Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Phys Chem Chem Phys ; 21(31): 16937-16948, 2019 Aug 21.
Article in English | MEDLINE | ID: mdl-31339131

ABSTRACT

Multi-frequency EPR spectroscopy can provide high-level structural information on high-spin Fe3+ sites in proteins and enzymes. Unfortunately, analysis of the EPR spectra of these spin systems is hindered by the presence of broad distributions in the zero-field-splitting (ZFS) parameters, which reflect conformational heterogeneity of the iron sites. We present the analysis of EPR spectra of high-spin Fe3+ bound to human serum transferrin. We apply a method termed the grid-of-errors to extract the distributions of the individual ZFS parameters from EPR spectra recorded in the high-field limit at a microwave frequency of 275 GHz. Study of a series of transferrin variants shows that the ZFS distributions are as characteristic of the structure of a high-spin Fe3+ site as the ZFS parameters themselves. Simulations based on the extracted ZFS distributions reproduce spectra recorded at 34 GHz (Q band) and 9.7 GHz (X band), including subtle variations that were previously difficult to quantify. The X-band spectrum of transferrin shows a characteristic double peak, which has puzzled researchers for decades. We show that the double peak is uniquely related to the term B4-3O4-3(S) in the spin Hamiltonian. Our method is generally applicable in the analysis of spectra that arise from a broad distribution of parameters.

2.
J Theor Biol ; 416: 88-98, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28065783

ABSTRACT

The transferrin (Tf) trafficking pathway is a promising mechanism for use in targeted cancer therapy due to the overexpression of transferrin receptors (TfRs) on cancerous cells. We have previously developed a mathematical model of the Tf/TfR trafficking pathway to improve the efficiency of Tf as a drug carrier. By using diphtheria toxin (DT) as a model toxin, we found that mutating the Tf protein to change its iron release rate improves cellular association and efficacy of the drug. Though this is an improvement upon using wild-type Tf as the targeting ligand, conjugated toxins like DT are unfortunately still highly cytotoxic at off-target sites. In this work, we address this hurdle in cancer research by developing a mathematical model to predict the efficacy and selectivity of Tf conjugates that use an alternative toxin. For this purpose, we have chosen to study a mutant of DT, cross-reacting material 107 (CRM107). First, we developed a mathematical model of the Tf-DT trafficking pathway by extending our Tf/TfR model to include intracellular trafficking via DT and DT receptors. Using this mathematical model, we subsequently investigated the efficacy of several conjugates in cancer cells: DT and CRM107 conjugated to wild-type Tf, as well as to our engineered mutant Tf proteins (K206E/R632A Tf and K206E/R534A Tf). We also investigated the selectivity of mutant Tf-CRM107 against non-neoplastic cells. Through the use of our mathematical model, we predicted that (i) mutant Tf-CRM107 exhibits a greater cytotoxicity than wild-type Tf-CRM107 against cancerous cells, (ii) this improvement was more drastic with CRM107 conjugates than with DT conjugates, and (iii) mutant Tf-CRM107 conjugates were selective against non-neoplastic cells. These predictions were validated with in vitro cytotoxicity experiments, demonstrating that mutant Tf-CRM107 conjugates is indeed a more suitable therapeutic agent. Validation from in vitro experiments also confirmed that such whole-cell kinetic models can be useful in cancer therapeutic design.


Subject(s)
Bacterial Toxins/genetics , Models, Theoretical , Neoplasms/drug therapy , Transferrin/genetics , Antineoplastic Agents/therapeutic use , Bacterial Toxins/therapeutic use , Diphtheria Toxin , Drug Screening Assays, Antitumor , Humans , Mutagenesis, Site-Directed , Mutation , Transferrin/analogs & derivatives , Transferrin/therapeutic use
3.
Biochim Biophys Acta ; 1863(12): 2859-2867, 2016 12.
Article in English | MEDLINE | ID: mdl-27627839

ABSTRACT

In erythroid cells, more than 90% of transferrin-derived iron enters mitochondria where ferrochelatase inserts Fe2+ into protoporphyrin IX. However, the path of iron from endosomes to mitochondrial ferrochelatase remains elusive. The prevailing opinion is that, after its export from endosomes, the redox-active metal spreads into the cytosol and mysteriously finds its way into mitochondria through passive diffusion. In contrast, this study supports the hypothesis that the highly efficient transport of iron toward ferrochelatase in erythroid cells requires a direct interaction between transferrin-endosomes and mitochondria (the "kiss-and-run" hypothesis). Using a novel method (flow sub-cytometry), we analyze lysates of reticulocytes after labeling these organelles with different fluorophores. We have identified a double-labeled population definitively representing endosomes interacting with mitochondria, as demonstrated by confocal microscopy. Moreover, we conclude that this endosome-mitochondrion association is reversible, since a "chase" with unlabeled holotransferrin causes a time-dependent decrease in the size of the double-labeled population. Importantly, the dissociation of endosomes from mitochondria does not occur in the absence of holotransferrin. Additionally, mutated recombinant holotransferrin, that cannot release iron, significantly decreases the uptake of 59Fe by reticulocytes and diminishes 59Fe incorporation into heme. This suggests that endosomes, which are unable to provide iron to mitochondria, cause a "traffic jam" leading to decreased endocytosis of holotransferrin. Altogether, our results suggest that a molecular mechanism exists to coordinate the iron status of endosomal transferrin with its trafficking. Besides its contribution to the field of iron metabolism, this study provides evidence for a new intracellular trafficking pathway of organelles.


Subject(s)
Endosomes/metabolism , Ferrochelatase/metabolism , Iron/metabolism , Mitochondria/metabolism , Protoporphyrins/metabolism , Reticulocytes/metabolism , Transferrin/metabolism , Animals , Biological Transport , Cell Differentiation , Endocytosis/physiology , Fetus , Fluorescent Dyes/chemistry , Heme/metabolism , Humans , Liver/cytology , Liver/metabolism , Mice , Mutation , Primary Cell Culture , Reticulocytes/cytology , Staining and Labeling/methods
4.
J Cell Biol ; 214(7): 831-45, 2016 09 26.
Article in English | MEDLINE | ID: mdl-27646275

ABSTRACT

Transient "kiss and run" interactions between endosomes containing iron-bound transferrin (Tf) and mitochondria have been shown to facilitate direct iron transfer in erythroid cells. In this study, we used superresolution three-dimensional (3D) direct stochastic optical reconstruction microscopy to show that Tf-containing endosomes directly interact with mitochondria in epithelial cells. We used live-cell time-lapse fluorescence microscopy, followed by 3D rendering, object tracking, and a distance transformation algorithm, to track Tf-endosomes and characterize the dynamics of their interactions with mitochondria. Quenching of iron sensor RDA-labeled mitochondria confirmed functional iron transfer by an interacting Tf-endosome. The motility of Tf-endosomes is significantly reduced upon interaction with mitochondria. To further assess the functional role of iron in the ability of Tf-endosomes to interact with mitochondria, we blocked endosomal iron release by using a Tf K206E/K534A mutant. Blocking intraendosomal iron release led to significantly increased motility of Tf-endosomes and increased duration of endosome-mitochondria interactions. Thus, intraendosomal iron regulates the kinetics of the interactions between Tf-containing endosomes and mitochondria in epithelial cells.


Subject(s)
Endosomes/metabolism , Iron/metabolism , Mitochondria/metabolism , Transferrin/metabolism , Animals , Dogs , Humans , Imaging, Three-Dimensional , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Models, Biological , Time-Lapse Imaging
5.
J Biol Inorg Chem ; 20(3): 487-96, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25537134

ABSTRACT

We report 275 GHz EPR spectra of human serum transferrin. At this high microwave frequency the zero-field splitting between the magnetic sublevels of the high-spin [Formula: see text] sites can be accurately determined. We find the zero-field splitting to be a sensitive probe of the structure of the transferrin iron-binding sites. Signals arising from iron bound to the transferrin N-lobe can clearly be distinguished from signals from iron bound to the C-lobe. Moreover, our spectra show that the structure of the iron site in the N-lobe is influenced by the presence and conformation of the C-lobe. The spectra of a series of N-lobe mutants altering the second-shell interaction of Arg124 with the synergistic anion carbonate reflect conformational changes induced at the iron site.


Subject(s)
Electron Spin Resonance Spectroscopy , Ferric Compounds/chemistry , Models, Molecular , Transferrin/chemistry , Binding Sites , Blood Chemical Analysis , Humans , Transferrin/genetics , Transferrin/metabolism
6.
Cell Host Microbe ; 14(5): 522-34, 2013 Nov 13.
Article in English | MEDLINE | ID: mdl-24237698

ABSTRACT

Arenaviruses and hantaviruses cause severe human disease. Little is known regarding host proteins required for their propagation. We identified human proteins that interact with the glycoproteins (GPs) of a prototypic arenavirus and hantavirus and show that the lectin endoplasmic reticulum (ER)-Golgi intermediate compartment 53 kDa protein (ERGIC-53), a cargo receptor required for glycoprotein trafficking within the early exocytic pathway, associates with arenavirus, hantavirus, coronavirus, orthomyxovirus, and filovirus GPs. ERGIC-53 binds to arenavirus GPs through a lectin-independent mechanism, traffics to arenavirus budding sites, and is incorporated into virions. ERGIC-53 is required for arenavirus, coronavirus, and filovirus propagation; in its absence, GP-containing virus particles form but are noninfectious, due in part to their inability to attach to host cells. Thus, we have identified a class of pathogen-derived ERGIC-53 ligands, a lectin-independent basis for their association with ERGIC-53, and a role for ERGIC-53 in the propagation of several highly pathogenic RNA virus families.


Subject(s)
Arenavirus/physiology , Coronavirus/physiology , Filoviridae/physiology , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Virus Assembly , Cell Line , Glycoproteins/metabolism , Humans , Protein Transport , Viral Proteins/metabolism
7.
Biochemistry ; 52(46): 8333-41, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24152109

ABSTRACT

It has been previously suggested that large amounts of oxalate in plasma could play a role in autism by binding to the bilobal iron transport protein transferrin (hTF), thereby interfering with iron metabolism by inhibiting the delivery of iron to cells. By examining the effect of the substitution of oxalate for the physiologically utilized synergistic carbonate anion in each lobe of hTF, we sought to provide a molecular basis for or against such a role. Our work clearly shows both qualitatively (6 M urea gels) and quantitatively (kinetic analysis by stopped-flow spectrofluorimetry) that the presence of oxalate in place of carbonate in each binding site of hTF does indeed greatly interfere with the removal of iron from each lobe (in the absence and presence of the specific hTF receptor). However, we also clearly demonstrate that once the iron is bound within each lobe of hTF, neither anion can displace the other. Additionally, as verified by urea gels and electrospray mass spectrometry, formation of completely homogeneous hTF-anion complexes requires that all iron must first be removed and hTF then reloaded with iron in the presence of either carbonate or oxalate. Significantly, experiments described here show that carbonate is the preferred binding partner; i.e., even if an equal amount of each anion is available during the iron loading process, the hTF-carbonate complex is formed.


Subject(s)
Anemia, Iron-Deficiency/physiopathology , Autistic Disorder/blood , Carbonates/metabolism , Child Development Disorders, Pervasive/blood , Iron/metabolism , Oxalates/blood , Transferrin/chemistry , Transferrin/metabolism , Anemia, Iron-Deficiency/blood , Animals , Cells, Cultured , Cricetinae , Humans , Kinetics
8.
Metallomics ; 5(6): 619-26, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23446908

ABSTRACT

Following an internal contamination event, the transport of actinide (An) and lanthanide (Ln) metal ions through the body is facilitated by endogenous ligands such as the human iron-transport protein transferrin (Tf). The recognition of resulting metallo-transferrin complexes (M2Tf) by the cognate transferrin receptor (TfR) is therefore a critical step for cellular uptake of these metal ions. A high performance liquid chromatography-based method has been used to probe the binding of M2Tf with TfR, yielding a direct measurement of the successive thermodynamic constants that correspond to the dissociation of TfR(M2Tf)2 and TfR(M2Tf) complexes for Fe(3+), Ga(3+), La(3+), Nd(3+), Gd(3+), Yb(3+), Lu(3+), (232)Th(4+), (238)UO2(2+), and (242)Pu(4+). Important features of this method are (i) its ability to distinguish both 1 : 1 and 1 : 2 complexes formed between the receptor and the metal-bound transferrin, and (ii) the requirement for very small amounts of each binding partner (<1 nmol of protein per assay). Consistent with previous reports, the strongest receptor affinity is found for Fe2Tf (Kd1 = 5 nM and Kd2 = 20 nM), while the lowest affinity was measured for Pu2Tf (Kd1 = 0.28 µM and Kd2 = 1.8 µM) binding to the TfR. Other toxic metal ions such as Th(IV) and U(VI), when bound to Tf, are well recognized by the TfR. Under the described experimental conditions, the relative stabilities of TfR:(MxTf)y adducts follow the order Fe(3+) >> Th(4+) ~ UO2(2+) ~ Cm(3+) > Ln(3+) ~ Ga(3+) >>> Yb(3+) ~ Pu(4+). This study substantiates a role for Tf in binding lanthanide fission products and actinides, and transporting them into cells by receptor-mediated endocytosis.


Subject(s)
Actinoid Series Elements/metabolism , Lanthanoid Series Elements/metabolism , Receptors, Transferrin/metabolism , Transferrin/metabolism , Actinoid Series Elements/chemistry , Biological Transport , Chromatography, High Pressure Liquid , Lanthanoid Series Elements/chemistry , Protein Binding , Transferrin/chemistry
9.
J Am Chem Soc ; 135(7): 2676-83, 2013 Feb 20.
Article in English | MEDLINE | ID: mdl-23363005

ABSTRACT

Worldwide stocks of actinides and lanthanide fission products produced through conventional nuclear spent fuel are increasing continuously, resulting in a growing risk of environmental and human exposure to these toxic radioactive metal ions. Understanding the biomolecular pathways involved in mammalian uptake, transport and storage of these f-elements is crucial to the development of new decontamination strategies and could also be beneficial to the design of new containment and separation processes. To start unraveling these pathways, our approach takes advantage of the unique spectroscopic properties of trivalent curium. We clearly show that the human iron transporter transferrin acts as an antenna that sensitizes curium luminescence through intramolecular energy transfer. This behavior has been used to describe the coordination of curium within the two binding sites of the protein and to investigate the recognition of curium-transferrin complexes by the cognate transferrin receptor. In addition to providing the first protein-curium spectroscopic characterization, these studies prove that transferrin receptor-mediated endocytosis is a viable mechanism of intracellular entry for trivalent actinides such as curium and provide a new tool utilizing the specific luminescence of curium for the determination of other biological actinide transport mechanisms.


Subject(s)
Actinoid Series Elements/chemistry , Curium/chemistry , Transferrin/chemistry , Actinoid Series Elements/metabolism , Chromatography, High Pressure Liquid , Coordination Complexes/chemistry , Curium/metabolism , Humans , Luminescence , Thermodynamics , Transferrin/metabolism
10.
Adv Drug Deliv Rev ; 65(8): 1012-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23183585

ABSTRACT

Highly proliferative cells have a dramatically increased need for iron which results in the expression of an increased number of transferrin receptors (TFR). This insight makes the transferrin receptor on these cells an excellent candidate for targeted therapeutics. In this regard, it is critical to understand at a molecular level exactly how the TFR interacts with its ligand, hTF. Understanding of the hTF/TFR pathway could, in theory, maximize the use of this system for development of more effective small molecules or toxin-conjugates to specifically target cancer cells. Many strategies have been attempted with the objective of utilizing the hTF/TFR system to deliver drugs; these include conjugation of a toxin or drug to hTF or direct targeting of the TFR by antibodies. To date, in spite of all of the effort, there is a conspicuous absence of any successful candidate drugs reaching the clinic. We suggest that a lack of quantitative data to determine the basic biochemical properties of the drug carrier and the effects of drug-conjugation on the hTF-TFR interaction may have contributed to the failure to realize the full potential of this system. This review provides some guidelines for developing a more quantitative approach for evaluation of current and future hTF-drug conjugates.


Subject(s)
Drug Carriers/administration & dosage , Receptors, Transferrin/metabolism , Transferrin/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Humans , Receptors, Transferrin/chemistry , Transferrin/chemistry
11.
Curr Top Membr ; 69: 3-35, 2012.
Article in English | MEDLINE | ID: mdl-23046645

ABSTRACT

Essential to iron homeostasis is the transport of iron by the bilobal protein human serum transferrin (hTF). Each lobe (N- and C-lobe) of hTF forms a deep cleft which binds a single Fe(3+). Iron-bearing hTF in the blood binds tightly to the specific transferrin receptor (TFR), a homodimeric transmembrane protein. After undergoing endocytosis, acidification of the endosome initiates the release of Fe(3+) from hTF in a TFR-mediated process. Iron-free hTF remains tightly bound to the TFR at acidic pH; following recycling back to the cell surface, it is released to sequester more iron. Efficient delivery of iron is critically dependent on hTF/TFR interactions. Therefore, identification of the pH-specific contacts between hTF and the TFR is crucial. Recombinant protein production has enabled deconvolution of this complex system. The studies reviewed herein support a model in which pH-induced interrelated events control receptor-stimulated iron release from each lobe of hTF.


Subject(s)
Iron/metabolism , Transferrin/metabolism , Anions/chemistry , Anions/metabolism , Biological Transport , Humans , Hydrogen-Ion Concentration , Kinetics , Protein Binding , Protein Structure, Tertiary , Receptors, Transferrin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transferrin/chemistry , Transferrin/genetics
12.
J Inorg Biochem ; 116: 37-44, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23010327

ABSTRACT

The Fe(3+) binding protein human serum transferrin (hTF) is well known for its role in cellular iron delivery via the transferrin receptor (TFR). A new application is the use of hTF as a therapy and targeted drug delivery system for a number of diseases. Recently, production of hTF in plants has been reported; such systems provide a relatively inexpensive, animal-free (eliminating potential contamination by animal pathogens) method to produce large amounts of recombinant proteins for such biopharmaceutical applications. Specifically, the production of Optiferrin (hTF produced in rice, Oryza sativa, from InVitria) has been shown to yield large amounts of functional protein for use in culture medium for cellular iron delivery to promote growth. In the present work we describe further purification (by gel filtration) and characterization of hTF produced in rice (purified Optiferrin) to determine its suitability in biopharmaceutical applications. The spectral, mass spectrometric, urea gel and kinetic analysis shows that purified Optiferrin is similar to recombinant nonglycosylated N-His tagged hTF expressed by baby hamster kidney cells and/or serum derived glycosylated hTF. Additionally, in a competitive immunoassay, iron-loaded Optiferrin is equivalent to iron-loaded N-His hTF in its ability to bind to the soluble portion of the TFR immobilized in an assay plate. As an essential requirement for any functional hTF, both lobes of purified Optiferrin bind Fe(3+) tightly yet reversibly. Although previously shown to be capable of delivering Fe(3+) to cells, the kinetics of iron release from iron-loaded Optiferrin™/sTFR and iron-loaded N-His hTF/sTFR complexes differ somewhat. We conclude that the purified Optiferrin might be suitable for consideration in biopharmaceutical applications.


Subject(s)
Oryza/genetics , Transferrin/chemistry , Amino Acid Sequence , Chromatography, Gel , Chromatography, High Pressure Liquid , Circular Dichroism , Culture Media , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Mass Spectrometry , Molecular Sequence Data , Peptide Mapping , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Homology, Amino Acid , Transferrin/isolation & purification
13.
Biochemistry ; 51(10): 2113-21, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22356162

ABSTRACT

The recent crystal structure of two monoferric human serum transferrin (Fe(N)hTF) molecules bound to the soluble portion of the homodimeric transferrin receptor (sTFR) has provided new details about this binding interaction that dictates the delivery of iron to cells. Specifically, substantial rearrangements in the homodimer interface of the sTFR occur as a result of the binding of the two Fe(N)hTF molecules. Mutagenesis of selected residues in the sTFR highlighted in the structure was undertaken to evaluate the effect on function. Elimination of Ca(2+) binding in the sTFR by mutating two of four coordinating residues ([E465A,E468A]) results in low production of an unstable and aggregated sTFR. Mutagenesis of two histidines ([H475A,H684A]) at the dimer interface had little effect on the kinetics of release of iron at pH 5.6 from either lobe, reflecting the inaccessibility of this cluster to solvent. Creation of an H318A sTFR mutant allows assignment of a small pH-dependent initial decrease in the magnitude of the fluorescence signal to His318. Removal of the four C-terminal residues of the sTFR, Asp757-Asn758-Glu759-Phe760, eliminates pH-stimulated release of iron from the C-lobe of the Fe(2)hTF/sTFR Δ757-760 complex. The inability of this sTFR mutant to bind and stabilize protonated hTF His349 (a pH-inducible switch) in the C-lobe of hTF accounts for the loss. Collectively, these studies support a model in which a series of pH-induced events involving both TFR residue His318 and hTF residue His349 occurs to promote receptor-stimulated release of iron from the C-lobe of hTF.


Subject(s)
Receptors, Transferrin/chemistry , Receptors, Transferrin/genetics , Transferrin/chemistry , Binding Sites/genetics , Calcium/metabolism , Dimerization , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Iron/metabolism , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Receptors, Transferrin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transferrin/genetics , Transferrin/metabolism
14.
Nature ; 483(7387): 53-8, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327295

ABSTRACT

Neisseria are obligate human pathogens causing bacterial meningitis, septicaemia and gonorrhoea. Neisseria require iron for survival and can extract it directly from human transferrin for transport across the outer membrane. The transport system consists of TbpA, an integral outer membrane protein, and TbpB, a co-receptor attached to the cell surface; both proteins are potentially important vaccine and therapeutic targets. Two key questions driving Neisseria research are how human transferrin is specifically targeted, and how the bacteria liberate iron from transferrin at neutral pH. To address these questions, we solved crystal structures of the TbpA-transferrin complex and of the corresponding co-receptor TbpB. We characterized the TbpB-transferrin complex by small-angle X-ray scattering and the TbpA-TbpB-transferrin complex by electron microscopy. Our studies provide a rational basis for the specificity of TbpA for human transferrin, show how TbpA promotes iron release from transferrin, and elucidate how TbpB facilitates this process.


Subject(s)
Bacterial Proteins/chemistry , Iron/metabolism , Neisseria/metabolism , Transferrin-Binding Protein A/chemistry , Transferrin-Binding Protein A/metabolism , Transferrin-Binding Protein B/chemistry , Transferrin-Binding Protein B/metabolism , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Binding Sites , Biological Transport , Cattle , Crystallography, X-Ray , Humans , Mice , Models, Molecular , Molecular Dynamics Simulation , Neisseria/pathogenicity , Protein Conformation , Scattering, Small Angle , Species Specificity , Structure-Activity Relationship , Transferrin/chemistry , Transferrin/metabolism , Transferrin/ultrastructure , Transferrin-Binding Protein A/ultrastructure , Transferrin-Binding Protein B/ultrastructure , X-Ray Diffraction
15.
Biochim Biophys Acta ; 1820(3): 161-87, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21856378

ABSTRACT

BACKGROUND: Not long after the Big Bang, iron began to play a central role in the Universe and soon became mired in the tangle of biochemistry that is the prima essentia of life. Since life's addiction to iron transcends the oxygenation of the Earth's atmosphere, living things must be protected from the potentially dangerous mix of iron and oxygen. The human being possesses grams of this potentially toxic transition metal, which is shuttling through his oxygen-rich humor. Since long before the birth of modern medicine, the blood-vibrant red from a massive abundance of hemoglobin iron-has been a focus for health experts. SCOPE OF REVIEW: We describe the current understanding of iron metabolism, highlight the many important discoveries that accreted this knowledge, and describe the perils of dysfunctional iron handling. GENERAL SIGNIFICANCE: Isaac Newton famously penned, "If I have seen further than others, it is by standing upon the shoulders of giants". We hope that this review will inspire future scientists to develop intellectual pursuits by understanding the research and ideas from many remarkable thinkers of the past. MAJOR CONCLUSIONS: The history of iron research is a long, rich story with early beginnings, and is far from being finished. This article is part of a Special Issue entitled Transferrins: Molecular mechanisms of iron transport and disorders.


Subject(s)
Iron Metabolism Disorders , Iron/metabolism , Transferrins/metabolism , Animals , Biological Transport , Erythrocytes/cytology , Erythrocytes/metabolism , Health , Hemoglobins/metabolism , History, 17th Century , History, 18th Century , History, 19th Century , History, 20th Century , History, 21st Century , Humans , Iron/blood , Iron/history , Iron Metabolism Disorders/history , Iron Metabolism Disorders/metabolism , Macrophages/metabolism , Oxygen/metabolism , Transferrins/chemistry
16.
Biochim Biophys Acta ; 1820(3): 326-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21699959

ABSTRACT

BACKGROUND: Human serum transferrin (hTF) is a bilobal glycoprotein that reversibly binds Fe(3+) and delivers it to cells by the process of receptor-mediated endocytosis. Despite decades of research, the precise events resulting in iron release from each lobe of hTF within the endosome have not been fully delineated. SCOPE OF REVIEW: We provide an overview of the kinetics of iron release from hTF±the transferrin receptor (TFR) at endosomal pH (5.6). A critical evaluation of the array of biophysical techniques used to determine accurate rate constants is provided. GENERAL SIGNIFICANCE: Delivery of Fe(3+)to actively dividing cells by hTF is essential; too much or too little Fe(3+) directly impacts the well-being of an individual. Because the interaction of hTF with the TFR controls iron distribution in the body, an understanding of this process at the molecular level is essential. MAJOR CONCLUSIONS: Not only does TFR direct the delivery of iron to the cell through the binding of hTF, kinetic data demonstrate that it also modulates iron release from the N- and C-lobes of hTF. Specifically, the TFR balances the rate of iron release from each lobe, resulting in efficient Fe(3+) release within a physiologically relevant time frame. This article is part of a Special Issue entitled Molecular Mechanisms of Iron Transport and Disorders.


Subject(s)
Iron/metabolism , Receptors, Transferrin/metabolism , Transferrin/metabolism , Biological Transport , Endocytosis , Endosomes/metabolism , Humans , Hydrogen-Ion Concentration , Protein Conformation , Receptors, Transferrin/chemistry
17.
Biochemistry ; 51(2): 686-94, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22191507

ABSTRACT

Efficient delivery of iron is critically dependent on the binding of diferric human serum transferrin (hTF) to its specific receptor (TFR) on the surface of actively dividing cells. Internalization of the complex into an endosome precedes iron removal. The return of hTF to the blood to continue the iron delivery cycle relies on the maintenance of the interaction between apohTF and the TFR after exposure to endosomal pH (≤6.0). Identification of the specific residues accounting for the pH-sensitive nanomolar affinity with which hTF binds to TFR throughout the cycle is important to fully understand the iron delivery process. Alanine substitution of 11 charged hTF residues identified by available structures and modeling studies allowed evaluation of the role of each in (1) binding of hTF to the TFR and (2) TFR-mediated iron release. Six hTF mutants (R50A, R352A, D356A, E357A, E367A, and K511A) competed poorly with biotinylated diferric hTF for binding to TFR. In particular, we show that Asp356 in the C-lobe of hTF is essential to the formation of a stable hTF-TFR complex: mutation of Asp356 in the monoferric C-lobe hTF background prevented the formation of the stoichiometric 2:2 (hTF:TFR monomer) complex. Moreover, mutation of three residues (Asp356, Glu367, and Lys511), whether in the diferric or monoferric C-lobe hTF, significantly affected iron release when in complex with the TFR. Thus, mutagenesis of charged hTF residues has allowed identification of a number of residues that are critical to formation of and release of iron from the hTF-TFR complex.


Subject(s)
Iron/metabolism , Receptors, Transferrin/metabolism , Transferrin/chemistry , Transferrin/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Mutation , Protein Binding , Protein Structure, Tertiary , Receptors, Transferrin/chemistry , Solubility , Transferrin/genetics
18.
Proc Natl Acad Sci U S A ; 108(32): 13089-94, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788477

ABSTRACT

Delivery of iron to cells requires binding of two iron-containing human transferrin (hTF) molecules to the specific homodimeric transferrin receptor (TFR) on the cell surface. Through receptor-mediated endocytosis involving lower pH, salt, and an unidentified chelator, iron is rapidly released from hTF within the endosome. The crystal structure of a monoferric N-lobe hTF/TFR complex (3.22-Å resolution) features two binding motifs in the N lobe and one in the C lobe of hTF. Binding of Fe(N)hTF induces global and site-specific conformational changes within the TFR ectodomain. Specifically, movements at the TFR dimer interface appear to prime the TFR to undergo pH-induced movements that alter the hTF/TFR interaction. Iron release from each lobe then occurs by distinctly different mechanisms: Binding of His349 to the TFR (strengthened by protonation at low pH) controls iron release from the C lobe, whereas displacement of one N-lobe binding motif, in concert with the action of the dilysine trigger, elicits iron release from the N lobe. One binding motif in each lobe remains attached to the same α-helix in the TFR throughout the endocytic cycle. Collectively, the structure elucidates how the TFR accelerates iron release from the C lobe, slows it from the N lobe, and stabilizes binding of apohTF for return to the cell surface. Importantly, this structure provides new targets for mutagenesis studies to further understand and define this system.


Subject(s)
Endosomes/metabolism , Iron/metabolism , Receptors, Transferrin/metabolism , Transferrin/metabolism , Amino Acid Motifs , Binding Sites , Endocytosis , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Receptors, Transferrin/chemistry , Transferrin/chemistry
19.
J Biol Inorg Chem ; 15(8): 1341-52, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20711621

ABSTRACT

His349 in human transferrin (hTF) is a residue critical to transferrin receptor (TFR)-stimulated iron release from the C-lobe. To evaluate the importance of His349 on the TFR interaction, it was replaced by alanine, aspartate, lysine, leucine, tryptophan, and tyrosine in a monoferric C-lobe hTF construct (Fe(C)hTF). Using a stopped-flow spectrofluorimeter, we determined rate processes assigned to iron release and conformational events (in the presence and in the absence of the TFR). Significantly, all mutant/TFR complexes feature dampened iron release rates. The critical contribution of His349 is most convincingly revealed by analysis of the kinetics as a function of pH (5.6-6.2). The Fe(C)hTF/TFR complex titrates with a pK(a) of approximately 5.9. By contrast, the H349A mutant/TFR complex releases iron at higher pH with a profile that is almost the inverse of that of the control complex. At the putative endosomal pH of 5.6 (in the presence of salt and chelator), iron is released from the H349W mutant/TFR and H349Y mutant/TFR complexes with a single rate constant similar to the iron release rate constant for the control; this suggests that these substitutions bypass the required pH-induced conformational change allowing the C-lobe to directly interact with the TFR to release iron. The H349K mutant proves that although the positive charge is crucial to complete iron release, the geometry at this position is also critical. The H349D mutant shows that a negative charge precludes complete iron release at pH 5.6 both in the presence and in the absence of the TFR. Thus, histidine uniquely drives the pH-induced conformational change in the C-lobe required for TFR interaction, which in turn promotes iron release.


Subject(s)
Histidine/chemistry , Iron/chemistry , Receptors, Transferrin/chemistry , Transferrin/chemistry , Histidine/genetics , Histidine/metabolism , Humans , Hydrogen-Ion Concentration , Iron/metabolism , Kinetics , Protein Binding , Protein Conformation , Receptors, Transferrin/metabolism , Transferrin/genetics , Transferrin/metabolism
20.
J Biol Chem ; 285(36): 27609-13, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20615872

ABSTRACT

The innate immune system in humans consists of both cellular and humoral components that collaborate to eradicate invading bacteria from the body. Here, we discover that the gram-positive bacterium Bacillus anthracis, the causative agent of anthrax, does not grow in human serum. Fractionation of serum by gel filtration chromatography led to the identification of human transferrin as the inhibiting factor. Purified transferrin blocks growth of both the fully virulent encapsulated B. anthracis Ames and the non-encapsulated Sterne strain. Growth inhibition was also observed in serum of wild-type mice but not of hypotransferrinemic mice that only have approximately 1% circulating transferrin levels. We were able to definitely assign the bacteriostatic activity of transferrin to its iron-binding function: neither iron-saturated transferrin nor a recombinant transferrin mutant unable to bind iron could inhibit growth of B. anthracis. Additional iron could restore bacterial growth in human serum. The observation that other important gram-positive pathogens are not inhibited by transferrin suggests they have evolved effective mechanisms to circumvent serum iron deprivation. These findings provide a better understanding of human host defense mechanisms against anthrax and provide a mechanistic basis for the antimicrobial activity of human transferrin.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Serum/immunology , Serum/microbiology , Transferrin/pharmacology , Animals , Bacillus anthracis/growth & development , Female , Humans , Immunity, Innate , Iron/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...