Open access peer-reviewed chapter - ONLINE FIRST

Dietary Iron Uptake and Absorption

Written By

Aderinola Awoniyi, Oreoluwa Daniel and Oladimeji Babatunde

Submitted: 22 May 2023 Reviewed: 06 February 2024 Published: 05 July 2024

DOI: 10.5772/intechopen.114277

Metabolism - Annual Volume 2024 IntechOpen
Metabolism - Annual Volume 2024 Authored by Yannis Karamanos

From the Annual Volume

Metabolism - Annual Volume 2024 [Working Title]

Prof. Yannis Karamanos

Chapter metrics overview

1 Chapter Downloads

View Full Metrics

Abstract

Iron is an essential element that participates in many physiological roles in the human body, including oxygen transport, DNA synthesis, cell division and differentiation, immunity, and electron transport. Iron absorption takes place primarily on the duodenum of the small intestine through the enterocyte cell. Its levels in the human body are controlled only by absorption since unlike most other essential nutrients, it has no regulated excretory system in humans. Hence, maintenance of iron level is critical to avoid adverse physiological consequences of iron deficiency or overload. Dietary iron exists in two forms; haem and non-haem; the bioavailability of these forms of iron is dependent on dietary inhibitors (calcium, phytates, and polyphenols) and enhancers (ascorbic acid). The mechanism that regulates iron absorption occurs in three stages; (i) luminal uptake and transport of iron across the apical membrane (ii) transfer of iron to the basolateral membrane and (iii) transport of iron across the basolateral membrane into circulation. The proteins that mediate iron uptake at the various stages include divalent metal transporter I, ferroportin, hephaestin, and hepcidin. This chapter will elaborate on dietary iron in its different forms, factors that enhance and inhibit iron absorption, the mechanism by which it is absorbed and iron supplementation and fortification.

Keywords

  • haem iron
  • non-haem iron
  • Iron absorption
  • duodenal cytochrome b
  • divalent metal transporter I
  • ferroprotin
  • transferrin
  • ferritin
  • hephaestin
  • hepcidin

1. Introduction

Healthy living requires an adequate supply of essential metals, and one such is iron. Iron plays a crucial role in enzyme catalysis, oxygen transport, electron transport, cell division and differentiation, and regulation of gene expression [1, 2, 3]. Despite its indispensability, when it is excessively present in diets, it can be toxic as it is capable of catalyzing the formation of reactive oxygen species [1]. Given the dual tendencies of iron, the concentration of the essential metal in the body must be kept within bounds [1]. The levels of iron in the human body are regulated mainly by the amount absorbed per time [4]. The mechanism of iron excretion is an unregulated process, and it occurs via loss of sweat, menstruation, shedding of hair and skin cells, and rapid turnover and excretion of enterocytes [4]. In general, iron is present in diets as either haem or non-haem. Of the two forms, the non-haem form is by far the most prevalent accounting for 80–90% of the iron in a standard diet, but the less absorbed [5, 6]. The haem form accounts for 10–20% of the iron in a standard diet and is the more absorbed of the two forms [5, 6]. The sources of the two forms of iron are diverse. The haem form is derived from hemoglobin and myoglobin of animal food sources such as meat, fish, and poultry; whereas, the non-haem form is derived from plants such as fruits, vegetables, dried beans, nuts, and grain products and iron-fortified foods [4, 7].

The absorption of iron is influenced by certain dietary factors including the type of food consumed and the phytochemical constituent. Inadequate iron absorption leads to iron deficiency anemia, and excessive iron absorption leads to iron overload, a condition often caused by transfusion therapies used to treat thalassemia and other anemias [8, 9]. Iron levels are controlled mainly by regulating iron absorption in the duodenum. This mechanism allows for an increase or decrease in iron levels depending on the physiological demand.

Advertisement

2. Haem and non-haem iron

Dietary iron exists as haem and non-haem iron. Haem iron can be obtained mainly from hemoglobin and myoglobin of meat. Haem iron is well absorbed, and it is not influenced by other dietary constituents [10]. It can be sourced from meat, poultry, liver, spleen, and boiled egg yolk (Figure 1) [11]. Non-haem iron contains largely ferrous iron and can be obtained from both animal and plant sources. Unlike heam iron, the absorption of non-haem iron is low and is influenced by other constituents of the diet [10]. Sources of non-haem iron include whole grains, cereals, pulses, green leafy vegetables like pumpkin leaves, radish leaves, mustard leaves, spinach, curry, mint, parsley, coriander, and drumstick (Figure 1) [11].

Figure 1.

Dietary sources of iron (A) sources of haem iron (B) sources of non-haem iron.

Advertisement

3. Dietary iron absorption

Iron absorption takes place in the duodenum and proximal jejunum, and it is essential for maintaining iron balance in the body as the body does not have an active excretory system [12]. The absorption of iron is dependent on many factors, one of which is the physical state of iron. Iron exists in the ferric state (Fe3+) at physiological pH; however, for it to be absorbed, it must be in the ferrous state (Fe2+) because ferrous ion does not undergo polymerization and their solubility is greater than ferric ions [13]. Also, the quantity of iron stores has an impact on its absorption. It has been gathered from previous studies that iron absorption has an inverse relationship to the quantity of iron stored in the body [14, 15].

3.1 Factors influencing dietary iron absorption

3.1.1 Factors influencing haem iron absorption

  • Amount of dietary haem iron,

  • Iron status of the subject: Haem iron from animal sources is well absorbed in humans and contributes to more than 10% of the total absorbed iron [16]. The absorption varies from 15 to 25% in normal subjects and 25 to 35% in iron-deficient subjects [11].

  • Content of calcium in meal (e.g., milk and cheese)

  • Food preparation (time and temperature): Studies have shown that baking and prolonged frying reduces absorption of haem iron by about 40% [11, 15]

3.1.2 Factors influencing non-haem iron Absorption

  • Amount of potentially available non-haem iron

  • Iron status of the subject: The absorption of non-heam iron varies between 2 and 20%. Absorption of non-haem iron is higher in individuals who are severely iron deficient compared to individuals with normal level. Individuals who are anemic and pregnant have the highest absorption rates (5–13%)

  • Other constituents in the diet: The type of food ingested along with plant food determines the rate at which the non-haem iron in the plant is absorbed. If the food contains reducing substances like ascorbic acid, the iron will be kept in the reduced ferrous form and will be better absorbed [11, 15].

3.2 Inhibitors and enhancers of iron absorption

The duodenal pH-dependent process of iron absorption is modulated by certain dietary compounds [4]. These dietary compounds can either be inhibitors or enhancers. Figure 2 shows the main enhancers and inhibitors of iron absorption. One primary enhancer of iron absorption is ascorbic acid. As an enhancer, ascorbic acid in the diet increases the absorption of non-haem iron [17]. Mechanistically, ascorbic acid chelates ferric ions in the stomach at an acidic pH, keeping the ferric ion soluble until it reaches the duodenum of the small intestine. At the duodenum, ascorbic acid donates an electron to a ferric ion, acting as a free radical scavenger and reducing it to a ferrous ion, the absorbable form of iron that crosses the brush border membrane of the enterocytes [18]. It is observed that the effect of ascorbic acid is dose-dependent and can increase the absorption of iron only when both nutrients are consumed together [19, 20, 21]. More still, ascorbic acid can overcome the effects of all dietary inhibitors of iron absorption when it is included in a diet with high non-haem iron [4]. Another enhancer of iron absorption is animal tissues such as those of beef, chicken, fish, pork, and lamb [22]. A study carried out by [23] observed that meat increases iron absorption by inactivating the luminal factors that prevent iron absorption. The authors further asserted that meat forms a luminal transporter that conveys iron from the intestinal lumen to the mucosal cell membrane and finally to the enterocyte cytoplasm.

Figure 2.

Main enhancer and inhibitors of iron absorption. Ascorbic acid is the main enhancer of iron absorption, while phytate, polyphenols, oxalate, and calcium ion act as inhibitors of iron absorption. Proteins can either act as inhibitors or enhancers of iron absorption depending on the source.

In contrast, inhibitors of iron transport prevent the bioavailability of the essential metal. The most prominent inhibitors are phytate and polyphenols found in plant-based foods [24]. Phytate is a naturally occurring component found in plants, and it has an inhibitory effect on the bioavailability of most minerals [25]. Phytate cannot be degraded by humans due to the lack of endophytases, and it is not absorbed in the small intestine. Hence, minerals chelated in phytate are not bioavailable [26, 27]. Polyphenols are abundantly found in human diets, namely vegetables, cereals, spices, tea, coffee, red wine, and cocoa and work by forming a complex with iron [28, 29, 30]. However, the precise mechanism by which polyphenols reduce the bioavailability of iron is still to a large extent unclear [31, 32]. Also, calcium is a known inhibitor of iron absorption. Whereas, calcium inhibits both haem and non-haem iron, phytate, and polyphenols inhibit only non-haem iron [33]. Although the exact mechanism of calcium inhibition is still unknown, however, certain possibilities have been proposed Ref. [33] suggested that calcium inhibition of iron absorption occurs during the efflux of iron into the portal circulation. Conversely, [34] proposed that calcium inhibition of iron absorption occurs during the initial entry of iron into the enterocytes. In either case, it is multiply attested that calcium is an inhibitor of iron absorption. Depending on their source, certain proteins have been reported to be inhibitors and enhancers of iron absorption [32]. While proteins from meat were reported to be enhancers of iron absorption, other proteins such as those from egg, soybean, and casein were reported to be inhibitors of iron absorption [23, 35, 36, 37]. To a large extent, oxalate is an inhibitor of mineral bioavailability [38, 39, 40]. However, its impact on iron absorption is debatable. Studies on rats have shown that the effect of oxalate on ferric ion absorption is insignificant; however, when the iron is in the ferrous form, oxalate has been observed to limit its absorption by forming insoluble ferrous oxalate [41].

Advertisement

4. Mechanism of iron absorption

4.1 Haem iron absorption

As asserted earlier, haem iron is more efficiently absorbed than non- haem iron. Before haem iron is made available for absorption, it must first be released from proteins such as hemoglobin and myoglobin by proteolytic cleavage in the intestinal lumen [42]. Afterward, the haem moiety, with its bound iron, is absorbed intact through the brush border membrane of the intestinal enterocytes [6]. Afterward, the haem moiety, with its bound iron, is absorbed intact through the brush border membrane of the intestinal enterocytes [6]. The fact that iron is not released from haem before absorption makes haem iron more efficiently absorbed than non- haem iron, which readily binds to a variety of substances in the diet reducing its bioavailability [43, 44, 45, 46]. Haem carrier protein (HCP1) mediates the absorption of haem iron and transports it across the apical membrane [47]. Upon arrival at the cytoplasm, iron is released from the protoporphyrin moiety most likely by haem oxygenase 1 (HO-1) [45, 48, 49]. Afterward, the protoporphyrin moiety is degraded, and the iron released joins the pathway observed in non-haem iron efflux into the portal circulation [1].

4.2 Non-haem iron absorption

Figure 3 shows the major pathways of non-haem iron absorption.

Figure 3.

Major pathways of non-heme iron absorption. Ferric ion is converted to ferrous ion by the ferric reductase enzyme, duodenal cytochrome B (Dcytb) on the brush border membrane of the enterocytes. Upon the reduction of Fe (III) to Fe (II), an integral membrane protein of the enterocytes called divalent metal cation transporter 1 (DMT1) transports Fe (II) across the membrane into the enterocyte cytoplasm. If iron is required by the body, it forms a complex with hephaestin and is rapidly transported across the basolateral membrane by the protein ferroportin 1 (FPN), and then, it binds to transferrin for distribution into the body through portal circulation. If iron is not required by the body, it is stored as ferritin in the enterocyte cytoplasm.

4.2.1 Ferric ion reduction

A large percentage of the non-haem iron present in diets is in the oxidized or ferric form, Fe (III), which must first be reduced to the ferrous form, Fe (II) before it can be taken up by the duodenum and proximal jejunum of the intestinal epithelial cells or enterocytes [1, 4]. A principal candidate for facilitating this reduction is the ferric reductase enzyme, duodenal cytochrome B (Dcytb) on the brush border membrane of the enterocytes (Figure 3) [3]. This enzyme is pH-sensitive and requires the low pH of the gastric acid in the proximal duodenum for activation.

4.2.2 Iron uptake across the apical membrane

Subsequent to reduction of Fe (III) to Fe (II), non-haem iron makes its entry into the enterocytes through a transporter known as divalent metal transporter (DMT1) (Figure 3) [50]. DMT1 is an apical brush border membrane transporter, and its expression is increased by the hypoxia- inducible factor-2α during iron deficiency [51]. DMT1 imports iron into the cytosol of absorptive cells after which it is either transferred across the basolateral membrane or bound to ferritin for storage; the body’s demand for iron determines the pathway it follows.

4.2.3 Iron efflux across basolateral membrane

When iron is in low demand, the body stores it as ferritin (a cytosolic iron storage protein). However, when required, ferrous iron from both haem and non-haem iron is transferred from the apical side to the basolateral side [47]. In a study by Das et al. [52], it was reported that a nuclear receptor coactivator NCOA4 is required for the transport of iron within the enterocytes. The NCOA4 is commonly expressed as a “coiled-coil” domain responsible for the promotion of iron release from ferritin. This occurs by aiding ferritin delivery to lysosomes for degradation through a process called ferritinophagy [53]. It was observed that the induction of NCOA4 messenger RNA and protein in the duodenum of mouse occurred as a form of response to iron deficiency induced by diet and hemolytic anemia induced chemically [52].

The proteins ferroportin 1 [(FPN1) (a transmembrane protein with 12 domains that can only transport ferrous iron)] and hephaestin [(HEPH), a multicopper ferroxidase] act to transport iron across the basolateral membrane (Figure 3) [5455]. After the exportation of ferrous iron from the enterocytes, hephaestin converts ferrous iron (Fe2+) to ferric iron (Fe3+) by oxidation. The ferric iron is then bound to transferrin for onward distribution throughout the body through plasma circulation [56]. Each transferrin has the ability to bind two Fe3+ to form TF-[Fe3+]2 complex which binds to transferrin receptor (TFR1) on the cell surface and is absorbed into cells to form endosome [57]. Subsequently, a six transmembrane prostate epithelial antigen 3 (STEAP3) reduces Fe3+ to Fe2+ and then transported by DMTI to the cytoplasm to perform physiological functions [50, 58].

4.3 Regulation of iron absorption

Iron homeostasis is a tightly regulated process. The regulation of non- haem iron absorption has been studied extensively, and since haem iron absorption shares many similarities with non-haem iron absorption, the regulatory mechanism employed in the latter can be extrapolated on the former. Most studies have shown that iron deficiency can stimulate the absorption of both haem and non-haem iron [43, 46, 49, 59, 60, 61, 62]. Furthermore, erythropoiesis is a factor that affects the rate of absorption of haem and non-haem iron. However, the influence of erythropoiesis on iron absorption is specie specific.

At the molecular level, the efflux of iron across the basolateral membrane is regulated almost exclusively by hepcidin levels [4, 63, 64]. Hepcidin is a twenty-five (25) amino acid peptide hormone, synthesized in the liver whose level decreases when body iron requirements are high and increases when iron requirements decline [54].

When iron levels are high, that is body iron requirements decline, hepcidin binds the ferroportin/hephaestin complex resulting in its internalization and degradation (Figure 4), effectively shunting cellular iron into ferritin stores, and preventing its absorption into the portal circulation [4]. The binding of ferroportin/hephastin to hepcidin results in the ubiquitination of major lysine residues, endocytosis, and degradation in lysosomes, inhibiting iron entry into the bloodstream [65].

Figure 4.

Regulation of iron absorption (A) increased hepcidin expression by the liver results in the internalization and degradation of ferroportin. Loss of ferroprotin on the cell surface results in low plasma iron level. (B) Normal hepcidin levels respond to iron demand and regulate iron import into the plasma. (C) Iron overload results from reduced levels of hepcidin causing an efflux of iron into the plasma.

Ferroportin undergoes ubiquitination by the oxidation and demethylation of 5-methylcytosines in DNA, carried out by an iron- and 2- oxoglutarate-dependent dioxygenase Tet. This leads to the induction of the ferroportin’s endocytosis and proteolysis. Ubiquitination is carried out by a flow of enzymes E1, E2, and E3, this they do by activating ubiquitin, transferring it, and ultimately ligating it to a lysine side chain in the target protein, respectively. Jiang et al. [66] reported that in the absence of Rnf217 in macrophages, there was a slower hepcidin-dependent ferroportin degradation, and this suggests that some other E3 ligases may also be involved in the regulation of the degradation of ferroportin [66]. In situations when body iron requirements are high and the concentration of hepcidin is decreased, ferroportin/hephastin complex readily signals the translocation of iron into the portal circulation (Figure 4).

Another protein involved in iron homeostasis is hereditary hemochromatosis protein (HFE). The HFE protein is expressed by the hepatocytes, and it competes with transferrin binding to TFR1, hence lowering iron uptake into cells [67]. Alternately, binding of transferrin to TFR1 releases HFE to bind to TFR2 in hepatocytes, thereby increasing hepcidin transcription [68].

Furthermore, iron homeostasis is regulated by iron-responsive element-binding protein (IRPs) which consists of both IRP1 and IRP2. In order to mediate homeostasis, IRP binds to the corresponding IRE on the untranslated region of mRNA encoding the protein essential for cellular iron regulation (TFR1, DMT1), thereby participating in iron uptake (TFR1), storage (ferritin), redistribution, and efflux (FPN1) [69]. When intracellular iron is low, IRP can inhibit the translation of FPN1 and ferritin but increase the synthesis of TFR1. On the contrary, when intracellular iron is surplus, synthesis of FPN1 and ferritin is increased while TFR1 is degraded [70].

Another mechanism contributing to systemic iron regulation is the hypoxia-inducible factor (HIF) prolyl hydroxylases, and they have been discovered to not only be sensitive to hypoxia but also concentrations of iron. These enzymes sense oxygen and iron, thereby regulating HIF, and through this regulation, they control some specific gene transcriptions which are responsible for the absorption, distribution, and utilization of iron [71].

Advertisement

5. Iron supplementation and fortification

The high prevalence of iron-deficiency anemia among pregnant women and young children has become an issue of global concern [72]. This prevalence is highest in resource-poor countries that can rarely afford diets rich in iron. Moreso, the poor diet alongside high levels of infection limits the uptake of iron and results in many pathological conditions [73]. Hence, prevention of iron deficiency and anemia has become a major public health goal. It is therefore a necessity to design ways of delivering additional iron effectively to the affected population. Therefore, WHO recommended that governments of countries with high prevalence implement programs that can help in the reduction of iron deficiency anemia. One of such programs is supplementation for young children and pregnant women [74, 75].

Oral administration of iron supplement is the most frequently used method for iron delivery because it is convenient, not expensive, and not restricted by external conditions [76]. The first generation of iron supplementation is ferrous sulfate (FeSO4) tablets. Although FeSO4 is easy to handle and affordable, it has the capability of reacting with sulfide and polyphenol to cause food decoloration and deterioration. This has a negative impact on the gastrointestinal tract, leading to poor absorption. FeSO4 and other inorganic iron can also trigger nausea, vomiting, and diarrhea in individuals that use them [77, 78]. The absorption of iron from the second-generation iron supplements (ferrous citrate, ferrous gluconate, ferrous fumarate, and ferrous lactate) had an improvement over the first generation. The supplements are organic acids iron salt chelated with a small molecule; therefore, the organic acid and iron interact together, and this helps in the gradual release of iron in the acidic gastric stage avoiding rapid surges [79]. However, despite this improvement, these second-generation iron supplements still caused adverse gastrointestinal reactions [80, 81]. The problem was overcome by developing chelated and encapsulated iron which improved the stability of iron in the intestinal tract. Therefore, iron microcapsules that are nanoparticle-based which demonstrated better bioavailability and safety have now emerged as novel oral iron supplements [79].

Another strategy for improving iron status is iron fortification. This process involves the addition of iron-containing substances to the product recipe, either as isolated compounds (e.g., iron salts or chelates) or as iron-rich ingredients (e.g., meat or its derivatives) [73]. It is believed that if iron is delivered through the natural matrix of food the adverse effects experienced with supplementation will be reduced. Iron fortification therefore involves delivering small doses of iron into a food vehicle at the point of manufacturing [73]. Although this method raises body iron levels slowly compared with iron supplementation, however, it is considered to be safer [82].

Iron fortification is usually done with staple food items like rice, oil, and wheat; condiments such as fish sauce, soy sauce, and lentils; salt and sugar; processed food items like infant complementary food, dairy products, and noodles [83]. However, the addition of iron to different food item has produced technical challenges over the years [84]. One of such is induction of organoleptic changes that renders the taste of the food item unacceptable to consumers. Since iron has the ability to transit between two oxidation states, it creates chemical instability with the food matrix, thereby resulting in changes in the taste [85]. Albeit, this technical obstacle was overcome by using insoluble, poorly soluble, or strongly chelated iron compounds which have limited chemical activity [86].Therefore, the recommended iron salts for fortification by WHO are ferrous sulfate, ferrous fumarate, ferric pyrophosphate, and electric iron powder [87, 88].

Fortification of cereal flour with iron is considered to be cost-effective and sustainable way of improving iron status in deficient populations [89]. Since wheat is widely consumed staple by about one-third of the world’s population [90], it will be a suitable vehicle for iron fortification. Moreso, WHO and other relevant stakeholders have endorsed NaFeEDTA as the only iron salt suitable for use in fortification of high-extraction flours [91]. This is because NaFeEDTA binds strongly to ferric iron at gastric juice pH in the stomach and exchanges it for other metals in the duodenum as the pH increases, thereby protecting iron from the phytic acid present in food consumed [73]. NaFeEDTA has also been used effectively as fortificant in food vehicles like curry powder, sugar, fish sauce, and maize flour [72, 88, 92, 93].

Technology has also been developed for the fortification of salt with iron since it is also widely consumed. The National Institute of Nutrition has developed standards whereby 1 g of salt is fortified with 1 mg of iron and 15 μg of iodine. This provides about 30–60% of RDA of 17 mg of an adult man consuming 5–10 g of salt per day [93]. This platform of fortification of salt with iron is preferred to fortification of cereals because a relatively smaller volume of the foodstuff is required. In addition, iodization of salt is nearly universal, and therefore, it will be easy to suggest a double-fortified salt (iron-fortified iodized salt) to the populace [11].

Advertisement

6. Conclusion

Iron is an element that is critical for human life, and due to its ability to exist in different oxidation state, it partakes in oxygen transport, DNA synthesis, energy production, and many more physiological functions. The regulation of iron levels in human is unique compared to other essential dietary nutrients because the body lacks a regulated excretory process for getting rid of excess iron. Therefore, an efficient regulatory mechanism has been developed to maintain body iron content at the level of absorption in the duodenum.

In view of the efficient absorption of haem iron, a precise mechanism of iron absorption is necessary to adequately manage diseases that bother on defects in the iron signaling pathway. Also, given the role of dietary factors in determining iron bioavailability and absorption, it is essential for individuals experiencing iron deficiency and anemia to pay keen attention to their dietary intake to forestall undesirable health implications. In order to achieve this, iron supplements and iron-fortified products can be included in the diet.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Anderson GJ, Frazer DM, McKie AT, Vulpe CD, Smith A. Mechanisms of haem and non-haem iron absorption: Lessons from inherited disorders of iron metabolism. Biometals. 2005;18(4):339-348
  2. 2. Boldt DH. New perspectives on iron: An introduction. The American Journal of the Medical Sciences. 1999;318(4):207-212
  3. 3. Guo Q, Li L, Hou S, Yuan Z, Li C, Zhang W, et al. The role of iron in cancer progression. Frontiers in Oncology. 2021;10(11):778492
  4. 4. Ems T, St. Lucia K, Huecker MR. Biochemistry, Iron Absorption. Treasure Island, FL: StatPearls Publishing; 2023
  5. 5. Carpenter CE, Mahoney AW. Contributions of heme and nonheme iron to human nutrition. Critical Reviews in Food Science and Nutrition. 1992;31(4):333-367
  6. 6. Hallberg L. Bioavailability of dietary iron in man. Annual Review of Nutrition. 1981;1:123-147
  7. 7. Hurrell RF. Preventing iron deficiency through food fortification. Nutrition Reviews. 1997;55(6):210-222
  8. 8. DeLoughery TG. Safety of oral and intravenous iron. Acta haematologica. 2019;142(1):8-12
  9. 9. Shokrgozar N, Golafshan HA. Molecular perspective of iron uptake, related diseases, and treatments. Blood Research. 2019;54(1):10-16
  10. 10. Gulec S, Anderson GJ, Collins JF. Mechanistic and regulatory aspects of intestinal iron absorption. American Journal of Physiology Gastrointestinal and Liver Physiology. 2014;307(4):1522-1547
  11. 11. Kouser F, Noor F. Dietary iron. In: Marwa Z, Tamer H, editors. Iron Metabolism. Rijeka: IntechOpen; 2021 p. Ch. 1
  12. 12. Piskin E, Cianciosi D, Gulec S, Tomas M, Capanoglu EA. Iron absorption: Factors, limitations, and improvement methods. ACS Omega. 2022;7(24):2470-1343
  13. 13. Jacobs A. Iron absorption. Journal of Clinical Pathology Supplement. 1971;5:55-59
  14. 14. Conrad ME. Humoral regulation of iron absorption. Gastroenterology. 1969;57(2):225-228
  15. 15. Sharma KK. Improving bioavailability of iron in Indian diets through food-based approaches for the control of iron deficiency anaemia. Food Nutrition and Agriculture. 2003;32:51-61
  16. 16. Thomas Ems KSL, Huecker MR. Biochemistry, Iron Absorption. Treasure Island (FL): StatPearls Publishing; 2022
  17. 17. Teucher B, Olivares M, Cori H. Enhancers of iron absorption: Ascorbic acid and other organic acids. International Journal for Vitamin and Nutrition Research Internationale Zeitschrift fur Vitamin- und Ernahrungsforschung Journal international de vitaminologie et de nutrition. 2004;74(6):403-419
  18. 18. Smirnoff N. Ascorbic acid metabolism and functions: A comparison of plants and mammals. Free Radical Biology & Medicine. 2018;122:116-129
  19. 19. Cook JD, Reddy MB. Effect of ascorbic acid intake on nonheme-iron absorption from a complete diet. The American Journal of Clinical Nutrition. 2001;73(1):93-98
  20. 20. Davidsson L, Walczyk T, Morris A, Hurrell RF. Influence of ascorbic acid on iron absorption from an iron-fortified, chocolate-flavored milk drink in Jamaican children. The American Journal of Clinical Nutrition. 1998;67(5):873-877
  21. 21. Mao X, Yao G. Effect of vitamin C supplementations on iron deficiency anemia in Chinese children. Biomedical and Environmental Sciences: BES. 1992;5(2):125-129
  22. 22. Cook JD, Monsen ER. Food iron absorption in human subjects. III. Comparison of the effect of animal proteins on nonheme iron absorption. The American Journal of Clinical Nutrition. 1976;29(8):859-867
  23. 23. Björn-Rasmussen E, Hallberg L. Effect of animal proteins on the absorption of food iron in man. Nutrition and Metabolism. 1979;23(3):192-202
  24. 24. Schönfeldt HC, Pretorius B, Hall N. Bioavailability of nutrients. In: Caballero B, Finglas PM, Toldrá F, editors. Encyclopedia of Food and Health. Oxford: Academic Press; 2016. pp. 401-406
  25. 25. Norhaizan ME Jr, Nor Faizadatul Ain AW. Determination of phytate, iron, zinc, calcium contents and their molar ratios in commonly consumed raw and prepared food in Malaysia. Malaysian Journal of Nutrition. 2009;15(2):213-222
  26. 26. Brouns F. Phytic acid and whole grains for health controversy. Nutrients. 2021;14(1)
  27. 27. Wilson MS, Bulley SJ, Pisani F, Irvine RF, Saiardi A. A novel method for the purification of inositol phosphates from biological samples reveals that no phytate is present in human plasma or urine. Open Biology. 2015;5(3):150014
  28. 28. Hart JJ, Tako E, Kochian LV, Glahn RP. Identification of black bean (Phaseolus vulgaris L.) polyphenols that inhibit and promote iron uptake by Caco-2 cells. Journal of Agricultural and Food Chemistry. 2015;63(25):5950-5956
  29. 29. Petry N, Egli I, Zeder C, Walczyk T, Hurrell R. Polyphenols and phytic acid contribute to the low iron bioavailability from common beans in young women. The Journal of Nutrition. 2010;140(11):1977-1982
  30. 30. Hurrell RF, Reddy M, Cook JD. Inhibition of non-haem iron absorption in man by polyphenolic-containing beverages. The British Journal of Nutrition. 1999;81(4):289-295
  31. 31. Lesjak M, Balesaria S, Skinner V, Debnam ES, Srai SKS. Quercetin inhibits intestinal non-haem iron absorption by regulating iron metabolism genes in the tissues. European Journal of Nutrition. 2019;58(2):743-753
  32. 32. Piskin E, Cianciosi D, Gulec S, Tomas M, Capanoglu E. Iron absorption: Factors, limitations, and improvement methods. ACS Omega. 2022;7(24):20441-20456
  33. 33. Hallberg L, Rossander-Hulthén L, Brune M, Gleerup A. Inhibition of haem-iron absorption in man by calcium. The British Journal of Nutrition. 1993;69(2):533-540
  34. 34. Roughead ZK, Zito CA, Hunt JR. Inhibitory effects of dietary calcium on the initial uptake and subsequent retention of heme and nonheme iron in humans: Comparisons using an intestinal lavage method. The American Journal of Clinical Nutrition. 2005;82(3):589-597
  35. 35. Hurrell RF, Reddy MB, Juillerat M, Cook JD. Meat protein fractions enhance nonheme iron absorption in humans. The Journal of Nutrition. 2006;136(11):2808-2812
  36. 36. Ishikawa SI, Tamaki S, Arihara K, Itoh M. Egg yolk protein and egg yolk phosvitin inhibit calcium, magnesium, and iron absorptions in rats. Journal of Food Science. 2007;72(6):S412-S419
  37. 37. Kim M, Lee D, Lee YS. Iron absorption and intestinal solubility in rats are influenced by dietary proteins. Nutrition Research. 1995;15:1705-1716
  38. 38. Heaney RP, Weaver CM. Oxalate: Effect on calcium absorbability. The American Journal of Clinical Nutrition. 1989;50(4):830-832
  39. 39. Heaney RP, Weaver CM, Recker RR. Calcium absorbability from spinach. The American Journal of Clinical Nutrition. 1988;47(4):707-709
  40. 40. Kelsay JL, Prather ES. Mineral balances of human subjects consuming spinach in a low-fiber diet and in a diet containing fruits and vegetables. The American Journal of Clinical Nutrition. 1983;38(1):12-19
  41. 41. Genannt Bonsmann SS, Walczyk T, Renggli S, Hurrell RF. Oxalic acid does not influence nonhaem iron absorption in humans: A comparison of kale and spinach meals. European Journal of Clinical Nutrition. 2008;62(3):336-341
  42. 42. Conrad ME, Cortell S, Williams HL, Foy AL. Polymerization and intraluminal factors in the absorption of hemoglobin-iron. The Journal of Laboratory and Clinical Medicine. 1966;68(4):659-668
  43. 43. Hallberg L, Sölvell L. Absorption of hemoglobin iron in man. Acta medica Scandinavica. 1967;181(3):335-354
  44. 44. Levine DS, Huebers HA, Rubin CE, Finch CA. Blocking action of parenteral desferrioxamine on iron absorption in rodents and men. Gastroenterology. 1988;95(5):1242-1248
  45. 45. Weintraub LR, Conrad ME, Crosby WH. Absorption of hemoglobin iron by the rat. Proceedings of the Society for Experimental Biology and Medicine Society for Experimental Biology and Medicine (New York, NY). 1965;120(3):840-843
  46. 46. Wheby MS, Suttle GE, Ford KT 3rd. Intestinal absorption of hemoglobin iron. Gastroenterology. 1970;58(5):647-654
  47. 47. Yanatori I, Kishi F. DMT1 and iron transport. Free Radical Biology and Medicine. 2019;133:55-63
  48. 48. Hartmann F, Bissell DM. Metabolism of heme and bilirubin in rat and human small intestinal mucosa. The Journal of Clinical Investigation. 1982;70(1):23-29
  49. 49. Raffin SB, Woo CH, Roost KT, Price DC, Schmid R. Intestinal absorption of hemoglobin iron-heme cleavage by mucosal heme oxygenase. The Journal of Clinical Investigation. 1974;54(6):1344-1352
  50. 50. Vogt AC, Arsiwala T, Mohsen M, Vogel M, Manolova V, Bachmann MF. On iron metabolism and its regulation. International Journal of Molecular Sciences. 2021;27;22(9):4591
  51. 51. Finberg KE. Blocking (iron) traffic in the gut. Blood. 2022;139(16):2424-2425
  52. 52. Das NK, Jain C, Sankar A, Schwartz AJ, Santana-Codina N, Solanki S, et al. Modulation of the HIF2α-NCOA4 axis in enterocytes attenuates iron loading in a mouse model of hemochromatosis. Blood, The Journal of the American Society of Hematology. 2022;21;139(16):2547-2552
  53. 53. Santana-Codina N, Mancias JD. The role of NCOA4-mediated ferritinophagy in health and disease. Pharmaceuticals. 2018;23;11(4):114
  54. 54. Anderson GJ, Frazer DM, McKie AT, Vulpe CD. The ceruloplasmin homolog hephaestin and the control of intestinal iron absorption. Blood Cells, Molecules & Diseases. 2002;29(3):367-375
  55. 55. McKie AT, Marciani P, Rolfs A, Brennan K, Wehr K, Barrow D, et al. A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation. Molecular Cell. 2000;5(2):299-309
  56. 56. Caballero Valcárcel AM, López Nicolás R, Frontela Saseta C, González Bermúdez CA, Martínez Graciá C, Santaella PM. Comparison of bioavailability and transporters gene expression of four iron fortificants added to infant cereals. Food Bioscience. 2022;50:102023
  57. 57. Kawabata H. Transferrin and transferrin receptors update. Free Radical Biology and Medicine. 2019;1;133:46-54
  58. 58. Bogdan AR, Miyazawa M, Hashimoto K, Tsuji Y. Regulators of iron homeostasis: new players in metabolism, cell death, and disease. Trends in Biochemical Sciences. 2016;1;41(3):274-286
  59. 59. Bannerman RM. Quantitative aspects of hemoglobin- iron absorption. The Journal of Laboratory and Clinical Medicine. 1965;65:944-950
  60. 60. Roberts SK, Henderson RW, Young GP. Modulation of uptake of heme by rat small intestinal mucosa in iron deficiency. The American Journal of Physiology. 1993;265(4 Pt 1):G712-G718
  61. 61. Turnbull A, Cleton F, Finch CA. Iron absorption. IV. The absorption of hemoglobin iron. The Journal of Clinical Investigation. 1962;41(10):1897-1907
  62. 62. Wyllie JC, Kaufman N. An electron microscopic study of heme uptake by rat duodenum. Laboratory Investigation: A Journal of Technical Methods and Pathology. 1982;47(5):471-476
  63. 63. Ganz T. Hepcidin and its role in regulating systemic iron metabolism. Hematology. American Society of Hematology. Education Program. 2006;29-35:507
  64. 64. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science. 2004;306(5704):2090-2093
  65. 65. Wang CY, Babitt JL. Hepcidin regulation in the anemia of inflammation. Current Opinion in Hematology. 2016;23(3):189-197
  66. 66. Jiang L, Wang J, Wang K, Wang H, Wu Q, Yang C, et al. RNF217 regulates iron homeostasis through its E3 ubiquitin ligase activity by modulating ferroportin degradation. Blood, The Journal of the American Society of Hematology. 2021;26;138(8):689-705
  67. 67. Drakesmith H, Sweetland E, Schimanski L, Edwards J, Cowley D, Ashraf M, et al. The hemochromatosis protein HFE inhibits iron export from macrophages. Proceedings of the National Academy of Sciences. 2002;26;99(24):15602-15607
  68. 68. Andrews NC, Schmidt PJ. Iron homeostasis. Annual Review of Physiology. 2007;17(69):69-85
  69. 69. Torti SV, Manz DH, Paul BT, Blanchette-Farra N, Torti FM. Iron and cancer. Annual Review of Nutrition. 2018;21(38):97-125
  70. 70. Anderson CP, Shen M, Eisenstein RS, Leibold EA. Mammalian iron metabolism and its control by iron regulatory proteins. Biochimica et Biophysica Acta (BBA)-Molecular Cell Research. 2012;1;1823(9):1468-1483
  71. 71. Mole DR. Iron homeostasis and its interaction with prolyl hydroxylases. Antioxidants & Redox Signaling. 2010;15;12(4):445-458
  72. 72. Muthayya S, Thankachan P, Hirve S, Amalrajan V, Thomas T, Lubree H, et al. Iron fortification of whole wheat flour reduces iron deficiency and iron deficiency anemia and increases body iron stores in indian school-aged children, 4. The Journal of Nutrition. 2012;1;142(11):1997-2003
  73. 73. Prentice AM, Mendoza YA, Pereira D, Cerami C, Wegmuller R, Constable A, et al. Dietary strategies for improving iron status: Balancing safety and efficacy. Nutrition Reviews. 2017;1;75(1):49-60
  74. 74. World Health O. Guideline: Daily Iron Supplementation in Infants and Children. Geneva: World Health Organization; 2016
  75. 75. World Health O. Guideline: Daily iron Supplementation in Adult Women and Adolescent Girls. Geneva: World Health Organization; 2016
  76. 76. Niehaus ED, Malhotra R, Cocca-Spofford D, Semigran M, Lewis GD. Repletion of iron stores with the use of oral iron supplementation in patients with systolic heart failure. Journal of Cardiac Failure. 2015;1;21(8):694-697
  77. 77. Mimura EC, Breganó JW, Dichi JB, Gregório EP, Dichi I. Comparison of ferrous sulfate and ferrous glycinate chelate for the treatment of iron deficiency anemia in gastrectomized patients. Nutrition. 2008;1;24(7-8):663-668
  78. 78. Kadota K, Kamata K, Kobayashi Y, Kagaya H, Shimada S, Yoshimoto‐Furuie K, Echizen H. Nomogram for individualizing supplementary iron doses during erythropoietin therapy in haemodialysis patients. Journal of Clinical Pharmacy and Therapeutics. 2002;27(2):111-119
  79. 79. Man Y, Xu T, Adhikari B, Zhou C, Wang Y, Wang B. Iron supplementation and iron-fortified foods: A review. Critical Reviews in Food Science and Nutrition. 2022;1;62(16):4504-4525
  80. 80. Cao GY, Li KX, Jin PF, Yue XY, Yang C, Hu X. Comparative bioavailability of ferrous succinate tablet formulations without correction for baseline circadian changes in iron concentration in healthy Chinese male subjects: A single-dose, randomized, 2-period crossover study. Clinical Therapeutics. 2011;1;33(12):2054-2059
  81. 81. Muñoz M, Gómez-Ramírez S, Besser M, Pavía J, Gomollón F, Liumbruno GM, et al. Current misconceptions in diagnosis and management of iron deficiency. Blood Transfusion. 2017;15(5):422
  82. 82. Dainty JR, Berry R, Lynch SR, Harvey LJ, Fairweather-Tait SJ. Estimation of dietary iron bioavailability from food iron intake and iron status. PLoS One. 2014;30;9(10):e111824
  83. 83. Podder R, Tar’an B, Tyler RT, Henry CJ, DellaValle DM, Vandenberg A. Iron fortification of lentil (Lens culinaris Medik.) to address iron deficiency. Nutrients. 2017;11;9(8):863
  84. 84. Backstrand JR. The history and future of food fortification in the United States: A public health perspective. Nutrition Reviews. 2002;1;60(1):15-26
  85. 85. Hurrell RF. Fortification: Overcoming technical and practical barriers. The Journal of Nutrition. 2002;1;132(4):806S-8012S
  86. 86. Hurrell RFE. Optimizing the Bioavailability of iron Compounds for Food Fortification. Basel, Switzerland: Sight and Life Press; 2007
  87. 87. Hurrell R, Egli I. Iron bioavailability and dietary reference values. The American Journal of Clinical Nutrition. 2010;1;91(5):1461S-1467S
  88. 88. Dobe M, Garg P, Bhalla G. Fortification as an effective strategy to bridge iron gaps during complementary feeding. Clinical Epidemiology and Global Health. 2018;6(4):168-171
  89. 89. Sue Horton VM, Wesley A. Best Practices Paper: Micronutrient Fortification (Iron and Salt Iodization). Frederiksberg, Denmark: Copenhagen Consensus Center; 2008
  90. 90. FAO. Joint Meeting of the Intergovernmental Group on Grains (30th Session) and the Intergovernmental Group on Rice (41st Session). Cereals and Other Starch-Based Staples: Are Consumption Patterns Changing? Rome, Italy: FAO; 2004
  91. 91. World Health O. WHO, FAO, UNICEF, Global Alliance for improved nutrition, micronutrient initiative, flour fortification initiative. Recommendations on wheat and maize flour fortification. Meeting report: interim consensus statement. Geneva. 2009
  92. 92. Blanco-Rojo R, Vaquero MP. Iron bioavailability from food fortification to precision nutrition. A review. Innovative Food Science & Emerging Technologies. 2019;51:126-138
  93. 93. Nair K. Contextualizing the principles of iron fortification of foods in India. NFI Bulletin. 2019;40:1-5

Written By

Aderinola Awoniyi, Oreoluwa Daniel and Oladimeji Babatunde

Submitted: 22 May 2023 Reviewed: 06 February 2024 Published: 05 July 2024