Current understanding of iron homeostasis

Gregory J Anderson, David M Frazer, Gregory J Anderson, David M Frazer

Abstract

Iron is an essential trace element, but it is also toxic in excess, and thus mammals have developed elegant mechanisms for keeping both cellular and whole-body iron concentrations within the optimal physiologic range. In the diet, iron is either sequestered within heme or in various nonheme forms. Although the absorption of heme iron is poorly understood, nonheme iron is transported across the apical membrane of the intestinal enterocyte by divalent metal-ion transporter 1 (DMT1) and is exported into the circulation via ferroportin 1 (FPN1). Newly absorbed iron binds to plasma transferrin and is distributed around the body to sites of utilization with the erythroid marrow having particularly high iron requirements. Iron-loaded transferrin binds to transferrin receptor 1 on the surface of most body cells, and after endocytosis of the complex, iron enters the cytoplasm via DMT1 in the endosomal membrane. This iron can be used for metabolic functions, stored within cytosolic ferritin, or exported from the cell via FPN1. Cellular iron concentrations are modulated by the iron regulatory proteins (IRPs) IRP1 and IRP2. At the whole-body level, dietary iron absorption and iron export from the tissues into the plasma are regulated by the liver-derived peptide hepcidin. When tissue iron demands are high, hepcidin concentrations are low and vice versa. Too little or too much iron can have important clinical consequences. Most iron deficiency reflects an inadequate supply of iron in the diet, whereas iron excess is usually associated with hereditary disorders. These disorders include various forms of hemochromatosis, which are characterized by inadequate hepcidin production and, thus, increased dietary iron intake, and iron-loading anemias whereby both increased iron absorption and transfusion therapy contribute to the iron overload. Despite major recent advances, much remains to be learned about iron physiology and pathophysiology.

Keywords: anemia; ferritin; hemochromatosis; hepcidin; iron deficiency; iron overload; iron physiology; transferrin.

© 2017 American Society for Nutrition.

Figures

FIGURE 1
FIGURE 1
Body iron homeostasis. Iron is present in the diet in both heme and nonheme forms. Although the mechanisms underlying heme absorption are poorly understood, nonheme iron enters the circulation after traversing the enterocyte apical membrane via DMT1 and the basolateral membrane via FPN1. Iron binds to plasma TF and is distributed to tissues throughout the body. Quantitatively, most iron is used by immature red blood cells in the bone marrow for hemoglobin production. Senescent erythrocytes are phagocytosed by macrophages, and the iron is released from catabolized hemoglobin and re-enters the circulation. The liver-derived peptide hepcidin plays a critical role in the regulation of body iron intake and distribution by binding to plasma membrane FPN1 on enterocytes, macrophages, and most body cells and facilitating its internalization and degradation. Hepcidin, in turn, is regulated by body iron demand. Thus, when the body is iron deficient, hepcidin concentrations are low, thereby favoring iron absorption and delivery to the plasma from storage sites; but when the body is iron replete, a higher hepcidin concentration reduces iron absorption and impairs iron release from stores. DCYTB, duodenal cytochrome b; DMT1, divalent metal-ion transporter 1; FPN, ferroportin; HP, hephaestin; TF, transferrin.
FIGURE 2
FIGURE 2
Cellular iron homeostasis. Cells can take up iron in a variety of forms, but all nucleated cells have the capacity to use TF-bound iron. Diferric TF binds to TFR1 on the plasma membrane, and the complex is internalized in endosomes. Acidification of the endosome, which is accompanied by iron reduction by a member of the STEAP family of proteins, releases iron from TF, which subsequently moves across the endosomal membrane via DMT1 and into the cytoplasm. The precise form of this cytosolic iron pool is unclear, but at least some iron is bound by PCBP proteins that act as iron chaperones. These complexes can deliver iron to newly synthesized iron-containing proteins (although whether they can deliver iron to mitochondria is unclear) and to the iron-storage protein ferritin. Iron in excess of cellular needs may be exported through FPN1 with the iron oxidase CP increasing the efficiency of this process. Some cell types can take up iron in other forms, including NTBI, or iron that is contained within ferritin, heme, or hemoglobin. Cellular iron intake and storage are regulated by the iron-responsive element and IRP system such that low iron concentrations favor the synthesis of more TFR1 and suppress ferritin expression, whereas high cellular iron leads to an increase in ferritin concentrations and a decrease in TFR1. CP, ceruloplasmin; DMT1, divalent metal-ion transporter 1; FPN1, ferroportin 1; IRP, iron regulatory protein; LRP, LDL-receptor–related protein; mRNA, messenger RNA; NTBI, non–transferrin-bound iron; PCBP, poly(rC)-binding protein; STEAP, 6-transmembrane epithelial antigen of the prostate; TF, transferrin; TFR1, transferrin receptor 1; ZIP14, Zrt/Irt-like protein 14.

References

    1. Ganz T. Systemic iron homeostasis. Physiol Rev 2013;93:1721–41.
    1. Carpenter CE, Mahoney AW. Contributions of heme and nonheme iron to human nutrition. Crit Rev Food Sci Nutr 1992;31:333–67.
    1. Fuqua BK, Vulpe CD, Anderson GJ. Intestinal iron absorption. J Trace Elem Med Biol 2012;26:115–9.
    1. 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.
    1. McKie AT, Barrow D, Latunde-Dada GO, Rolfs A, Sager G, Mudaly E, Mudaly M, Richardson C, Barlow D, Bomford A, et al. . An iron-regulated ferric reductase associated with the absorption of dietary iron. Science 2001;291:1755–9.
    1. Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine S, Andrews NC. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab 2005;1:191–200.
    1. Vulpe CD, Kuo YM, Murphy TL, Cowley L, Askwith C, Libina N, Gitschier J, Anderson GJ. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nat Genet 1999;21:195–9.
    1. Theil EC, Chen H, Miranda C, Janser H, Elsenhans B, Nunez MT, Pizarro F, Schumann K. Absorption of iron from ferritin is independent of heme iron and ferrous salts in women and rat intestinal segments. J Nutr 2012;142:478–83.
    1. Frazer DM, Anderson GJ. The regulation of iron transport. Biofactors 2014;40:206–14.
    1. Fleming RE, Ponka P. Iron overload in human disease. N Engl J Med 2012;366:348–59.
    1. Bainton DF, Finch CA. The diagnosis of iron deficiency anemia. Am J Med 1964;37:62–70.
    1. Bullock GC, Delehanty LL, Talbot AL, Gonias SL, Tong WH, Rouault TA, Dewar B, Macdonald JM, Chruma JJ, Goldfarb AN. Iron control of erythroid development by a novel aconitase-associated regulatory pathway. Blood 2010;116:97–108.
    1. Ohgami RS, Campagna DR, Greer EL, Antiochos B, McDonald A, Chen J, Sharp JJ, Fujiwara Y, Barker JE, Fleming MD. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat Genet 2005;37:1264–9.
    1. Fleming MD, Romano MA, Su MA, Garrick LM, Garrick MD, Andrews NC. Nramp2 is mutated in the anemic Belgrade (b) rat: evidence of a role for Nramp2 in endosomal iron transport. Proc Natl Acad Sci USA 1998;95:1148–53.
    1. Sharp P. The molecular basis of copper and iron interactions. Proc Nutr Soc 2004;63:563–9.
    1. Jenkitkasemwong S, Wang CY, Coffey R, Zhang W, Chan A, Biel T, Kim JS, Hojyo S, Fukada T, Knutson MD. SLC39A14 is required for the development of hepatocellular iron overload in murine models of hereditary hemochromatosis. Cell Metab 2015;22:138–50.
    1. Gelvan D, Fibach E, Meyron-Holtz EG, Konijn AM. Ferritin uptake by human erythroid precursors is a regulated iron uptake pathway. Blood 1996;88:3200–7.
    1. Anderson GJ, Frazer DM. Hepatic iron metabolism. Semin Liver Dis 2005;25:420–32.
    1. Leidgens S, Bullough KZ, Shi H, Li F, Shakoury-Elizeh M, Yabe T, Subramanian P, Hsu E, Natarajan N, Nandal A, et al. . Each member of the poly-r(C)-binding protein 1 (PCBP) family exhibits iron chaperone activity toward ferritin. J Biol Chem 2013;288:17791–802.
    1. Theil EC. Ferritin: the protein nanocage and iron biomineral in health and in disease. Inorg Chem 2013;52:12223–33.
    1. Wilkinson N, Pantopoulos K. The IRP/IRE system in vivo: insights from mouse models. Front Pharmacol 2014;5:176.
    1. Harms K, Kaiser T. Beyond soluble transferrin receptor: old challenges and new horizons. Best Pract Res Clin Endocrinol Metab 2015;29:799–810.
    1. Speeckaert MM, Speeckaert R, Delanghe JR. Biological and clinical aspects of soluble transferrin receptor. Crit Rev Clin Lab Sci 2010;47:213–28.
    1. Peyssonnaux C, Nizet V, Johnson RS. Role of the hypoxia inducible factors HIF in iron metabolism. Cell Cycle 2008;7:28–32.
    1. Guo W, Bachman E, Li M, Roy CN, Blusztajn J, Wong S, Chan SY, Serra C, Jasuja R, Travison TG, et al. . Testosterone administration inhibits hepcidin transcription and is associated with increased iron incorporation into red blood cells. Aging Cell 2013;12:280–91.
    1. Hou Y, Zhang S, Wang L, Li J, Qu G, He J, Rong H, Ji H, Liu S. Estrogen regulates iron homeostasis through governing hepatic hepcidin expression via an estrogen response element. Gene 2012;511:398–403.
    1. Anderson GJ, Vulpe CD. Mammalian iron transport. Cell Mol Life Sci 2009;66:3241–61.
    1. Ganz T, Nemeth E. Iron homeostasis in host defence and inflammation. Nat Rev Immunol 2015;15:500–10.
    1. Lakhal-Littleton S, Wolna M, Carr CA, Miller JJ, Christian HC, Ball V, Santos A, Diaz R, Biggs D, Stillion R, et al. . Cardiac ferroportin regulates cellular iron homeostasis and is important for cardiac function. Proc Natl Acad Sci USA 2015;112:3164–9.
    1. Finch C. Regulators of iron balance in humans. Blood 1994;84:1697–702.
    1. Anderson GJ, Powell LW, Halliday JW. Transferrin receptor distribution and regulation in the rat small intestine. Effect of iron stores and erythropoiesis. Gastroenterology 1990;98:576–85.
    1. Guiang SF III, Georgieff MK, Lambert DJ, Schmidt RL, Widness JA. Intravenous iron supplementation effect on tissue iron and hemoproteins in chronically phlebotomized lambs. Am J Physiol 1997;273:R2124–31.
    1. Milman N. Iron prophylaxis in pregnancy–general or individual and in which dose? Ann Hematol 2006;85:821–8.
    1. Schroeder BO, Backhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nat Med 2016;22:1079–89.
    1. Zimmermann MB, Chassard C, Rohner F, N’Goran EK, Nindjin C, Dostal A, Utzinger J, Ghattas H, Lacroix C, Hurrell RF. The effects of iron fortification on the gut microbiota in African children: a randomized controlled trial in Cote d’Ivoire. Am J Clin Nutr 2010;92:1406–15.
    1. Jaeggi T, Kortman GA, Moretti D, Chassard C, Holding P, Dostal A, Boekhorst J, Timmerman HM, Swinkels DW, Tjalsma H, et al. . Iron fortification adversely affects the gut microbiome, increases pathogen abundance and induces intestinal inflammation in Kenyan infants. Gut 2015;64:731–42.
    1. Zimmermann MB, Hurrell RF. Nutritional iron deficiency. Lancet 2007;370:511–20.
    1. Beard JL, Durward C. The liabilities of iron deficiency Anderson GJ, McLaren GD, editors. Iron physiology and pathophysiology in humans New York: Springer; 2012. p. 283–302.
    1. Finberg KE, Heeney MM, Campagna DR, Aydinok Y, Pearson HA, Hartman KR, Mayo MM, Samuel SM, Strouse JJ, Markianos K, et al. . Mutations in TMPRSS6 cause iron-refractory iron deficiency anemia (IRIDA). Nat Genet 2008;40:569–71.
    1. Pasricha SR, Flecknoe-Brown SC, Allen KJ, Gibson PR, McMahon LP, Olynyk JK, Roger SD, Savoia HF, Tampi R, Thomson AR, et al. . Diagnosis and management of iron deficiency anaemia: a clinical update. Med J Aust 2010;193:525–32.
    1. Lopez A, Cacoub P, Macdougall IC, Peyrin-Biroulet L. Iron deficiency anaemia. Lancet 2016;387:907–16.
    1. Cancelo-Hidalgo MJ, Castelo-Branco C, Palacios S, Haya-Palazuelos J, Ciria-Recasens M, Manasanch J, Perez-Edo L. Tolerability of different oral iron supplements: a systematic review. Curr Med Res Opin 2013;29:291–303.
    1. Anderson GJ. Mechanisms of iron loading and toxicity. Am J Hematol 2007;82:1128–31.
    1. Barton JC, Edwards CQ, Acton RT. HFE gene: structure, function, mutations, and associated iron abnormalities. Gene 2015;574:179–92.
    1. Deugnier Y, Turlin B. The pathology of hepatic iron overload In: Anderson GJ, McLaren GD, editors. Iron physiology and pathophysiology in humans. New York: Springer; 2012. p. 345–83.
    1. Aigner E, Theurl I, Theurl M, Lederer D, Haufe H, Dietze O, Strasser M, Datz C, Weiss G. Pathways underlying iron accumulation in human nonalcoholic fatty liver disease. Am J Clin Nutr 2008;87:1374–83.
    1. Reid DW, Anderson GJ, Lamont IL. Cystic fibrosis: ironing out the problem of infection? Am J Physiol Lung Cell Mol Physiol 2008;295:L23–4.
    1. Farina M, Avila DS, da Rocha JB, Aschner M. Metals, oxidative stress and neurodegeneration: a focus on iron, manganese and mercury. Neurochem Int 2013;62:575–94.
    1. Olivieri NF, Brittenham GM. Management of the thalassemias. Cold Spring Harb Perspect Med 2013;3:a011601.
    1. Chua K, Fung E, Micewicz ED, Ganz T, Nemeth E, Ruchala P. Small cyclic agonists of iron regulatory hormone hepcidin. Bioorg Med Chem Lett 2015;25:4961–9.
    1. Arezes J, Nemeth E. Hepcidin and iron disorders: new biology and clinical approaches. Int J Lab Hematol 2015;37 Suppl 1:92–8.
    1. Weiss G, Goodnough LT. Anemia of chronic disease. N Engl J Med 2005;352:1011–23.
    1. Guida C, Altamura S, Klein FA, Galy B, Boutros M, Ulmer AJ, Hentze MW, Muckenthaler MU. A novel inflammatory pathway mediating rapid hepcidin-independent hypoferremia. Blood 2015;125:2265–75.

Source: PubMed

3
S'abonner