Stoichiometries of transferrin receptors 1 and 2 in human liver

https://doi.org/10.1016/j.bcmd.2009.09.004Get rights and content

Abstract

Mutations in either the hereditary hemochromatosis protein, HFE, or transferrin receptor 2, TfR2, result in a similarly severe form of the most common type of iron overload disease called hereditary hemochromatosis. Models of the interactions between HFE, TfR1, and TfR2 imply that these proteins are present in different molar concentrations in the liver, where they control expression of the iron regulatory hormone, hepcidin, in response to body iron loading. The aim of this study was to determine in vivo levels of mRNA by quantitative RT-PCR and concentrations of these proteins by quantitative immunoblotting in human liver tissues. The level of TfR2 mRNA was 21- and 63-fold higher than that of TfR1 and HFE, respectively. Molar concentration of TfR2 protein was the highest and determined to be 1.95 nmol/g protein in whole cell lysates and 10.89 nmol/g protein in microsomal membranes. Molar concentration of TfR1 protein was 4.5- and 6.1-fold lower than that of TfR2 in whole cell lysates and membranes, respectively. The level of HFE protein was below 0.53 nmol/g of total protein. HFE is thus present in substoichiometric concentrations with respect to both TfR1 and TfR2 in human liver tissue. This finding supports a model, in which availability of HFE is limiting for formation of complexes with TfR1 or TfR2.

Introduction

Hereditary hemochromatosis (HH)1 is an autosomal, inherited disorder of iron homeostasis characterized by hepatocellular iron overload that ranges from mild to severe (reviewed in ref. [1], [2], [3]). HH is associated with mutations in at least five genes. On the basis of the gene involved, HH is classified as type 1 (hereditary hemochromatosis, HFE) [4], type 2A (hemojuvelin, HFE2) [5], type 2B (hepcidin, HAMP) [6], type 3 (transferrin receptor 2, TFR2) [7], and type 4 (ferroportin, FPN) [8]. Type 1 is the most common form of HH [4].

HFE is a type I transmembrane protein that belongs to the MHC-I like family of proteins. Like MHC-I proteins, HFE also forms a heterodimer with β2-microglobulin (β2M) [4], [9]. The most common mutation in the HFE protein, C282Y [4], results in destabilization of the α3 domain, which abrogates the interaction between HFE and β2M [4]. As a result, the mutant C282Y-HFE protein has impaired ability to reach the cell surface [4], [10], [11]. The second most common mutation is H63D [4], but the mechanism by which this mutation causes HH is unknown. Interestingly, there is a considerable variation in iron loading in individuals with these two mutations [1], [2]. Such heterogeneity suggests that HFE function depends on the presence of modifiers, which might be proteins that interact with HFE.

The first identified binding partner of HFE was the transferrin receptor 1 (TfR1) [12], [13], a ubiquitous cell surface receptor that binds and internalizes iron-loaded transferrin (holo-Tf). HFE/TfR1 complex dissociates in the presence of holo-Tf because holo-Tf competes with HFE for binding to TfR1 [14], [15], [16]. The discovery that hepcidin, an iron regulatory hormone predominantly expressed in hepatocytes [17], is decreased in both HH type 1 patients [18] and Hfe-/- mice [19], [20] and that HFE is also predominantly expressed in hepatocytes [21] indicated that the primary site of HFE effects on iron homeostasis is the liver. These observations lead to a “hepcidin hypothesis,” in which HFE is an upstream regulator of hepcidin expression (reviewed in ref. [22]). Observations that mice lacking Hfe in the crypt- and villi-enterocytes have no detectable iron loading [23] while mice lacking Hfe in hepatocytes manifest iron overload [24] emphasize the importance of HFE expression in hepatocytes.

Recently, transferrin receptor 2 (TfR2), a homolog of TfR1 that is predominantly expressed in hepatocytes [25], was reported to bind to HFE [26]. Interestingly, the interacting domains of HFE and TfR2 [27] are different from those of HFE and TfR1. First, HFE interacts with TfR2 via its α3 domain, versus with TfR1 via its α1 and α2 domains. Second, the Tf binding site of TfR2 does not overlap with the HFE binding site as it does in TfR1. Thus, in contrast to the HFE/TfR1 complex, the HFE/TfR2 complex does not dissociate, even in the presence of high Tf concentrations [27]. This finding suggests a new model of HFE-dependent regulation of hepcidin expression, in which HFE is released from TfR1 and binds to TfR2, with increasing iron-loaded Tf concentrations. Two recent studies expand these findings. The first work analyzes Hfe and Tfr1 interactions in mice models of HH type 1. Expression of mutant forms of mouse Tfr1 that either prevent or stabilize Hfe/Tfr1 interactions results in an Hfe-dependent induction of hepcidin expression, which occurs only when Hfe is dissociated from Tfr1 [28]. The second study demonstrates that in the presence of holo-Tf, human hepatoma cells that express undetectable HFE but readily detectable TfR1 and TfR2 proteins regulate hepcidin expression only when exogenous HFE is expressed [29]. These studies lead to the proposal that TfR1 sequesters HFE from TfR2 under low iron conditions, but under high iron conditions, the increased iron saturation of Tf shifts the balance towards creation of an HFE/TFR2 hepcidin signaling complex [28]. In this process, HFE represents the limiting factor during reorganization of HFE/TfR1 and HFE/TfR2 complexes [29].

In order to better understand the mechanism by which these complexes are formed as well as their response to iron levels, it is important to know the relative amounts of HFE, TfR1, and TfR2 in the liver. Thus, we tested the hypothesis that in human liver, where the HFE-dependent regulation of hepcidin expression occurs, the molar concentration of HFE is similar to or lower than that of TfR1 or TfR2. Both the mRNA and protein levels of HFE, TfR1, and TfR2 in human liver tissues were measured. Our results showed that mRNA and protein levels of TfR2 are significantly higher than TfR1 and HFE levels. The least abundant is the HFE protein, supporting the proposed model of hepcidin regulation in vivo.

Section snippets

Cell culture

TRVb cells that lack the endogenous transferrin receptor 1 (TFRC) and that do not express detectable HFE and TfR2 (kindly provided by Dr. Timothy McGraw, Cornell University, NY) were grown in F-12 Coon's Modification, 5% fetal bovine serum (FBS) and 2 mg/ml glucose. TRVb/HFE/β2M cells stably expressing HFE with C-terminal FLAG epitope tag (HFE-FLAG) and β2M [30] were grown in the same medium supplemented with 300 μg/ml hygromycin. HeLa/tTA-HFE-FLAG cells [31] that stably express HFE-FLAG in a

Levels of HFE, TfR1, and TfR2 mRNAs

Recently, our group and others have focused on studies of HFE complexes with TfR1 and TfR2 in cultured cells [26], [27] and in mouse models [28] of HH. In this study, we decided to concentrate on analysis of in vivo levels of these individual players because such information is critical for interpretation of biochemical studies performed in hepatic tissues and hepatic cells. First, mRNA levels were analyzed. Total RNA was isolated from eight control liver samples, and the levels of HFE, TFRC,

Discussion

In this work, we determined the stoichiometries of TfR1 and TfR2, two key proteins involved in the regulation of hepcidin expression in response to holo-Tf levels. A recent model suggests that TfR1, via its interaction with HFE, controls levels of HFE that are available for interaction with another binding partner such as TfR2 [28], [29]. However, no in vivo data on the amounts of individual candidates existed that would support these suggestions. Therefore, we tested the hypothesis that HFE is

Aknowledgments

We thank to Kristin Diez-Sauter, Tul-Dim Cing, and Dara Partovi for technical assistance. We are also grateful to Katarina Luciakova, Juxing Chen, Junwei Gao, Julia Maxson and Kristina Nicholson for critical reading of the manuscript. This work was supported by National Institutes of Health Grants DK072166 and DK54488 (to C.A.E.) and in part by Medical Research Foundation of Oregon ACEBD0082 (to M.C.).

References (43)

  • L.M. Cullen et al.

    Genetics of hemochromatosis

    Annu. Rev. Med.

    (1999)
  • T.H. Bothwell et al.

    Hereditary hemochromatosis: etiologic, pathologic, and clinical aspects

    Sem. Hematol.

    (1998)
  • K.P. Batts

    Iron overload syndromes and the liver

    Mod. Pathol.

    (2007)
  • J.N. Feder et al.

    A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis

    Nat. Genet.

    (1996)
  • G. Papanikolaou et al.

    Mutations in HFE2 cause iron overload in chromosome 1q-linked juvenile hemochromatosis

    Nat. Genet.

    (2004)
  • A. Roetto et al.

    Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis

    Nat. Genet.

    (2003)
  • C. Camaschella et al.

    The gene TFR2 is mutated in a new type of haemochromatosis mapping to 7q22

    Nat. Genet.

    (2000)
  • G. Montosi et al.

    Autosomal-dominant hemochromatosis is associated with a mutation in the ferroportin (SLC11A3) gene

    J. Clin. Invest.

    (2001)
  • I.A. Wilson et al.

    Unusual MHC-like molecules: CD1, Fc receptor, the hemochromatosis gene product, and viral homologs

    Curr. Opin. Immunol.

    (1998)
  • J.N. Feder et al.

    The hemochromatosis founder mutation in HLA-H disrupts β2-microglobulin interaction and cell surface expression

    J. Cell. Biol.

    (1997)
  • A. Waheed et al.

    Hereditary hemochromatosis: effects of C282Y and H63D mutations on association with beta2-microglobulin, intracellular processing, and cell surface expression of the HFE protein in COS-7 cells

    Proc. Natl. Acad. Sci. U. S. A.

    (1997)
  • J.N. Feder et al.

    The hemochromatosis gene product complexes with the transferrin receptor and lowers its affinity for ligand binding

    Proc. Natl. Acad. Sci. U. S. A.

    (1998)
  • S. Parkkila et al.

    Association of the transferrin receptor in human placenta with HFE, the protein defective in hereditary hemochromatosis

    Proc. Natl. Acad. Sci. U. S. A.

    (1997)
  • J.A. Lebron et al.

    The hemochromatosis protein HFE competes. with transferrin for binding to the transferrin receptor

    J. Mol. Biol.

    (1999)
  • J.A. Lebron et al.

    The transferrin receptor binding site on HFE, the class I MHC-related protein mutated in hereditary hemochromatosis

    J. Mol. Biol.

    (1999)
  • M.J. Bennett et al.

    Crystal structure of the hereditary haemochromatosis protein HFE complexed with transferrin receptor

    Nature

    (2000)
  • T. Ganz

    Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation

    Blood

    (2003)
  • K.R. Bridle et al.

    Disrupted hepcidin regulation in HFE-associated haemochromatosis and the liver as a regulator of body iron homoeostasis

    Lancet

    (2003)
  • M. Muckenthaler et al.

    Regulatory defects in liver and intestine implicate abnormal hepcidin and Cybrd1 expression in mouse hemochromatosis

    Nat. Genet.

    (2003)
  • G. Nicolas et al.

    Constitutive hepcidin expression prevents iron overload in a mouse model of hemochromatosis

    Nat. Genet.

    (2003)
  • A.S. Zhang et al.

    Localization of iron metabolism-related mRNAs in rat liver indicate that HFE is expressed predominantly in hepatocytes

    Blood

    (2004)
  • Cited by (18)

    • Extrahepatic deficiency of transferrin receptor 2 is associated with increased erythropoiesis independent of iron overload

      2020, Journal of Biological Chemistry
      Citation Excerpt :

      Much like its homologue TFR1, TFR2 is capable of binding and internalizing iron-loaded transferrin (holo-Tf) (5). However, although TFR1 is expressed ubiquitously, TFR2 is largely restricted to hepatocytes and maturing erythrons (6), with liver having ˜6-fold more TFR2 than TFR1 (7). Unlike Tfr1, liver-specific knockout of murine Tfr2 results in iron overload (8, 9), implying that iron uptake is not the primary function of TFR2.

    • Physiology and pathophysiology of iron in hemoglobin-associated diseases

      2014, Free Radical Biology and Medicine
      Citation Excerpt :

      TfR2 is exclusively expressed in the liver and intestine and at levels 5.8 times higher in the liver than the intestine. Levels of TfR2 are much higher than those of TfR1 in human liver [32]. Both receptors preferentially bind diferric Tf, but the affinity of TfR1 for iron is 25 times higher than that of TfR2.

    • Hepatocyte-targeted HFE and TFR2 control hepcidin expression in mice

      2010, Blood
      Citation Excerpt :

      Importantly, we determined that, in wild-type mice, Hfe mRNA is a limiting factor in hepcidin signaling and that overexpression of Hfe increases hepcidin levels, which in turn decreases iron levels. This result is consistent with the observation that the molar amount of HFE protein is lower than that of TfR1 and TfR2 in solubilized human liver extracts.47 Superficially, these results appear to be in conflict with the idea that type 1 HH is a recessive disease.

    • Molecular basis of iron-loading disorders

      2010, Expert Reviews in Molecular Medicine
    View all citing articles on Scopus

    In this article, we have followed the numbering system of HFE, which starts at the first amino acid translated [43].

    View full text