ScholarPubMed
Osaki, S, Johnson, DA, Frieden, E. The possible significance of the ferrous oxidase activity of ceruloplasmin in normal human serum. J Biol Chem 1966; 241: 2746–51.Google ScholarPubMed
Roeser, HP, Lee, GR, Nacht, S, Cartwright, GE. The role of ceruloplasmin in iron metabolism. J Clin Invest 1970; 49: 2408–17.CrossRefGoogle ScholarPubMed
Vulpe, CD, Kuo, YM, Murphy, TL, et al. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nat Genet 1999; 21: 195–99.CrossRefGoogle ScholarPubMed
Anderson, GJ, Frazer, DM, Wilkins, SJ, et al. Relationship between intestinal iron-transporter expression, hepatic hepcidin levels and the control of iron absorption. Biochem Soc Trans 2002; 30: 724–6.CrossRefGoogle ScholarPubMed
Domenico, I, Ward, DM, di Patti, MC, et al. Ferroxidase activity is required for the stability of cell surface ferroportin in cells expressing GPI-ceruloplasmin. EMBO J 2007; 26: 2823–31.CrossRefGoogle ScholarPubMed
Parmley, RT, Barton, JC, Conrad, ME. Ultrastructural localization of transferrin, transferrin receptor, and iron-binding sites on human placental and duodenal microvilli. Br J Haematol 1985; 60: 81–9.CrossRefGoogle ScholarPubMed
Feder, JN, Penny, DM, Irrinki, A, et al. The hemochromatosis gene product complexes with the transferrin receptor and lowers its affinity for ligand binding. Proc Natl Acad Sci USA 1998; 95: 1472–7.CrossRefGoogle ScholarPubMed
Lebron, JA, Bennett, MJ, Vaughn, , et al. Crystal structure of the hemochromatosis protein HFE and characterization of its interaction with transferrin receptor. Cell 1998; 93: 111–23.CrossRefGoogle ScholarPubMed
Shaw, GC, Cope, JJ, Li, L, et al. Mitoferrin is essential for erythroid iron assimilation. Nature 2006; 440: 96–100.CrossRefGoogle ScholarPubMed
Quigley, JG, Yang, Z, Worthington, MT, et al. Identification of a human heme exporter that is essential for erythropoiesis. Cell 2004; 118: 757–66.CrossRefGoogle ScholarPubMed
Keel, SB, Doty, RT, Yang, Z, et al. A heme export protein is required for red blood cell differentiation and iron homeostasis. Science 2008; 319: 825–8.CrossRefGoogle ScholarPubMed
Ponka, P, Sheftel, AD. It's hepcidin again, but is it the only master?Blood 2006; 108: 3631–2.CrossRefGoogle Scholar
Yoon, D, Pastore, YD, Divoky, V, et al. Hypoxia-inducible factor-1 deficiency results in dysregulated erythropoiesis signaling and iron homeostasis in mouse development. J Biol Chem 2006; 281: 25 703–11.CrossRefGoogle ScholarPubMed
Jaakkola, P, Mole, DR, Tian, YM, et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001; 292: 468–72.CrossRefGoogle ScholarPubMed
Takeda, K, Aguila, HL, Parikh, NS, et al. Regulation of adult erythropoiesis by prolyl hydroxylase domain proteins. Blood 2008; 111: 3229–35.CrossRefGoogle ScholarPubMed
Yamashita, T, Ohneda, O, Sakiyama, A, Iwata, F, Ohneda, K, Fujii-Kuriyama, Y. The microenvironment for erythropoiesis is regulated by HIF-2alpha through VCAM-1 in endothelial cells. Blood 2008; 112: 1482–92.CrossRefGoogle ScholarPubMed
Chong, ZZ, Kang, JQ, Maiese, K. Hematopoietic factor erythropoietin fosters neuroprotection through novel signal transduction cascades. J Cereb Blood Flow Metab 2002; 22: 503–14.CrossRefGoogle ScholarPubMed
Vokurka, M, Krijt, J, Sulc, K, Necas, E. Hepcidin mRNA levels in mouse liver respond to inhibition of erythropoiesis. Physiol Res 2006; 55: 667–74.Google ScholarPubMed
Pak, M, Lopez, MA, Gabayan, V, Ganz, T, Rivera, S. Suppression of hepcidin during anemia requires erythropoietic activity. Blood 2006; 108: 3730–5.CrossRefGoogle ScholarPubMed
Tanno, T, Bhanu, NV, Oneal, PA, et al. High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin. Nat Med 2007; 13: 1096–101.CrossRefGoogle ScholarPubMed
Mast, AE, Foster, TM, Pinder, HL, et al. Behavioral, biochemical, and genetic analysis of iron metabolism in high-intensity blood donors. Transfusion 2008; 48: 2197–204.CrossRefGoogle ScholarPubMed
Brown, KE, Bacon, BR. Hepatic iron metabolism in hemochromatosis. In: Barton, JC, Edwards, CQ, eds. Hemochromatosis: Genetics, Pathophysiology, Diagnosis and Treatment. Cambridge, Cambridge University Press. 2000; 157–62.CrossRefGoogle Scholar
Hershko, C, Cook, JD, Finch, DA.Storage iron kinetics, 3. Study of desferrioxamine action by selective radioiron labels of RE and parenchymal cells. J Lab Clin Med 1973; 81: 876–86.Google ScholarPubMed
Morgan, EH, Smith, GD, Peters, TJ. Uptake and subcellular processing of 59Fe-125I-labeled transferrin by rat liver. Biochem J 1986; 237: 163–73.CrossRefGoogle ScholarPubMed
Young, SP, Roberts, S, Bomford, A. Intracellular processing of transferrin and iron by isolated rat hepatocytes. Biochem J 1985; 232: 819–23.CrossRefGoogle ScholarPubMed
Donovan, A, Lima, CA, Pinkus, JL, et al. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab 2005; 1: 191–200.CrossRefGoogle ScholarPubMed
Bastin, J, Drakesmith, H, Rees, M, Sargent, I, Townsend, A. Localization of proteins of iron metabolism in the human placenta and liver. Br J Haematol 2006; 134: 532–43.CrossRefGoogle Scholar
Canonne-Hergaux, F, Donovan, A, Delaby, C, Wang, HJ, Gros, P. Comparative studies of duodenal and macrophage ferroportin proteins. Am J Physiol Gastrointest Liver Physiol 2006; 290: G156–63.CrossRefGoogle ScholarPubMed
Lu, JP, Hayashi, K, Awai, M. Transferrin receptor expression in normal, iron-deficient and iron-overloaded rats. Acta Pathol Jpn 1989; 39: 759–64.Google ScholarPubMed
Trinder, D, Morgan, EH, Baker, E. The effects of an antibody to the rat transferrin receptor and of rat serum albumin on the uptake of diferric transferrin by rat hepatocytes. Biochim Biophys Acta 1988; 943: 440–6.CrossRefGoogle ScholarPubMed
Sciot, R, Paterson, AC, Oord, JJ, Desmet, VJ. Lack of hepatic transferrin receptor expression in hemochromatosis. Hepatology 1987; 7: 831–7.CrossRefGoogle ScholarPubMed
Lombard, M, Bomford, A, Hynes, M, et al. Regulation of the hepatic transferrin receptor in hereditary hemochromatosis. Hepatology 1989; 9: 1–5.CrossRefGoogle ScholarPubMed
Morgan, EH, Baker, E. Iron uptake and metabolism by hepatocytes. Fed Proc 1986; 45: 2810–16.Google ScholarPubMed
Trinder, D, Zak, O, Aisen, P. Transferrin receptor-independent uptake of differic transferrin by human hepatoma cells with antisense inhibition of receptor expression. Hepatology 1996; 23: 1512–20.CrossRefGoogle ScholarPubMed
Thorstensen, K, Romslo, I. Albumin prevents non-specific transferrin-binding and iron uptake by isolated hepatocytes. Biochim Biophys Acta 1984; 804: 393–7.CrossRefGoogle Scholar
Cole, ES, Glass, J. Transferrin-binding and iron uptake in mouse hepatocytes. Biochim Biophys Acta 1983; 762: 102–10.CrossRefGoogle ScholarPubMed
Kawabata, H, Yang, R, Hirama, T, et al. Molecular cloning of transferrin receptor 2. A new member of the transferrin receptor-like family. J Biol Chem 1999; 274: 20826–32.CrossRefGoogle ScholarPubMed
Chua, AC, Delima, RD, Morgan, EH, et al. Iron uptake from plasma transferrin by a transferrin receptor 2 mutant mouse model of haemochromatosis, J Hepatol 2010; 52: 425–31.CrossRefGoogle ScholarPubMed
Merle, U, Theilig, F, Fein, E, et al. Localization of the iron regulatory proteins hemojuvelin and transferrin receptor 2 to the basolateral membrane domain of hepatocytes. Histochem Cell Biol 2007; 127: 221–6.CrossRefGoogle ScholarPubMed
Goswami, T, Andrews, NC. Hereditary hemochromatosis protein, HFE, interaction with transferrin receptor 2 suggests a molecular mechanism for mammalian iron sensing. J Biol Chem 2006; 281: 28 494–8.CrossRefGoogle ScholarPubMed
Fleming, RE, Migas, MC, Holden, CC, et al. Transferrin receptor 2: continued expression in mouse liver in the face of iron overload and in hereditary hemochromatosis. Proc Natl Acad Sci USA 2000; 97: 2214–19.CrossRefGoogle ScholarPubMed
Deaglio, S, Capobianco, A, Cali, A, et al. Structural, functional, and tissue distribution analysis of human transferrin receptor 2 by murine monoclonal antibodies and a polyclonal antiserum. Blood 2002; 100: 3782–9.CrossRefGoogle Scholar
Lee, AW, Oates, PS, Trinder, D. Effects of cell proliferation on the uptake of transferrin-bound iron by human hepatoma cells. Hepatology 2003; 38: 967–77.CrossRefGoogle ScholarPubMed
Robb, A, Wessling-Resnick, M. Regulation of transferrin receptor 2 protein levels by transferrin. Blood 2004; 104: 4294–9.CrossRefGoogle ScholarPubMed
Johnson, MB, Enns, CA. Diferric transferrin regulates transferrin receptor 2 protein stability. Blood 2004; 104: 4287–93.CrossRefGoogle ScholarPubMed
Camaschella, C. Why do humans need two types of transferrin receptor? Lessons from a rare genetic disorder. Haematologica 2005; 90: 296.Google ScholarPubMed
Sarkar, B. State of iron(3) in normal human serum: low molecular weight and protein ligands besides transferrin. Can J Biochem 1970; 48: 1339–50.CrossRefGoogle ScholarPubMed
Batey, RG, Lai Chung, FP, Shamir, S, Sherlock, S. A non-transferrin-bound serum iron in idiopathic hemochromatosis. Dig Dis Sci 1980; 25: 340–6.CrossRefGoogle ScholarPubMed
Stojkovski, S, Goumakos, W, Sarkar, B. Iron(III)-binding polypeptide in human cord and adult serum: isolation, purification and partial characterization. Biochim Biophys Acta 1992; 1137: 155–61.CrossRefGoogle ScholarPubMed
Chua, AC, Olynyk, JK, Leedman, PJ, Trinder, D. Non-transferrin-bound iron uptake by hepatocytes is increased in the Hfe knockout mouse model of hereditary hemochromatosis. Blood 2004; 104: 1519–25.CrossRefGoogle Scholar
Trinder, D, Morgan, E. Inhibition of uptake of transferrin-bound iron by human hepatoma cells by non-transferrin-bound iron. Hepatology 1997; 26: 691–8.CrossRefGoogle Scholar
Graham, RM, Morgan, EH, Baker, E. Ferric citrate uptake by cultured rat hepatocytes is inhibited in the presence of transferrin. Eur J Biochem 1998; 253: 139–45.CrossRefGoogle ScholarPubMed
Scheiber-Mojdehkar, B, Zimmermann, I, Dresow, B, Goldenberg, H. Differential response of non-transferrin-bound iron uptake in rat liver cells on long-term and short-term treatment with iron. J Hepatol 1999; 31: 61–70.CrossRefGoogle ScholarPubMed
Trinder, D, Oates, PS, Thomas, C, Sadleir, J, Morgan, EH. Localization of divalent metal transporter 1 (DMT1) to the microvillus membrane of rat duodenal enterocytes in iron deficiency, but to hepatocytes in iron overload. Gut 2000; 46: 270–6.CrossRefGoogle Scholar
Worwood, M, Dawkins, S, Wagstaff, M, Jacobs, A. The purification and properties of ferritin from human serum. Biochem J 1976; 157: 97–103.CrossRefGoogle ScholarPubMed
Mack, U, Powell, LW, Halliday, JW. Detection and isolation of a hepatic membrane receptor for ferritin. J Biol Chem 1983; 258: 4672–5.Google ScholarPubMed
Osterloh, K, Aisen, P. Pathways in the binding and uptake of ferritin by hepatocytes. Biochim Biophys Acta 1989; 1011: 40–5.CrossRefGoogle ScholarPubMed
Kristiansen, M, Graversen, JH, Jacobsen, C, et al. Identification of the haemoglobin scavenger receptor. Nature 2001; 409: 198–201.CrossRefGoogle ScholarPubMed
Hvidberg, V, Maniecki, MB, Jacobsen, C, Hojrup, P, Moller, HJ, Moestrup, SK. Identification of the receptor scavenging hemopexin-heme complexes. Blood 2005; 106: 2572–9.CrossRefGoogle ScholarPubMed
Oshiro, S, Nakajima, H. Intrahepatocellular site of the catabolism of heme and globin moiety of hemoglobin-haptoglobin after intravenous administration to rats. J Biol Chem 1988; 263: 16032–8.Google ScholarPubMed
Hinton, RH, Dobrota, M, Mullock, BM. Haptoglobin-mediated transfer of haemoglobin from serum into bile. FEBS Lett 1980; 112: 247–50.CrossRefGoogle ScholarPubMed
Meilinger, M, Haumer, M, Szakmary, KA, et al. Removal of lactoferrin from plasma is mediated by binding to low-density lipoprotein receptor-related protein/alpha 2-macroglobulin receptor and transport to endosomes. FEBS Lett 1995; 360: 70–4.CrossRefGoogle ScholarPubMed
Ziere, GJ, Dijk, MC, Bijsterbosch, MK, Berkel, TJ.Lactoferrin uptake by the rat liver. Characterization of the recognition site and effect of selective modification of arginine residues. J Biol Chem 1992; 267: 11 229–35.Google ScholarPubMed
Bennatt, DJ, Ling, YY, McAbee, DD. Isolated rat hepatocytes bind lactoferrins by the RHL-1 subunit of the asialoglycoprotein receptor in a galactose-independent manner. Biochemistry 1997; 36: 8367–76.CrossRefGoogle Scholar
Niederkofler, V, Salie, R, Arber, S. Hemojuvelin is essential for dietary iron sensing, and its mutation leads to severe iron overload. J Clin Invest 2005; 115: 2180–6.CrossRefGoogle ScholarPubMed
Huang, FW, Pinkus, JL, Pinkus, GS, Fleming, MD, Andrews, NC. A mouse model of juvenile hemochromatosis. J Clin Invest 2005; 115: 2187–91.CrossRefGoogle ScholarPubMed
Feder, JN, Tsuchihashi, Z, Irrinki, A, et al. The hemochromatosis founder mutation in HLA-H disrupts beta2-microglobulin interaction and cell surface expression. J Biol Chem 1997; 272: 14,025–8.CrossRefGoogle ScholarPubMed
Ganz, T. Iron homeostasis: fitting the puzzle pieces together. Cell Metab 2008; 7: 288–90.CrossRefGoogle ScholarPubMed
Terpstra, V, Berkel, TJ. Scavenger receptors on liver Kupffer cells mediate the in vivo uptake of oxidatively damaged red blood cells in mice. Blood 2000; 95: 2157–63.Google ScholarPubMed
Dennis, C. Haemoglobin scavenger. Nature 2001; 409: 141.CrossRefGoogle ScholarPubMed
Schaer, DJ, Schaer, CA, Buehler, PW, et al. CD163 is the macrophage scavenger receptor for native and chemically modified hemoglobins in the absence of haptoglobin. Blood 2006; 107: 373–80.CrossRefGoogle ScholarPubMed
Knutson, MD, Vafa, MR, Haile, DJ, Wessling-Resnick, M. Iron loading and erythrophagocytosis increase ferroportin 1 (FPN1) expression in J774 macrophages. Blood 2003; 102: 4191–7.CrossRefGoogle ScholarPubMed
Zhang, AS, Xiong, S, Tsukamoto, H, Enns, CA. Localization of iron metabolism-related mRNAs in rat liver indicate that HFE is expressed predominantly in hepatocytes. Blood 2004; 103: 1509–14.CrossRefGoogle ScholarPubMed
Maines, MD, Gibbs, PE.Thirty some years of heme oxygenase: from a “molecular wrecking ball” to a “mesmerizing” trigger of cellular events. Biochem Biophys Res Commun 2005; 338: 568–77.CrossRefGoogle Scholar
Delaby, C, Pilard, N, Puy, H, Canonne-Hergaux, F. Sequential regulation of ferroportin expression after erythrophagocytosis in murine macrophages: early mRNA induction by heme followed by iron-dependent protein expression. Biochem J 2008; 411: 123–31.CrossRefGoogle ScholarPubMed
Delaby, C, Pilard, N, Goncalves, AS, Beaumont, C, Canonne-Hergaux, F. Presence of the iron exporter ferroportin at the plasma membrane of macrophages is enhanced by iron loading and down-regulated by hepcidin. Blood 2005; 106: 3979–84.CrossRefGoogle ScholarPubMed
Knutson, MD, Oukka, M, Koss, LM, Aydemir, F, Wessling-Resnick, M. Iron release from macrophages after erythrophagocytosis is up-regulated by ferroportin 1 over expression and down-regulated by hepcidin. Proc Natl Acad Sci USA 2005; 102: 1324–8.CrossRefGoogle Scholar
Kondo, H, Saito, K, Grasso, JP, Aisen, P. Iron metabolism in the erythrophagocytosing Kupffer cell. Hepatology 1988; 8: 32–8.CrossRefGoogle ScholarPubMed
Galli, A, Bergamaschi, G, Recalde, H, et al. Ferroportin gene silencing induces iron retention and enhances ferritin synthesis in human macrophages. Br J Haematol 2004; 127: 598–603.CrossRefGoogle ScholarPubMed
Valenti, L, Guido, M, Dongiovanni, P, Cremonesi, L, Fracanzani, AL, Fargion, S. Ferroportin 1 in the recurrence of hepatic iron overload after liver transplantation. Dig Liver Dis 2009; 41: e17–20.CrossRefGoogle ScholarPubMed
Jouanolle, AM, Douabin-Gicquel, V, Halimi, C, et al. Novel mutation in ferroportin 1 gene is associated with autosomal dominant iron overload. J Hepatol 2003; 39: 286–9.CrossRefGoogle ScholarPubMed
Wallace, DF, Clark, RM, Harley, HA, Subramaniam, VN. Autosomal dominant iron overload due to a novel mutation of ferroportin 1 associated with parenchymal iron loading and cirrhosis. J Hepatol 2004; 40: 710–13.CrossRefGoogle ScholarPubMed
Drakesmith, H, Schimanski, LM, Ormerod, E, et al. Resistance to hepcidin is conferred by hemochromatosis-associated mutations of ferroportin. Blood 2005; 106: 1092–7.CrossRefGoogle ScholarPubMed
Domenico, I, Ward, DM, Musci, G, Kaplan, J. Iron overload due to mutations in ferroportin. Haematologica 2006; 91: 92–5.Google ScholarPubMed
Birgens, HS, Kristensen, LO, Borregaard, N, Karle, H, Hansen, NE. Lactoferrin-mediated transfer of iron to intracellular ferritin in human monocytes. Eur J Haematol 1988; 41: 52–7.CrossRefGoogle ScholarPubMed
Butler, TW, Heck, LW, Berkow, R, Barton, JC. Radioimmunometric quantification of surface lactoferrin in blood mononuclear cells. Am J Med Sci 1994; 307: 102–7.CrossRefGoogle ScholarPubMed
Andreesen, R, Sephton, RG, Gadd, S, Atkins, RC, Abrew, S. Human macrophage maturation in vitro: expression of functional transferrin-binding sites of high affinity. Blut 1988; 57: 77–83.CrossRefGoogle ScholarPubMed
Berkel, TJ, Dekker, CJ, Kruijt, JK, Eijk, HG. The interaction in vivo of transferrin and asialotransferrin with liver cells. Biochem J 1987; 243: 715–22.CrossRefGoogle ScholarPubMed
Bastin, JM, Jones, M, O'Callaghan, CA, Schimanski, L, Mason, DY, Townsend, AR. Kupffer cell staining by an HFE-specific monoclonal antibody: implications for hereditary haemochromatosis. Br J Haematol 1998; 103: 931–41.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×