Next Article in Journal
The Bioactive Gamma-Oryzanol from Oryza sativa L. Promotes Neuronal Differentiation in Different In Vitro and In Vivo Models
Previous Article in Journal
Exploring Plasma Coenzyme Q10 Status in Paediatric Dyslipidaemia
Previous Article in Special Issue
Erythrocyte Oxidative Status in People with Obesity: Relation to Tissue Losses, Glucose Levels, and Weight Reduction
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes

1
Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
2
Department of Dermatology and Allergology, School of Medicine and Health, Technical University of Munich, Biedersteinerstr. 29, 80802 München, Germany
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(8), 968; https://doi.org/10.3390/antiox13080968
Submission received: 25 July 2024 / Revised: 6 August 2024 / Accepted: 7 August 2024 / Published: 9 August 2024
(This article belongs to the Special Issue Blood Cells and Redox Homeostasis in Health and Disease)

Abstract

:
In the small intestine, nutrients from ingested food are absorbed and broken down by enterocytes, which constitute over 95% of the intestinal epithelium. Enterocytes demonstrate diet- and segment-dependent metabolic flexibility, enabling them to take up large amounts of glutamine and glucose to meet their energy needs and transfer these nutrients into the bloodstream. During glycolysis, ATP, lactate, and H+ ions are produced within the enterocytes. Based on extensive but incomplete glutamine oxidation large amounts of alanine or lactate are produced. Lactate, in turn, promotes hypoxia-inducible factor-1α (Hif-1α) activation and Hif-1α-dependent transcription of various proton channels and exchangers, which extrude cytoplasmic H+-ions into the intestinal lumen. In parallel, the vitamin C-dependent and duodenal cytochrome b-mediated conversion of ferric iron into ferrous iron progresses. Finally, the generated electrochemical gradient is utilized by the divalent metal transporter 1 for H+-coupled uptake of non-heme Fe2+-ions. Iron efflux from enterocytes, subsequent binding to the plasma protein transferrin, and systemic distribution supply a wide range of cells with iron, including erythroid precursors essential for erythropoiesis. In this review, we discuss the impact of vitamin C on the redox capacity of human erythrocytes and connect enterocyte function with iron metabolism, highlighting its effects on erythropoiesis.

1. Introduction

A considerable proportion of the cell volume of organelle-free mature human erythrocytes (hRBC) consists of hemoglobin, the molecule responsible for respiratory gas exchange of CO2, O2, and CO. Heme is an iron-containing heterocyclic molecule with an iron ion (Fe2+) at its center. Iron has the ability to reversibly switch between its two most common oxidation states: ferrous (Fe2+) and ferric (Fe3+) forms. Both CO and O2 solely bind to Fe2+, whereas NO binds to both Fe2+ and Fe3+. Methemoglobin (Hb-Fe3+), produced during the auto-oxidation of oxyhemoglobin (HbFe2+-O2), has an essential physiological function. hRBCs as well as diverse cell types in our body (e.g., hepatocytes and astrocytes), produce the anti-inflammatory signaling molecule hydrogen sulfide (H2S) [1,2,3]. hRBCs with their methemoglobin, are significantly involved in the degradation of H2S and thus regulate H2S levels in blood and tissues [4]. A remarkable property of hydrophobic and cell membrane permeable H2S is its carrier- and facilitator-independent transmembrane diffusion [5]. hRBCs are versatile, polyfunctional, and highly complex. They interact with endogenous cells (platelets and lymphocytes) as well as with pathogens (bacteria and viruses). Both complement receptor 1 (CR-1) and glycophorin A (GPA) mediate the attachment of hRBCs to bacteria and viruses [6], respectively, leading to phagocytosis and final elimination of RBCs-bound bacteria and viruses in the liver and spleen [7]. hRBCs also promote the proliferation of activated CD8+ T cells [8] and actively absorb infectious HIV-1 virions [9]. Thus, RBCs considerably relieve our immune system. hRBCs can rapidly and reversibly bind an array of chemokines, including IL-8, a leukocyte chemotaxin [10,11,12]. As a result, excessive stimulation of leukocytes and uncontrolled inflammatory responses are avoided. Hemoglobin α and β chains bind LPS (lipopolysaccharide, an endotoxin of Gram-negative bacteria) and neutralize its activity [13,14]. Functioning hRBCs and their production (erythropoiesis), however, require a functioning gastrointestinal tract that makes the components of the ingested food, such as ions and vitamins, available systemically. In this review, we connect vitamin C-dependent uptake of non-heme iron in enterocytes, highlighting their systemic effects on erythropoiesis (Figure 1).

2. pH-Dependent Solubility and Uptake of Vitamin C and Dietary Non-Heme Iron

The proper uptake, storage, systemic distribution, and utilization of iron are prerequisites for general health and well-being. For hemoproteins formation (e.g., hemoglobin [16] and ascorbic acid-dependent transmembrane ferrireductases of the cytochrome b561 class [17]), various body tissues store iron in the cytosolic protein complex ferritin. Macrophages of the spleen, liver, bone marrow, and skeletal muscle are further prominent storage sites for iron. Regarding one electron transport capacity, iron shows mixed valence states, designated as ferrous (Fe2+) and ferric (Fe3+) forms. Freely available ferrous iron becomes cytotoxic in the presence of the respiratory by-product hydrogen peroxide (H2O2) through highly reactive hydroxyl radical formation via Fenton’s reaction [18]. The environment of the digestive tract, especially of the duodenum is complex and adaptable. Following ingestion and upon entry of food into the duodenum, a dual and coincident stimulation of the external secretory functions of the liver and pancreas occurs, i.e., the flow of bile and pancreatic juice into the duodenum. Enterocytes covering the lumen of the intestinal mucosa can absorb ions, nutrients, vitamins, hormones, and water and transfer them to the blood. These cells non-competitively absorb both forms of dietary iron: heme and non-heme iron. Heme, released by hydrolysis of hemoproteins through intraluminal proteases and maintained in a soluble form by globin breakdown products, is absorbed intact by the enterocytes [19,20]. Afterwards, microsomal heme oxygenase-catalyzed heme degradation releases inorganic iron [21]. The latter is then either stored in ferritin molecules or transported to the basolateral membrane of the enterocytes for subsequent release into the blood.
The following equation shows the mucosal heme-splitting activity:
Heme + 3 O2 + 31/2 NADPH + 31/2 H+ + 7 e → biliverdin + CO + 31/2 NADP+ + 3H2O + Fe2+.
However, iron intake does not directly reflect iron bioavailability. Uptake by enterocytes of dietary non-heme iron, with ferric iron (Fe3+) being the most prevalent, is vitamin C-dependent. Ascorbic acid (AA) pH-dependently exerts both a reducing and a chelating effect on iron salts [22]. Both AA and ferric chloride are totally soluble in the acidic milieu of the stomach. This acidic pH causes the displacement of hydrogen ions from AA to ferric iron, leading to AA-iron chelate formation, which remains in solution over a pH range of 2 to 11. This iron chelate can thus mainly be absorbed at the slightly acidic pH of the duodenum [22,23]. In contrast to iron in heme complex, the uptake of non-heme iron is strongly regulated by dietary constituents.

3. Roles of Copper Ion (Cu+), Regulatory Proteins, and Vitamin C in Non-Heme Iron Absorption across Human Enterocytes

Two carrier systems accomplish heme-bound iron absorption: (1) heme-carrier protein 1 (HCP-1), a primarily H+-coupled folate transporter, and (2) receptor-mediated endocytosis. The apical influx of non-heme iron, especially ferrous iron (Fe2+) into the human enterocytes, its basolateral efflux, and re-oxidation to ferric iron (Fe3+), engages several regulatory and transporter proteins including their coordinated interactions. A sequence of steps is required prior to loading of monomeric plasma protein transferrin with two ferric iron ions. Both transmembrane proteins duodenal cytochrome B (Dcytb) and divalent metal transporter 1A-I (DMT1), with the former being ascorbic acid (AA)-reducible [17,24,25,26], are highly abundant in the brush-border membrane of duodenal enterocytes. The mammalian di-heme-containing [25,27] and iron-regulated ferric reductase Dcytb reduces ferric iron (Fe3+) to ferrous iron (Fe2+) prior to its transport by DMT1 [28]. The secondary active transporter DMT1, displays a pH dependence and, in an acidic environment operates as an H+/Fe2+ cotransporter [29,30,31]. This leads, in the case of enterocytes located at the proximal duodenum (pH 6.0) to rapid intracellular acidification (Figure 2).

4. Intracellular Enterocytes’ Lactate Production by Glutamine and Glucose Metabolism

Enterocytes absorb large amounts of glutamine, glucose, and ketone bodies to cover their energy needs. However, they show substrate preference for oxidative metabolism which can be altered by the availability of other substrates. Both villus and crypt cells have mitochondrial glutaminase activity. Glutamate generated by glutaminase can be transaminated to produce alanine, aspartate, and α-ketoglutarate. The latter an intermediate in the Krebs cycle, contributes to ATP production. The intestinal mucosa consisting of 75% non-lymphoid and 25% lymphoid tissues by mass [32], plays an important role in mucosal immunity [33,34,35,36]. Both cell types—enterocytes and intraepithelial immune cells—residing in this area, utilize glutamine at high and comparable intensity [32,37]. Based on extensive but incomplete glutamine oxidation in the intestinal mucosa, large amounts of glutamine undergo two steps of decarboxylation with the final product being either alanine or lactate depending on the pyruvate pool [38,39]. It is important to mention the gastrointestinal pH profile of healthy subjects. The stomach has a pH of 1.3 to 2. In the small intestine, consisting of three successive sections (duodenum, jejunum, and ileum), the pH values increase. These are about 6.0 in the proximal part of the duodenum, 7.0 at the duodenojejunal junction, and 7.4 in the terminal ileum [40,41]. Figure 3A shows the inverse correlation between DMT1 abundance and pH along the small intestine. Figure 3B illustrates the uptake of glucose in human enterocytes and its utilization by the glycolysis pathway which culminates in the production of two ATP, lactate and H+-ions each. Glycolysis is more intense in enterocytes of the proximal than in the distal intestine. Thus, glutamine and glucose metabolism are mainly involved in intracellular enterocyte lactate production.

5. Lactate-Induced Activation of Hypoxia-Inducible Factor 1-Alpha (Hif-1α) in Enterocytes and Other Cell Types

Glycolysis, which is independent of intact mitochondrial function, represents a positive selective pressure of evolution for obtaining energy (ATP) within the shortest time and is not restricted to enterocytes. Several healthy cells, e.g., human cytotoxic CD8+ T lymphocytes [42] and murine embryonic stem cells [43] temporarily exhibit a significant increase in glycolysis rate during activation and proliferation. This also applies to growing and proliferating tumor cells. However, for the continuation of glycolysis, the extrusion of lactate and H+-ions into the extracellular environment is mandatory [44]. This is performed by H+-linked monocarboxylate transporters (MCTs) [45]. The generated lactate also activates and stabilizes heme-containing hypoxia-inducible factor 1-α (Hif-1α) [46,47,48], a master regulator of glycolysis and oxygen homeostasis [49]. Hif-1α, discovered by Goldberg et al. [50], in turn, activates the transcription of numerous genes encoding MCT1, MCT4, Na+/H+-antiporter 1 (NHE-1) [51], vacuolar-type proton pump ATPase (V-ATPase), inducible nitric oxide synthase (i-NOS), heme oxygenase 1 (HO-1), transferrin (Tf), erythropoietin (EPO), ecto-enzyme carbonic anhydrase IX and glucose transporters 1 and 3 (GLUT-1 & -3), of which some are important contributors to erythropoiesis. For lactate-dependent and Hif-1α-mediated control of pH regulating pathways see the following review: [52].

6. Hif-1α Mediated Control of pH Regulating Pathways and Their Interplay with Divalent Metal Transporter 1 (DMT1) for Non-Heme Iron Transport

Intracellular pH [pH]i homeostasis is vital to the functioning of cells. Several ion transport mechanisms are involved in this process, e.g., exchangers (NHEs), proton (H+) pumps (V-ATPases), and H+-MCTs co-transporters, resulting in an alkaline shift in [pH]i. As described above in Section 3, DMT1 displays pH dependence and, in an acidic environment (for instance, in the duodenum), operates as an H+/Fe2+ cotransporter, leading to rapid intracellular acidification of enterocytes. To counteract this alteration of [pH]i, reciprocal NHE-3/Na+-K+-ATPase interplay (concerning Na+-ions translocation) as well as the contribution of V-ATPase are needed to apically efflux cytoplasmic H+-ions into the intestine lumen. This leads to the generation of an acidic microclimate at the brush border membrane of duodenal enterocytes and the formation of an H+-electrochemical gradient. The latter is subsequently used by enterocytes for a DMT1-dependent and H+-coupled uptake of Fe2+-ions. The most important physiological role of Na+-K+-ATPase is to channel the free energy of ATP-hydrolysis to intracellularly keep K+-ions at high and Na+-ions at low concentrations. On the basolateral site of the enterocyte membrane, MCT-1 regulates the equimolar and electroneutral co-extrusion of lactate and H+-ions into the circulation. For more details see Figure 2.

7. Iron Flux across Enterocytes Membrane, Its Release into the Blood and Distribution by Plasma Transferrin

For iron flux across enterocytes, they require not only apically located influx carriers Dcytb and DMT1 but also basolaterally located transmembrane efflux proteins ferroportin-1 (FPN1) [53] and Cu1+-dependent ferroxidase hephaestin [54,55,56,57,58,59,60,61,62]. Fe2+ exported by FPN1 is rapidly oxidized back to Fe3+ by hephaestin. Subsequently, Fe3+-binding plasma protein transferrin (Tf), responsible for systemic iron circulation—Tf-(Fe3+)2—supplies a wide range of cells with iron, including erythroid precursors essential for erythropoiesis [63,64] and Figure 2.

8. Roles of Folates, Vitamin B12, Ferrous Iron (Fe2+), Erythropoietin, Testosterone and Hepcidin in Erythropoiesis

The progressive differentiation of short-term hematopoietic stem cells in the bone marrow leads, among other things, to the formation of an erythroid lineage from which terminally differentiated hemoglobin-containing hRBCs arise. This dynamic production process of erythrocytes, referred to as erythropoiesis [65,66,67,68], requires an adequate supply of folates, vitamin B12 (cobalamin), and ferrous iron (Fe2+). Deficiency in one or more of these substances results in nutrition-related anemia. Folates are primarily absorbed in the duodenum and jejunum [69,70], whereas intrinsic factor-bound vitamin B12 (vit B12) is mainly absorbed in the terminal ileum [71,72,73]. Stomach acid (which is decreased in subjects with atrophic gastritis), its digestive enzymes (e.g., pepsin), and vit B12-binding glycoproteins haptocorrin and intrinsic factor positively regulate vit B12 absorption [71]. In adults, the kidney serves as the major site (peritubular fibroblasts in the renal cortex) [74,75,76,77], and the liver to a much lesser extent (hepatocytes and perisinusoidal Ito cells) [78,79,80] produces the circulating plasma protein hormone erythropoietin (EPO), the principal regulator of erythropoiesis. For reviews see [81,82,83]. Testosterone suppresses hepcidin [84,85]. The treatment of hypogonadal and especially middle-aged and elderly men with testosterone increases hematocrit levels [86]. This might explain gender-based differences in hematocrit content. The hepatic peptide hormone hepcidin, first discovered by Park et al. 2001 and Pigeon et al. 2001 [87,88], is directly involved in the maintenance of iron homeostasis, and its regulation is tightly controlled at the transcriptional level [89]. Hepcidin synthesis and release from the liver are positively correlated with inflammation and increased plasma and tissue iron levels. Under iron overload, hepcidin binds to its receptor FPN-1, leading to FPN-1 internalization [90,91], ubiquitination, and subsequent lysosomal degradation [91]. Thus, hepcidin-mediated FPN1-downregulation leads to diminished iron efflux, resulting in intracellular iron retention in iron-releasing target cells, e.g., hepatocytes, tissue macrophages, duodenal enterocytes, and placental cells; see also Figure 2. If persistent, this condition impairs iron-dependent erythropoiesis, as systemic iron levels decrease. The following is also of physiological importance: anemia and hypoxia significantly inhibit hepatocellular hepcidin gene expression [92]. Thus, the sophisticated interplay between plasma and tissue iron, testosterone, EPO, hepcidin, anemia, and hypoxia might be understood as a homeostatic loop to maintain the dynamic balance between iron deficiency and overload.

9. Glutathione and NADH-Dependent Vitamin C Reduction, Essential Contributors to Maintaining the Redox Capacity of hRBCs

In mammals, the plasma concentration of the antioxidant glutathione (GSH), a tripeptide with the structure γ-L-glutamyl-L-cysteinylglycine, is about 25 µM, with typical intracellular concentrations between 1 and 5 mM. The low micromolar plasma concentration of ascorbic acid (AA) is slightly higher than that of GSH, i.e., 40–60 µM. Intraerythrocytic concentrations of GSH and AA are considerably high and amount to 1–2 mM each. AA and DHA uptake are carrier-mediated: the former Na+-dependent and the latter Na+-independent, carried out by sodium-dependent vitamin C transporters (SVCTs) [93,94,95,96] and members of the glucose-transporter family (GLUT-1, -3 and -4) [94,97,98,99,100], respectively. The driving force for such substrate movements is the protein carrier-mediated secondary active transport with net accumulation of substrate on the other side of the membrane (here: outside → inside direction). Primarily, the mammalian Na+/K+-ATPase, discovered in 1965 [101], utilizes the glycolytically produced ATP to generate Na+/K+ asymmetry between cells and their surroundings, i.e., low Na+/high K+ content within the cell. This gradient is then used to drive diverse secondary active transports [102]. In contrast to nucleated cells, AA is a poor substrate for hRBCs. GLUT-1 transports DHA into hRBCs [103,104,105]. Once within the cells, GSH-dependent two-electron regeneration of AA occurs (DHA + 2 GSH → AA + GSSG), i.e., without involving the monoascorbyl free radical (AFR) intermediate [106,107,108]. This GSH-dependent reduction in DHA is not solely restricted to hRBCs [109]. GSH and DHA are interconnected and form a functional unit. The rapid entry of DHA inflicts on cells a high need for GSH for its reduction to AA. In this context, DHA stimulates the NADPH-generating pentose phosphate pathway (PPP) [110,111]. Subsequently, glutathione reductase (GR) catalyzes NADPH-dependent glutathione disulfide reduction (GSSG + NADPH + H+ → 2 GSH + NADP+). This self-supporting machinery, as a positive feedback loop, ensures permanent AA regeneration and accumulation within the hRBCs. The subsequent intracellular consumption and extracellular transport of AA (inside → outside direction) and the re-entry of its two-electron oxidized form DHA back into the cells (outside → inside direction) lead to the maintenance of a large intracellular electron pool. This culminates in the vitamin C-dependent high redox capacity of hRBCs; see also Figure 4. NADH generated during glycolysis represents another endogenous source for DHA reduction [112]. The groundbreaking discovery of this group was that a mixture of each mole of AA with 2 moles of ferricyanide instantly resulted in the generation of one mole DHA and two moles of forrocyanide, i.e., without involving the AFR intermediate (AA + 2 ferricyanide → DHA + 2 ferrocyanide) (see Figure 4).

10. Impact of V-ATPase on Endosomal pH and DMT1B-II Mediated Iron (Fe2+) Release into the Cytosol, and Its Relevance for Erythropoiesis

In previous sections, we summarized the apical uptake and basolateral efflux of iron across the cell membrane of enterocytes. Now, we will address its subsequent distribution into the blood and utilization in different target cells using the example of erythroid precursors. With almost 30 trillion eukaryotic cells in our body, RBCs, with ~25 trillion, represent nearly 84% of the total cells [115]. Circulating hRBCs have a lifespan of approximately 120 days [116]. Daily, about 1% (~250 billion) of senescent hRBCs are engulfed and degraded by macrophages and replaced to the same extent through erythropoiesis, i.e., two million RBCs are produced per second. A single intact hRBC contains over 270 million hemoglobin molecules. Hemoglobin (Hb) is an iron-porphyrin protein complex consisting of four polypeptide chains, each having a heme prosthetic group with a ferrous iron (Fe2+) at its center. Thus, a single hRBC possesses 1.1 billion heme groups or 1.1 billion Fe2+ ions. In other words, humans acquire the major part of body iron by catabolizing Hb obtained from senescent RBCs. Dietary iron absorbed in the small intestine and excess iron stored within ferritin in the liver hepatocytes are also available to most body cells. The diferric transferrin—Tf-(Fe3+)2—is the key iron transport machinery for heme biosynthesis in erythroid precursors. It binds to the transferrin receptor (Tf-R), which clusters in specialized areas of the cell surface, called ‘coated pits’. Coated pits, whose assembly is potassium dependent [117], are pooled into the cytosol to rapidly form coated endocytic vesicles. The latter lose the majority of their coat proteins and are then referred to as primary endosomes (Figure 5). Membrane-embedded V-ATPases are ATP-dependent proton (H+)-pumps. They are present in both endomembrane organelles and cell membrane and lead to alkalinization of the cytoplasm associated with acidification of both extracellular milieu (Figure 2) and intracellular compartments, e.g., endosomal and lysosomal lumen [118,119,120] (for details, see Figure 5). In the acidified endosome, ferric iron (Fe3+) readily dissociates from Tf [121,122] and is subsequently reduced to ferrous iron (Fe2+) by NADPH-dependent endosomal ferrireductase Steap3 (six-transmembrane epithelial antigen of the prostate) [123], prior to its transport into the cytosol by H+-coupled endosomal DMT1B-II [124] (see also Figure 5). Cytosolic iron (Fe2+) is the essential substrate for heme biosynthesis in erythroid precursors and erythropoiesis. Heme itself controls the synthesis of globin chains—needed for hemoglobin synthesis—at both transcriptional and translational levels. It is important to note that genetic ablation of Steap3 leads to severe hypochromic [125] and microcytic anemia [126].

11. Disruption of Lysosomal pH by Tricyclic Antidepressant Desipramine, Its Possible Negative Effect on Endosomal pH, Iron Supply and Erythropoiesis: A High-Risk Drug during Pregnancy?

Tricyclic antidepressants (TCAs) influence norepinephrine (NE) and serotonin (SER) transporters [127]. Desipramine, a representative of TCAs, has two primary targets. On one hand, it preferentially interacts with the NE-transporter and increases NE synaptic transmission by inhibiting NE reuptake, thereby relieving depressive symptoms [128,129,130,131]. On the other hand, desipramine has a direct inhibitory effect on lysosomal acid ceramidase [132] and acid sphingomyelinase [133,134]. Both acid ceramidase [135,136,137,138] and acid sphingomyelinase [139,140,141] are aberrantly over-expressed and highly active in patients with dysregulated sphingolipid metabolism. Under acidic conditions, e.g., in lysosomes, endosomes, or in the cytoplasm of glycolytically active cells, TCAs act as cationic amphiphilic drugs (CADs) (TCAs + H+ → CADs), their reactions resembling the formation of ammonium from ammonia and a proton (NH3 + H+ → NH4+). The secondary amine and basic lipophilic drug desipramine follow the same principle: it acts as a proton (H+-ion) acceptor, depleting the free proton and thus increasing the intracellular pH. Elojeimy et al. (2006) showed that in cancer cell lines, desipramine, even at a relatively low dose of 5 µM, neutralizes lysosomal pH [132]. By the same principle, desipramine and other CADs like imipramine, amitriptyline, chlorpromazine, and chloroquine would be able to neutralize the luminal acidification of endosomes in glycolytically active erythroid precursors. Consequently, H+-coupled Fe2+ transport into the cytosol of these cells, and thus the proper heme biosynthesis and heme-dependent erythropoiesis, might be severely affected (see Figure 5).
In addition to this, desipramine inhibits NHE-1 activity [142], a major regulator of intracellular pH. As shown in Figure 2, NHE-3 plays an essential role in H+-coupled Fe2+ transport by DMT1 in enterocytes. To avoid anemia and preserve the naturally increased erythropoietic activity during pregnancy, pregnant women suffering from depression should avoid medications with CAD properties if possible. However, this does not diminish the importance of CAD’s clinical applicability regarding cancer [143,144,145] and anti-viral [146,147] therapies. It is to be noted that the above-mentioned low-dose concentration of desipramine (5 µM) has no significant detectable biological effects on cell organelle-free mature hRBCs [148].

Author Contributions

M.G. designed this project. M.G. mainly and X.P. partly wrote the manuscript. X.P. and M.G. searched the literature and selected the references. M.G. made Figure 1 and Figure 2. M.K. and M.G. made Figure 3. X.P. and M.G. made Figure 4 and Figure 5. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge support from the Open Access Publication Fund of the University of Tübingen.

Conflicts of Interest

The authors declare that no competing financial interests or otherwise exist.

References

  1. Stipanuk, M.H.; Beck, P.W. Characterization of the enzymic capacity for cysteine desulphhydration in liver and kidney of the rat. Biochem. J. 1982, 206, 267–277. [Google Scholar] [CrossRef] [PubMed]
  2. Shibuya, N.; Mikami, Y.; Kimura, Y.; Nagahara, N.; Kimura, H. Vascular endothelium expresses 3-mercaptopyruvate sulfurtransferase and produces hydrogen sulfide. J. Biochem. 2009, 146, 623–626. [Google Scholar] [CrossRef] [PubMed]
  3. Tanizawa, K. Production of H2S by 3-mercaptopyruvate sulphurtransferase. J. Biochem. 2011, 149, 357–359. [Google Scholar] [CrossRef] [PubMed]
  4. Vitvitsky, V.; Yadav, P.K.; Kurthen, A.; Banerjee, R. Sulfide oxidation by a noncanonical pathway in red blood cells generates thiosulfate and polysulfides. J. Biol. Chem. 2015, 290, 8310–8320. [Google Scholar] [CrossRef] [PubMed]
  5. Mathai, J.C.; Missner, A.; Kugler, P.; Saparov, S.M.; Zeidel, M.L.; Lee, J.K.; Pohl, P. No facilitator required for membrane transport of hydrogen sulfide. Proc. Natl. Acad. Sci. USA 2009, 106, 16633–16638. [Google Scholar] [CrossRef] [PubMed]
  6. Allaway, G.P.; Burness, A.T. Site of attachment of encephalomyocarditis virus on human erythrocytes. J. Virol. 1986, 59, 768–770. [Google Scholar] [CrossRef] [PubMed]
  7. Craig, M.L.; Bankovich, A.J.; Taylor, R.P. Visualization of the transfer reaction: Tracking immune complexes from erythrocyte complement receptor 1 to macrophages. Clin. Immunol. 2002, 105, 36–47. [Google Scholar] [CrossRef] [PubMed]
  8. Fonseca, A.M.; Pereira, C.F.; Porto, G.; Arosa, F.A. Red blood cells promote survival and cell cycle progression of human peripheral blood T cells independently of CD58/LFA-3 and heme compounds. Cell Immunol. 2003, 224, 17–28. [Google Scholar] [CrossRef] [PubMed]
  9. Beck, Z.; Brown, B.K.; Wieczorek, L.; Peachman, K.K.; Matyas, G.R.; Polonis, V.R.; Rao, M.; Alving, C.R. Human erythrocytes selectively bind and enrich infectious HIV-1 virions. PLoS ONE 2009, 4, e8297. [Google Scholar] [CrossRef]
  10. Darbonne, W.C.; Rice, G.C.; Mohler, M.A.; Apple, T.; Hebert, C.A.; Valente, A.J.; Baker, J.B. Red blood cells are a sink for interleukin 8, a leukocyte chemotaxin. J. Clin. Investig. 1991, 88, 1362–1369. [Google Scholar] [CrossRef]
  11. Horuk, R.; Colby, T.J.; Darbonne, W.C.; Schall, T.J.; Neote, K. The human erythrocyte inflammatory peptide (chemokine) receptor. Biochemical characterization, solubilization, and development of a binding assay for the soluble receptor. Biochemistry 1993, 32, 5733–5738. [Google Scholar] [CrossRef]
  12. de Winter, R.J.; Manten, A.; de Jong, Y.P.; Adams, R.; van Deventer, S.J.; Lie, K.I. Interleukin 8 released after acute myocardial infarction is mainly bound to erythrocytes. Heart 1997, 78, 598–602. [Google Scholar] [CrossRef] [PubMed]
  13. Bahl, N.; Du, R.; Winarsih, I.; Ho, B.; Tucker-Kellogg, L.; Tidor, B.; Ding, J.L. Delineation of lipopolysaccharide (LPS)-binding sites on hemoglobin: From in silico predictions to biophysical characterization. J. Biol. Chem. 2011, 286, 37793–37803. [Google Scholar] [CrossRef] [PubMed]
  14. Liepke, C.; Baxmann, S.; Heine, C.; Breithaupt, N.; Standker, L.; Forssmann, W.G. Human hemoglobin-derived peptides exhibit antimicrobial activity: A class of host defense peptides. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2003, 791, 345–356. [Google Scholar] [CrossRef]
  15. Ghashghaeinia, M.; Mrowietz, U. Human erythrocytes, nuclear factor kappaB (NFkappaB) and hydrogen sulfide (H(2)S)—From non-genomic to genomic research. Cell Cycle 2021, 20, 2091–2101. [Google Scholar] [CrossRef]
  16. Perutz, M.F. Submicroscopic structure of the red cell. Nature 1948, 161, 204. [Google Scholar] [CrossRef] [PubMed]
  17. Su, D.; Asard, H. Three mammalian cytochromes b561 are ascorbate-dependent ferrireductases. FEBS J. 2006, 273, 3722–3734. [Google Scholar] [CrossRef]
  18. Lloyd, R.V.; Hanna, P.M.; Mason, R.P. The origin of the hydroxyl radical oxygen in the Fenton reaction. Free Radic. Biol. Med. 1997, 22, 885–888. [Google Scholar] [CrossRef]
  19. Turnbull, A.; Cleton, F.; Finch, C.A. Iron absorption. IV. The absorption of hemoglobin iron. J. Clin. Investig. 1962, 41, 1897–1907. [Google Scholar] [CrossRef]
  20. Young, G.P.; Rose, I.S.; St John, D.J. Haem in the gut. I. Fate of haemoproteins and the absorption of haem. J. Gastroenterol. Hepatol. 1989, 4, 537–545. [Google Scholar] [CrossRef]
  21. Raffin, S.B.; Woo, C.H.; Roost, K.T.; Price, D.C.; Schmid, R. Intestinal absorption of hemoglobin iron-heme cleavage by mucosal heme oxygenase. J. Clin. Investig. 1974, 54, 1344–1352. [Google Scholar] [CrossRef]
  22. Conrad, M.E.; Schade, S.G. Ascorbic acid chelates in iron absorption: A role for hydrochloric acid and bile. Gastroenterology 1968, 55, 35–45. [Google Scholar] [CrossRef] [PubMed]
  23. Lynch, S.R.; Cook, J.D. Interaction of vitamin C and iron. Ann. N. Y. Acad. Sci. 1980, 355, 32–44. [Google Scholar] [CrossRef] [PubMed]
  24. May, J.M.; Qu, Z.C.; Whitesell, R.R. Ascorbate is the major electron donor for a transmembrane oxidoreductase of human erythrocytes. Biochim. Biophys. Acta 1995, 1238, 127–136. [Google Scholar] [CrossRef] [PubMed]
  25. Oakhill, J.S.; Marritt, S.J.; Gareta, E.G.; Cammack, R.; McKie, A.T. Functional characterization of human duodenal cytochrome b (Cybrd1): Redox properties in relation to iron and ascorbate metabolism. Biochim. Biophys. Acta 2008, 1777, 260–268. [Google Scholar] [CrossRef] [PubMed]
  26. Lane, D.J.; Bae, D.H.; Merlot, A.M.; Sahni, S.; Richardson, D.R. Duodenal cytochrome b (DCYTB) in iron metabolism: An update on function and regulation. Nutrients 2015, 7, 2274–2296. [Google Scholar] [CrossRef] [PubMed]
  27. Ludwiczek, S.; Rosell, F.I.; Ludwiczek, M.L.; Mauk, A.G. Recombinant expression and initial characterization of the putative human enteric ferric reductase Dcytb. Biochemistry 2008, 47, 753–761. [Google Scholar] [CrossRef] [PubMed]
  28. McKie, A.T.; Barrow, D.; Latunde-Dada, G.O.; 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, 1755–1759. [Google Scholar] [CrossRef]
  29. Gunshin, H.; Mackenzie, B.; Berger, U.V.; Gunshin, Y.; Romero, M.F.; Boron, W.F.; Nussberger, S.; Gollan, J.L.; Hediger, M.A. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature 1997, 388, 482–488. [Google Scholar] [CrossRef]
  30. Mackenzie, B.; Ujwal, M.L.; Chang, M.H.; Romero, M.F.; Hediger, M.A. Divalent metal-ion transporter DMT1 mediates both H+ -coupled Fe2+ transport and uncoupled fluxes. Pflugers Arch. 2006, 451, 544–558. [Google Scholar] [CrossRef]
  31. Ehrnstorfer, I.A.; Manatschal, C.; Arnold, F.M.; Laederach, J.; Dutzler, R. Structural and mechanistic basis of proton-coupled metal ion transport in the SLC11/NRAMP family. Nat. Commun. 2017, 8, 14033. [Google Scholar] [CrossRef] [PubMed]
  32. Newsholme, E.A.; Carrie, A.L. Quantitative aspects of glucose and glutamine metabolism by intestinal cells. Gut 1994, 35, S13–S17. [Google Scholar] [CrossRef] [PubMed]
  33. Trejdosiewicz, L.K. What is the role of human intestinal intraepithelial lymphocytes? Clin. Exp. Immunol. 1993, 94, 395–397. [Google Scholar] [CrossRef] [PubMed]
  34. Dahan, S.; Roth-Walter, F.; Arnaboldi, P.; Agarwal, S.; Mayer, L. Epithelia: Lymphocyte interactions in the gut. Immunol. Rev. 2007, 215, 243–253. [Google Scholar] [CrossRef] [PubMed]
  35. Lutter, L.; Hoytema van Konijnenburg, D.P.; Brand, E.C.; Oldenburg, B.; van Wijk, F. The elusive case of human intraepithelial T cells in gut homeostasis and inflammation. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 637–649. [Google Scholar] [CrossRef] [PubMed]
  36. Ma, H.; Tao, W.; Zhu, S. T lymphocytes in the intestinal mucosa: Defense and tolerance. Cell Mol. Immunol. 2019, 16, 216–224. [Google Scholar] [CrossRef] [PubMed]
  37. Newsholme, E.A.; Crabtree, B.; Ardawi, M.S.M. Glutamine metabolism in lymphocytes: Its biochemical, physiological and clinical importance. Q. J. Exp. Physiol. Transl. Integr. 1985, 70, 473–489. [Google Scholar] [CrossRef] [PubMed]
  38. Duée, P.-H.; Darcy-Vrillon, B.; Blachier, F.; Morel, M.-T. Fuel selection in intestinal cells. Proc. Nutr. Soc. 1995, 54, 83–94. [Google Scholar] [CrossRef]
  39. Kight, C.E.; Fleming, S.E. Transamination processes promote incomplete glutamine oxidation in small intestine epithelial cells. J. Nutr. Biochem. 1995, 6, 27–37. [Google Scholar] [CrossRef]
  40. Fallingborg, J. Intraluminal pH of the human gastrointestinal tract. Dan. Med. Bull. 1999, 46, 183–196. [Google Scholar]
  41. Fallingborg, J.; Christensen, L.A.; Ingeman-Nielsen, M.; Jacobsen, B.A.; Abildgaard, K.; Rasmussen, H.H. pH-profile and regional transit times of the normal gut measured by a radiotelemetry device. Aliment. Pharmacol. Ther. 1989, 3, 605–613. [Google Scholar] [CrossRef] [PubMed]
  42. Cham, C.M.; Gajewski, T.F. Glucose availability regulates IFN-gamma production and p70S6 kinase activation in CD8+ effector T cells. J. Immunol. 2005, 174, 4670–4677. [Google Scholar] [CrossRef] [PubMed]
  43. Kondoh, H.; Lleonart, M.E.; Nakashima, Y.; Yokode, M.; Tanaka, M.; Bernard, D.; Gil, J.; Beach, D. A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxid. Redox Signal. 2007, 9, 293–299. [Google Scholar] [CrossRef]
  44. Spencer, T.L.; Lehninger, A.L. L-lactate transport in Ehrlich ascites-tumour cells. Biochem. J. 1976, 154, 405–414. [Google Scholar] [CrossRef] [PubMed]
  45. Broer, S.; Rahman, B.; Pellegri, G.; Pellerin, L.; Martin, J.L.; Verleysdonk, S.; Hamprecht, B.; Magistretti, P.J. Comparison of lactate transport in astroglial cells and monocarboxylate transporter 1 (MCT 1) expressing Xenopus laevis oocytes. Expression of two different monocarboxylate transporters in astroglial cells and neurons. J. Biol. Chem. 1997, 272, 30096–30102. [Google Scholar] [CrossRef] [PubMed]
  46. Kozlov, A.M.; Lone, A.; Betts, D.H.; Cumming, R.C. Lactate preconditioning promotes a HIF-1alpha-mediated metabolic shift from OXPHOS to glycolysis in normal human diploid fibroblasts. Sci. Rep. 2020, 10, 8388. [Google Scholar] [CrossRef] [PubMed]
  47. Sonveaux, P.; Copetti, T.; De Saedeleer, C.J.; Vegran, F.; Verrax, J.; Kennedy, K.M.; Moon, E.J.; Dhup, S.; Danhier, P.; Frerart, F.; et al. Targeting the lactate transporter MCT1 in endothelial cells inhibits lactate-induced HIF-1 activation and tumor angiogenesis. PLoS ONE 2012, 7, e33418. [Google Scholar] [CrossRef] [PubMed]
  48. Wu, Y.; Wang, M.; Feng, H.; Peng, Y.; Sun, J.; Qu, X.; Li, C. Lactate induces osteoblast differentiation by stabilization of HIF1alpha. Mol. Cell Endocrinol. 2017, 452, 84–92. [Google Scholar] [CrossRef] [PubMed]
  49. Semenza, G.L. Hypoxia-inducible factor 1: Master regulator of O2 homeostasis. Curr. Opin. Genet. Dev. 1998, 8, 588–594. [Google Scholar] [CrossRef]
  50. Goldberg, M.A.; Dunning, S.P.; Bunn, H.F. Regulation of the erythropoietin gene: Evidence that the oxygen sensor is a heme protein. Science 1988, 242, 1412–1415. [Google Scholar] [CrossRef]
  51. Shimoda, L.A.; Fallon, M.; Pisarcik, S.; Wang, J.; Semenza, G.L. HIF-1 regulates hypoxic induction of NHE1 expression and alkalinization of intracellular pH in pulmonary arterial myocytes. Am. J. Physiol. Lung Cell Mol. Physiol. 2006, 291, L941–L949. [Google Scholar] [CrossRef] [PubMed]
  52. Ghashghaeinia, M.; Koberle, M.; Mrowietz, U.; Bernhardt, I. Proliferating tumor cells mimick glucose metabolism of mature human erythrocytes. Cell Cycle 2019, 18, 1316–1334. [Google Scholar] [CrossRef] [PubMed]
  53. Donovan, A.; Brownlie, A.; Zhou, Y.; Shepard, J.; Pratt, S.J.; Moynihan, J.; Paw, B.H.; Drejer, A.; Barut, B.; Zapata, A.; et al. Positional cloning of zebrafish ferroportin1 identifies a conserved vertebrate iron exporter. Nature 2000, 403, 776–781. [Google Scholar] [CrossRef] [PubMed]
  54. Fuqua, B.K.; Lu, Y.; Darshan, D.; Frazer, D.M.; Wilkins, S.J.; Wolkow, N.; Bell, A.G.; Hsu, J.; Yu, C.C.; Chen, H.; et al. The multicopper ferroxidase hephaestin enhances intestinal iron absorption in mice. PLoS ONE 2014, 9, e98792. [Google Scholar] [CrossRef] [PubMed]
  55. Kuo, Y.M.; Su, T.; Chen, H.; Attieh, Z.; Syed, B.A.; McKie, A.T.; Anderson, G.J.; Gitschier, J.; Vulpe, C.D. Mislocalisation of hephaestin, a multicopper ferroxidase involved in basolateral intestinal iron transport, in the sex linked anaemia mouse. Gut 2004, 53, 201–206. [Google Scholar] [CrossRef] [PubMed]
  56. Reeves, P.G.; Demars, L.C.; Johnson, W.T.; Lukaski, H.C. Dietary copper deficiency reduces iron absorption and duodenal enterocyte hephaestin protein in male and female rats. J. Nutr. 2005, 135, 92–98. [Google Scholar] [CrossRef] [PubMed]
  57. Stearman, R.; Yuan, D.S.; Yamaguchi-Iwai, Y.; Klausner, R.D.; Dancis, A. A permease-oxidase complex involved in high-affinity iron uptake in yeast. Science 1996, 271, 1552–1557. [Google Scholar] [CrossRef] [PubMed]
  58. Doguer, C.; Ha, J.H.; Collins, J.F. Intersection of Iron and Copper Metabolism in the Mammalian Intestine and Liver. Compr. Physiol. 2018, 8, 1433–1461. [Google Scholar] [CrossRef] [PubMed]
  59. Petrak, J.; Vyoral, D. Hephaestin—A ferroxidase of cellular iron export. Int. J. Biochem. Cell Biol. 2005, 37, 1173–1178. [Google Scholar] [CrossRef]
  60. Anderson, G.J.; Frazer, D.M.; McKie, A.T.; Vulpe, C.D. The ceruloplasmin homolog hephaestin and the control of intestinal iron absorption. Blood Cells Mol. Dis. 2002, 29, 367–375. [Google Scholar] [CrossRef]
  61. Helman, S.L.; Zhou, J.; Fuqua, B.K.; Lu, Y.; Collins, J.F.; Chen, H.; Vulpe, C.D.; Anderson, G.J.; Frazer, D.M. The biology of mammalian multi-copper ferroxidases. Biometals 2023, 36, 263–281. [Google Scholar] [CrossRef]
  62. Vashchenko, G.; MacGillivray, R.T. Multi-copper oxidases and human iron metabolism. Nutrients 2013, 5, 2289–2313. [Google Scholar] [CrossRef]
  63. Mackenzie, B.; Hediger, M.A. SLC11 family of H+-coupled metal-ion transporters NRAMP1 and DMT1. Pflugers Arch. 2004, 447, 571–579. [Google Scholar] [CrossRef]
  64. Gkouvatsos, K.; Papanikolaou, G.; Pantopoulos, K. Regulation of iron transport and the role of transferrin. Biochim. Biophys. Acta 2012, 1820, 188–202. [Google Scholar] [CrossRef]
  65. Nandakumar, S.K.; Ulirsch, J.C.; Sankaran, V.G. Advances in understanding erythropoiesis: Evolving perspectives. Br. J. Haematol. 2016, 173, 206–218. [Google Scholar] [CrossRef]
  66. Palis, J. Primitive and definitive erythropoiesis in mammals. Front. Physiol. 2014, 5, 3. [Google Scholar] [CrossRef]
  67. Dzierzak, E.; Philipsen, S. Erythropoiesis: Development and differentiation. Cold Spring Harb. Perspect. Med. 2013, 3, a011601. [Google Scholar] [CrossRef]
  68. Koury, M.J.; Ponka, P. New insights into erythropoiesis: The roles of folate, vitamin B12, and iron. Annu. Rev. Nutr. 2004, 24, 105–131. [Google Scholar] [CrossRef]
  69. Visentin, M.; Diop-Bove, N.; Zhao, R.; Goldman, I.D. The intestinal absorption of folates. Annu. Rev. Physiol. 2014, 76, 251–274. [Google Scholar] [CrossRef]
  70. Baker, H.; Thomson, A.D.; Feingold, S.; Frank, O. Role of the jejunum in the absorption of folic acid and its polyglutamates. Am. J. Clin. Nutr. 1969, 22, 124–132. [Google Scholar] [CrossRef]
  71. Kozyraki, R.; Cases, O. Vitamin B12 absorption: Mammalian physiology and acquired and inherited disorders. Biochimie 2013, 95, 1002–1007. [Google Scholar] [CrossRef]
  72. Cooper, B.A. Complex of intrinsic factor and B12 in human ileum during vitamin B12 absorption. Am. J. Physiol. 1968, 214, 832–835. [Google Scholar] [CrossRef]
  73. Valman, H.B.; Roberts, P.D. Vitamin B12 absorption after resection of ileum in childhood. Arch. Dis. Child. 1974, 49, 932–935. [Google Scholar] [CrossRef]
  74. Jacobson, L.O.; Goldwasser, E.; Fried, W.; Plzak, L. Role of the kidney in erythropoiesis. Nature 1957, 179, 633–634. [Google Scholar] [CrossRef]
  75. Bachmann, S.; Le Hir, M.; Eckardt, K.U. Co-localization of erythropoietin mRNA and ecto-5′-nucleotidase immunoreactivity in peritubular cells of rat renal cortex indicates that fibroblasts produce erythropoietin. J. Histochem. Cytochem. 1993, 41, 335–341. [Google Scholar] [CrossRef]
  76. Koury, S.T.; Bondurant, M.C.; Koury, M.J. Localization of erythropoietin synthesizing cells in murine kidneys by in situ hybridization. Blood 1988, 71, 524–527. [Google Scholar] [CrossRef]
  77. Lacombe, C.; Da Silva, J.L.; Bruneval, P.; Fournier, J.G.; Wendling, F.; Casadevall, N.; Camilleri, J.P.; Bariety, J.; Varet, B.; Tambourin, P. Peritubular cells are the site of erythropoietin synthesis in the murine hypoxic kidney. J. Clin. Investig. 1988, 81, 620–623. [Google Scholar] [CrossRef]
  78. Tojo, Y.; Sekine, H.; Hirano, I.; Pan, X.; Souma, T.; Tsujita, T.; Kawaguchi, S.; Takeda, N.; Takeda, K.; Fong, G.H.; et al. Hypoxia Signaling Cascade for Erythropoietin Production in Hepatocytes. Mol. Cell Biol. 2015, 35, 2658–2672. [Google Scholar] [CrossRef]
  79. Erslev, A.J.; Caro, J.; Kansu, E.; Silver, R. Renal and extrarenal erythropoietin production in anaemic rats. Br. J. Haematol. 1980, 45, 65–72. [Google Scholar] [CrossRef]
  80. Maxwell, P.H.; Ferguson, D.J.; Osmond, M.K.; Pugh, C.W.; Heryet, A.; Doe, B.G.; Johnson, M.H.; Ratcliffe, P.J. Expression of a homologously recombined erythopoietin-SV40 T antigen fusion gene in mouse liver: Evidence for erythropoietin production by Ito cells. Blood 1994, 84, 1823–1830. [Google Scholar] [CrossRef]
  81. Suzuki, N.; Yamamoto, M. Roles of renal erythropoietin-producing (REP) cells in the maintenance of systemic oxygen homeostasis. Pflugers Arch. 2016, 468, 3–12. [Google Scholar] [CrossRef]
  82. Weidemann, A.; Johnson, R.S. Nonrenal regulation of EPO synthesis. Kidney Int. 2009, 75, 682–688. [Google Scholar] [CrossRef]
  83. Moritz, K.M.; Lim, G.B.; Wintour, E.M. Developmental regulation of erythropoietin and erythropoiesis. Am. J. Physiol. 1997, 273, R1829–R1844. [Google Scholar] [CrossRef]
  84. Latour, C.; Kautz, L.; Besson-Fournier, C.; Island, M.L.; Canonne-Hergaux, F.; Loreal, O.; Ganz, T.; Coppin, H.; Roth, M.P. Testosterone perturbs systemic iron balance through activation of epidermal growth factor receptor signaling in the liver and repression of hepcidin. Hepatology 2014, 59, 683–694. [Google Scholar] [CrossRef]
  85. Hennigar, S.R.; Berryman, C.E.; Harris, M.N.; Karl, J.P.; Lieberman, H.R.; McClung, J.P.; Rood, J.C.; Pasiakos, S.M. Testosterone Administration during Energy Deficit Suppresses Hepcidin and Increases Iron Availability for Erythropoiesis. J. Clin. Endocrinol. Metab. 2020, 105, e1316–e1321. [Google Scholar] [CrossRef]
  86. Coviello, A.D.; Kaplan, B.; Lakshman, K.M.; Chen, T.; Singh, A.B.; Bhasin, S. Effects of graded doses of testosterone on erythropoiesis in healthy young and older men. J. Clin. Endocrinol. Metab. 2008, 93, 914–919. [Google Scholar] [CrossRef]
  87. Park, C.H.; Valore, E.V.; Waring, A.J.; Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem. 2001, 276, 7806–7810. [Google Scholar] [CrossRef]
  88. Pigeon, C.; Ilyin, G.; Courselaud, B.; Leroyer, P.; Turlin, B.; Brissot, P.; Loreal, O. 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, 7811–7819. [Google Scholar] [CrossRef]
  89. Katsarou, A.; Pantopoulos, K. Hepcidin Therapeutics. Pharmaceuticals 2018, 11, 127. [Google Scholar] [CrossRef]
  90. Nemeth, E.; Tuttle, M.S.; Powelson, J.; Vaughn, M.B.; Donovan, A.; Ward, D.M.; Ganz, T.; Kaplan, J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004, 306, 2090–2093. [Google Scholar] [CrossRef]
  91. De Domenico, I.; Ward, D.M.; Langelier, C.; Vaughn, M.B.; Nemeth, E.; Sundquist, W.I.; Ganz, T.; Musci, G.; Kaplan, J. The molecular mechanism of hepcidin-mediated ferroportin down-regulation. Mol. Biol. Cell 2007, 18, 2569–2578. [Google Scholar] [CrossRef]
  92. Nicolas, G.; Chauvet, C.; Viatte, L.; Danan, J.L.; Bigard, X.; Devaux, I.; Beaumont, C.; Kahn, A.; Vaulont, S. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J. Clin. Investig. 2002, 110, 1037–1044. [Google Scholar] [CrossRef]
  93. Tsukaguchi, H.; Tokui, T.; Mackenzie, B.; Berger, U.V.; Chen, X.Z.; Wang, Y.; Brubaker, R.F.; Hediger, M.A. A family of mammalian Na+-dependent L-ascorbic acid transporters. Nature 1999, 399, 70–75. [Google Scholar] [CrossRef]
  94. Ulloa, V.; Garcia-Robles, M.; Martinez, F.; Salazar, K.; Reinicke, K.; Perez, F.; Godoy, D.F.; Godoy, A.S.; Nualart, F. Human choroid plexus papilloma cells efficiently transport glucose and vitamin C. J. Neurochem. 2013, 127, 403–414. [Google Scholar] [CrossRef]
  95. Amano, A.; Aigaki, T.; Maruyama, N.; Ishigami, A. Ascorbic acid depletion enhances expression of the sodium-dependent vitamin C transporters, SVCT1 and SVCT2, and uptake of ascorbic acid in livers of SMP30/GNL knockout mice. Arch. Biochem. Biophys. 2010, 496, 38–44. [Google Scholar] [CrossRef]
  96. Kobayashi, T.A.; Shimada, H.; Sano, F.K.; Itoh, Y.; Enoki, S.; Okada, Y.; Kusakizako, T.; Nureki, O. Dimeric transport mechanism of human vitamin C transporter SVCT1. Nat. Commun. 2024, 15, 5569. [Google Scholar] [CrossRef]
  97. Mann, G.V.; Newton, P. The membrane transport of ascorbic acid. Ann. N. Y. Acad. Sci. 1975, 258, 243–252. [Google Scholar] [CrossRef]
  98. Ingermann, R.L.; Stankova, L.; Bigley, R.H.; Bissonnette, J.M. Effect of monosaccharide on dehydroascorbic acid uptake by placental membrane vesicles. J. Clin. Endocrinol. Metab. 1988, 67, 389–394. [Google Scholar] [CrossRef]
  99. Hornung, T.C.; Biesalski, H.K. Glut-1 explains the evolutionary advantage of the loss of endogenous vitamin C-synthesis: The electron transfer hypothesis. Evol. Med. Public Health 2019, 2019, 221–231. [Google Scholar] [CrossRef]
  100. Goldenberg, H.; Schweinzer, E. Transport of vitamin C in animal and human cells. J. Bioenerg. Biomembr. 1994, 26, 359–367. [Google Scholar] [CrossRef]
  101. Skou, J.C. Enzymatic Basis for Active Transport of Na+ and K+ across Cell Membrane. Physiol. Rev. 1965, 45, 596–617. [Google Scholar] [CrossRef]
  102. Cereijido, M.; Shoshani, L.; Contreras, R.G. The polarized distribution of Na+, K+-ATPase and active transport across epithelia. J. Membr. Biol. 2001, 184, 299–304. [Google Scholar] [CrossRef]
  103. Sage, J.M.; Carruthers, A. Human erythrocytes transport dehydroascorbic acid and sugars using the same transporter complex. Am. J. Physiol. Cell Physiol. 2014, 306, C910–C917. [Google Scholar] [CrossRef]
  104. Montel-Hagen, A.; Kinet, S.; Manel, N.; Mongellaz, C.; Prohaska, R.; Battini, J.L.; Delaunay, J.; Sitbon, M.; Taylor, N. Erythrocyte Glut1 triggers dehydroascorbic acid uptake in mammals unable to synthesize vitamin C. Cell 2008, 132, 1039–1048. [Google Scholar] [CrossRef]
  105. Montel-Hagen, A.; Sitbon, M.; Taylor, N. Erythroid glucose transporters. Curr. Opin. Hematol. 2009, 16, 165–172. [Google Scholar] [CrossRef]
  106. May, J.M.; Qu, Z.C.; Whitesell, R.R.; Cobb, C.E. Ascorbate recycling in human erythrocytes: Role of GSH in reducing dehydroascorbate. Free Radic. Biol. Med. 1996, 20, 543–551. [Google Scholar] [CrossRef]
  107. Hughes, R.E. Reduction of Dehydroasorbic Acid by Animal Tissues. Nature 1964, 203, 1068–1069. [Google Scholar] [CrossRef]
  108. Dereven’kov, I.A.; Makarov, S.V.; Bui Thi, T.T.; Makarova, A.S.; Koifman, O.I. Studies on the reduction of dehydroascorbic acid by glutathione in the presence of aquahydroxocobinamide. Eur. J. Inorg. Chem. 2018, 2018, 2987–2992. [Google Scholar] [CrossRef]
  109. Sasaki, H.; Giblin, F.J.; Winkler, B.S.; Chakrapani, B.; Leverenz, V.; Shu, C.C. A protective role for glutathione-dependent reduction of dehydroascorbic acid in lens epithelium. Investig. Ophthalmol. Vis. Sci. 1995, 36, 1804–1817. [Google Scholar]
  110. Cisternas, P.; Silva-Alvarez, C.; Martinez, F.; Fernandez, E.; Ferrada, L.; Oyarce, K.; Salazar, K.; Bolanos, J.P.; Nualart, F. The oxidized form of vitamin C, dehydroascorbic acid, regulates neuronal energy metabolism. J. Neurochem. 2014, 129, 663–671. [Google Scholar] [CrossRef]
  111. Puskas, F.; Gergely, P., Jr.; Banki, K.; Perl, A. Stimulation of the pentose phosphate pathway and glutathione levels by dehydroascorbate, the oxidized form of vitamin C. FASEB J. 2000, 14, 1352–1361. [Google Scholar] [CrossRef]
  112. Orringer, E.P.; Roer, M.E. An ascorbate-mediated transmembrane-reducing system of the human erythrocyte. J. Clin. Investig. 1979, 63, 53–58. [Google Scholar] [CrossRef]
  113. D’Alessandro, A.; Keele, G.R.; Hay, A.; Nemkov, T.; Earley, E.J.; Stephenson, D.; Vincent, M.; Deng, X.; Stone, M.; Dzieciatkowska, M.; et al. Ferroptosis regulates hemolysis in stored murine and human red blood cells. bioRxiv 2024. [Google Scholar] [CrossRef]
  114. Howie, H.L.; Hay, A.M.; de Wolski, K.; Waterman, H.; Lebedev, J.; Fu, X.; Culp-Hill, R.; D’Alessandro, A.; Gorham, J.D.; Ranson, M.S.; et al. Differences in Steap3 expression are a mechanism of genetic variation of RBC storage and oxidative damage in mice. Blood Adv. 2019, 3, 2272–2285. [Google Scholar] [CrossRef]
  115. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
  116. Callender, S.T.; Powell, E.; Witts, L. The life-span of the red cell in man. J. Pathol. Bacteriol. 1945, 57, 129–139. [Google Scholar] [CrossRef]
  117. Larkin, J.M.; Donzell, W.C.; Anderson, R.G. Potassium-dependent assembly of coated pits: New coated pits form as planar clathrin lattices. J. Cell Biol. 1986, 103, 2619–2627. [Google Scholar] [CrossRef]
  118. Song, Q.; Meng, B.; Xu, H.; Mao, Z. The emerging roles of vacuolar-type ATPase-dependent Lysosomal acidification in neurodegenerative diseases. Transl. Neurodegener. 2020, 9, 17. [Google Scholar] [CrossRef]
  119. Harvey, W.R. Physiology of V-ATPases. J. Exp. Biol. 1992, 172, 1–17. [Google Scholar] [CrossRef]
  120. Touret, N.; Furuya, W.; Forbes, J.; Gros, P.; Grinstein, S. Dynamic traffic through the recycling compartment couples the metal transporter Nramp2 (DMT1) with the transferrin receptor. J. Biol. Chem. 2003, 278, 25548–25557. [Google Scholar] [CrossRef]
  121. Eckenroth, B.E.; Steere, A.N.; Chasteen, N.D.; Everse, S.J.; Mason, A.B. How the binding of human transferrin primes the transferrin receptor potentiating iron release at endosomal pH. Proc. Natl. Acad. Sci. USA 2011, 108, 13089–13094. [Google Scholar] [CrossRef]
  122. Dautry-Varsat, A.; Ciechanover, A.; Lodish, H.F. pH and the recycling of transferrin during receptor-mediated endocytosis. Proc. Natl. Acad. Sci. USA 1983, 80, 2258–2262. [Google Scholar] [CrossRef]
  123. Ohgami, R.S.; Campagna, D.R.; Greer, E.L.; Antiochos, B.; McDonald, A.; Chen, J.; Sharp, J.J.; Fujiwara, Y.; Barker, J.E.; Fleming, M.D. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat. Genet. 2005, 37, 1264–1269. [Google Scholar] [CrossRef]
  124. Yanatori, I.; Kishi, F. DMT1 and iron transport. Free Radic. Biol. Med. 2019, 133, 55–63. [Google Scholar] [CrossRef]
  125. Grandchamp, B.; Hetet, G.; Kannengiesser, C.; Oudin, C.; Beaumont, C.; Rodrigues-Ferreira, S.; Amson, R.; Telerman, A.; Nielsen, P.; Kohne, E.; et al. A novel type of congenital hypochromic anemia associated with a nonsense mutation in the STEAP3/TSAP6 gene. Blood 2011, 118, 6660–6666. [Google Scholar] [CrossRef]
  126. Blanc, L.; Papoin, J.; Debnath, G.; Vidal, M.; Amson, R.; Telerman, A.; An, X.; Mohandas, N. Abnormal erythroid maturation leads to microcytic anemia in the TSAP6/Steap3 null mouse model. Am. J. Hematol. 2015, 90, 235–241. [Google Scholar] [CrossRef]
  127. Zhou, Z.; Zhen, J.; Karpowich, N.K.; Goetz, R.M.; Law, C.J.; Reith, M.E.; Wang, D.N. LeuT-desipramine structure reveals how antidepressants block neurotransmitter reuptake. Science 2007, 317, 1390–1393. [Google Scholar] [CrossRef]
  128. Ordway, G.A.; Jia, W.; Li, J.; Zhu, M.Y.; Mandela, P.; Pan, J. Norepinephrine transporter function and desipramine: Residual drug effects versus short-term regulation. J. Neurosci. Methods 2005, 143, 217–225. [Google Scholar] [CrossRef]
  129. Zhu, M.Y.; Kyle, P.B.; Hume, A.S.; Ordway, G.A. The persistent membrane retention of desipramine causes lasting inhibition of norepinephrine transporter function. Neurochem. Res. 2004, 29, 419–427. [Google Scholar] [CrossRef]
  130. Andersen, J.; Kristensen, A.S.; Bang-Andersen, B.; Stromgaard, K. Recent advances in the understanding of the interaction of antidepressant drugs with serotonin and norepinephrine transporters. Chem. Commun. 2009, 15, 3677–3692. [Google Scholar] [CrossRef]
  131. Hyman, S.E.; Nestler, E.J. Initiation and adaptation: A paradigm for understanding psychotropic drug action. Am. J. Psychiatry 1996, 153, 151–162. [Google Scholar] [CrossRef] [PubMed]
  132. Elojeimy, S.; Holman, D.H.; Liu, X.; El-Zawahry, A.; Villani, M.; Cheng, J.C.; Mahdy, A.; Zeidan, Y.; Bielwaska, A.; Hannun, Y.A.; et al. New insights on the use of desipramine as an inhibitor for acid ceramidase. FEBS Lett. 2006, 580, 4751–4756. [Google Scholar] [CrossRef] [PubMed]
  133. Hurwitz, R.; Ferlinz, K.; Sandhoff, K. The tricyclic antidepressant desipramine causes proteolytic degradation of lysosomal sphingomyelinase in human fibroblasts. Biol. Chem. Hoppe Seyler 1994, 375, 447–450. [Google Scholar] [CrossRef] [PubMed]
  134. Kolzer, M.; Werth, N.; Sandhoff, K. Interactions of acid sphingomyelinase and lipid bilayers in the presence of the tricyclic antidepressant desipramine. FEBS Lett. 2004, 559, 96–98. [Google Scholar] [CrossRef] [PubMed]
  135. Lai, M.; Realini, N.; La Ferla, M.; Passalacqua, I.; Matteoli, G.; Ganesan, A.; Pistello, M.; Mazzanti, C.M.; Piomelli, D. Complete Acid Ceramidase ablation prevents cancer-initiating cell formation in melanoma cells. Sci. Rep. 2017, 7, 7411. [Google Scholar] [CrossRef] [PubMed]
  136. Saad, A.F.; Meacham, W.D.; Bai, A.; Anelli, V.; Elojeimy, S.; Mahdy, A.E.; Turner, L.S.; Cheng, J.; Bielawska, A.; Bielawski, J.; et al. The functional effects of acid ceramidase overexpression in prostate cancer progression and resistance to chemotherapy. Cancer Biol. Ther. 2007, 6, 1455–1460. [Google Scholar] [CrossRef] [PubMed]
  137. Seelan, R.S.; Qian, C.; Yokomizo, A.; Bostwick, D.G.; Smith, D.I.; Liu, W. Human acid ceramidase is overexpressed but not mutated in prostate cancer. Genes. Chromosomes Cancer 2000, 29, 137–146. [Google Scholar] [CrossRef] [PubMed]
  138. Beckham, T.H.; Lu, P.; Cheng, J.C.; Zhao, D.; Turner, L.S.; Zhang, X.; Hoffman, S.; Armeson, K.E.; Liu, A.; Marrison, T.; et al. Acid ceramidase-mediated production of sphingosine 1-phosphate promotes prostate cancer invasion through upregulation of cathepsin B. Int. J. Cancer 2012, 131, 2034–2043. [Google Scholar] [CrossRef] [PubMed]
  139. Awojoodu, A.O.; Keegan, P.M.; Lane, A.R.; Zhang, Y.; Lynch, K.R.; Platt, M.O.; Botchwey, E.A. Acid sphingomyelinase is activated in sickle cell erythrocytes and contributes to inflammatory microparticle generation in SCD. Blood 2014, 124, 1941–1950. [Google Scholar] [CrossRef]
  140. Lang, P.A.; Schenck, M.; Nicolay, J.P.; Becker, J.U.; Kempe, D.S.; Lupescu, A.; Koka, S.; Eisele, K.; Klarl, B.A.; Rubben, H.; et al. Liver cell death and anemia in Wilson disease involve acid sphingomyelinase and ceramide. Nat. Med. 2007, 13, 164–170. [Google Scholar] [CrossRef]
  141. Momchilova, A.; Pankov, R.; Alexandrov, A.; Markovska, T.; Pankov, S.; Krastev, P.; Staneva, G.; Vassileva, E.; Krastev, N.; Pinkas, A. Sphingolipid Catabolism and Glycerophospholipid Levels Are Altered in Erythrocytes and Plasma from Multiple Sclerosis Patients. Int. J. Mol. Sci. 2022, 23, 7592. [Google Scholar] [CrossRef] [PubMed]
  142. Choi, S.Y.; Li, J.; Jo, S.H.; Lee, S.J.; Oh, S.B.; Kim, J.S.; Lee, J.H.; Park, K. Desipramine inhibits Na+/H+ exchanger in human submandibular cells. J. Dent. Res. 2006, 85, 839–843. [Google Scholar] [CrossRef] [PubMed]
  143. Ellegaard, A.M.; Dehlendorff, C.; Vind, A.C.; Anand, A.; Cederkvist, L.; Petersen, N.H.T.; Nylandsted, J.; Stenvang, J.; Mellemgaard, A.; Osterlind, K.; et al. Repurposing Cationic Amphiphilic Antihistamines for Cancer Treatment. EBioMedicine 2016, 9, 130–139. [Google Scholar] [CrossRef] [PubMed]
  144. Petersen, N.H.; Olsen, O.D.; Groth-Pedersen, L.; Ellegaard, A.M.; Bilgin, M.; Redmer, S.; Ostenfeld, M.S.; Ulanet, D.; Dovmark, T.H.; Lonborg, A.; et al. Transformation-associated changes in sphingolipid metabolism sensitize cells to lysosomal cell death induced by inhibitors of acid sphingomyelinase. Cancer Cell 2013, 24, 379–393. [Google Scholar] [CrossRef] [PubMed]
  145. Arimochi, H.; Morita, K. Desipramine induces apoptotic cell death through nonmitochondrial and mitochondrial pathways in different types of human colon carcinoma cells. Pharmacology 2008, 81, 164–172. [Google Scholar] [CrossRef] [PubMed]
  146. Pakkanen, K.; Salonen, E.; Makela, A.R.; Oker-Blom, C.; Vattulainen, I.; Vuento, M. Desipramine induces disorder in cholesterol-rich membranes: Implications for viral trafficking. Phys. Biol. 2009, 6, 046004. [Google Scholar] [CrossRef] [PubMed]
  147. Salata, C.; Calistri, A.; Parolin, C.; Baritussio, A.; Palu, G. Antiviral activity of cationic amphiphilic drugs. Expert. Rev. Anti Infect. Ther. 2017, 15, 483–492. [Google Scholar] [CrossRef]
  148. Pan, X.; Giustarini, D.; Lang, F.; Rossi, R.; Wieder, T.; Koberle, M.; Ghashghaeinia, M. Desipramine induces eryptosis in human erythrocytes, an effect blunted by nitric oxide donor sodium nitroprusside and N-acetyl-L-cysteine but enhanced by Calcium depletion. Cell Cycle 2023, 22, 1827–1853. [Google Scholar] [CrossRef]
Figure 1. Transmembrane H2S diffusion and Band-3 mediated Cl/HS exchange in hRBCs. Methemoglobin (Hb-Fe3+)-mediated H2S degradation ensures the maintenance of physiological plasma and tissue concentration of free H2S. The Cl/HS/H2S cycle is also efficiently involved in net acid (H+-ions) efflux; for more details, see the following review [15]. For interactions of hRBCs with endogenous cells and pathogens, see the main text.
Figure 1. Transmembrane H2S diffusion and Band-3 mediated Cl/HS exchange in hRBCs. Methemoglobin (Hb-Fe3+)-mediated H2S degradation ensures the maintenance of physiological plasma and tissue concentration of free H2S. The Cl/HS/H2S cycle is also efficiently involved in net acid (H+-ions) efflux; for more details, see the following review [15]. For interactions of hRBCs with endogenous cells and pathogens, see the main text.
Antioxidants 13 00968 g001
Figure 2. Uptake and systemic circulation of non-heme iron require several carriers located in the cell membrane of human enterocytes, plasma protein transferrin, and transferrin receptor. Erythropoiesis requires liver- and kidney-dependent production of erythropoietin.
Figure 2. Uptake and systemic circulation of non-heme iron require several carriers located in the cell membrane of human enterocytes, plasma protein transferrin, and transferrin receptor. Erythropoiesis requires liver- and kidney-dependent production of erythropoietin.
Antioxidants 13 00968 g002
Figure 3. (A) Inverse correlation between DMT1 abundance and pH along the small intestinal. (B) GLUT-1-dependent influx of glucose into human enterocytes. DMT1: divalent metal transporter 1; GLUT-1: glucose transporter-1.
Figure 3. (A) Inverse correlation between DMT1 abundance and pH along the small intestinal. (B) GLUT-1-dependent influx of glucose into human enterocytes. DMT1: divalent metal transporter 1; GLUT-1: glucose transporter-1.
Antioxidants 13 00968 g003
Figure 4. GLUT-1-mediated influx of oxidized form of vitamin C (DHA) into the mature hRBCs. The interplay between DHA, PPP, GSH, AA, and the subsequent reduction of vitamin E prevents lipid peroxidation. As a result, cell membrane integrity is maintained and in vivo hemolysis of erythrocytes is minimized. Recently, a link between iron metabolism, lipid peroxidation, and hemolysis was found in stored human and mice erythrocytes [113,114]. Vitamin E is located inside the cell membrane.
Figure 4. GLUT-1-mediated influx of oxidized form of vitamin C (DHA) into the mature hRBCs. The interplay between DHA, PPP, GSH, AA, and the subsequent reduction of vitamin E prevents lipid peroxidation. As a result, cell membrane integrity is maintained and in vivo hemolysis of erythrocytes is minimized. Recently, a link between iron metabolism, lipid peroxidation, and hemolysis was found in stored human and mice erythrocytes [113,114]. Vitamin E is located inside the cell membrane.
Antioxidants 13 00968 g004
Figure 5. Iron transport into erythroid precursors. This process comprises the endocytosis of Transferrin-bound iron, DMT1B-II-mediated Fe2+ export from acidified endosomes into the cytoplasm. MT: mitochondria.
Figure 5. Iron transport into erythroid precursors. This process comprises the endocytosis of Transferrin-bound iron, DMT1B-II-mediated Fe2+ export from acidified endosomes into the cytoplasm. MT: mitochondria.
Antioxidants 13 00968 g005
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pan, X.; Köberle, M.; Ghashghaeinia, M. Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes. Antioxidants 2024, 13, 968. https://doi.org/10.3390/antiox13080968

AMA Style

Pan X, Köberle M, Ghashghaeinia M. Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes. Antioxidants. 2024; 13(8):968. https://doi.org/10.3390/antiox13080968

Chicago/Turabian Style

Pan, Xia, Martin Köberle, and Mehrdad Ghashghaeinia. 2024. "Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes" Antioxidants 13, no. 8: 968. https://doi.org/10.3390/antiox13080968

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop