Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Phys Chem Chem Phys ; 21(31): 16937-16948, 2019 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-31339131

RESUMO

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.
Nature ; 483(7387): 53-8, 2012 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-22327295

RESUMO

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.


Assuntos
Proteínas de Bactérias/química , Ferro/metabolismo , Neisseria/metabolismo , Proteína A de Ligação a Transferrina/química , Proteína A de Ligação a Transferrina/metabolismo , Proteína B de Ligação a Transferrina/química , Proteína B de Ligação a Transferrina/metabolismo , Animais , Apoproteínas/química , Apoproteínas/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Sítios de Ligação , Transporte Biológico , Bovinos , Cristalografia por Raios X , Humanos , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Neisseria/patogenicidade , Conformação Proteica , Espalhamento a Baixo Ângulo , Especificidade da Espécie , Relação Estrutura-Atividade , Transferrina/química , Transferrina/metabolismo , Transferrina/ultraestrutura , Proteína A de Ligação a Transferrina/ultraestrutura , Proteína B de Ligação a Transferrina/ultraestrutura , Difração de Raios X
3.
Biochim Biophys Acta ; 1863(12): 2859-2867, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27627839

RESUMO

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.


Assuntos
Endossomos/metabolismo , Ferroquelatase/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Protoporfirinas/metabolismo , Reticulócitos/metabolismo , Transferrina/metabolismo , Animais , Transporte Biológico , Diferenciação Celular , Endocitose/fisiologia , Feto , Corantes Fluorescentes/química , Heme/metabolismo , Humanos , Fígado/citologia , Fígado/metabolismo , Camundongos , Mutação , Cultura Primária de Células , Reticulócitos/citologia , Coloração e Rotulagem/métodos
4.
J Theor Biol ; 416: 88-98, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28065783

RESUMO

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.


Assuntos
Toxinas Bacterianas/genética , Modelos Teóricos , Neoplasias/tratamento farmacológico , Transferrina/genética , Antineoplásicos/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Toxina Diftérica , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Mutagênese Sítio-Dirigida , Mutação , Transferrina/análogos & derivados , Transferrina/uso terapêutico
5.
J Biol Inorg Chem ; 20(3): 487-96, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25537134

RESUMO

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.


Assuntos
Espectroscopia de Ressonância de Spin Eletrônica , Compostos Férricos/química , Modelos Moleculares , Transferrina/química , Sítios de Ligação , Análise Química do Sangue , Humanos , Transferrina/genética , Transferrina/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(32): 13089-94, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21788477

RESUMO

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.


Assuntos
Endossomos/metabolismo , Ferro/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Endocitose , Humanos , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Ligação Proteica , Estrutura Terciária de Proteína , Receptores da Transferrina/química , Transferrina/química
7.
Biochemistry ; 52(46): 8333-41, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24152109

RESUMO

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.


Assuntos
Anemia Ferropriva/fisiopatologia , Transtorno Autístico/sangue , Carbonatos/metabolismo , Transtornos Globais do Desenvolvimento Infantil/sangue , Ferro/metabolismo , Oxalatos/sangue , Transferrina/química , Transferrina/metabolismo , Anemia Ferropriva/sangue , Animais , Células Cultivadas , Cricetinae , Humanos , Cinética
8.
Biochim Biophys Acta ; 1820(3): 161-87, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21856378

RESUMO

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.


Assuntos
Distúrbios do Metabolismo do Ferro , Ferro/metabolismo , Transferrinas/metabolismo , Animais , Transporte Biológico , Eritrócitos/citologia , Eritrócitos/metabolismo , Saúde , Hemoglobinas/metabolismo , História do Século XVII , História do Século XVIII , História do Século XIX , História do Século XX , História do Século XXI , Humanos , Ferro/sangue , Ferro/história , Distúrbios do Metabolismo do Ferro/história , Distúrbios do Metabolismo do Ferro/metabolismo , Macrófagos/metabolismo , Oxigênio/metabolismo , Transferrinas/química
9.
Biochim Biophys Acta ; 1820(3): 326-33, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21699959

RESUMO

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.


Assuntos
Ferro/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Transporte Biológico , Endocitose , Endossomos/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Conformação Proteica , Receptores da Transferrina/química
10.
J Am Chem Soc ; 135(7): 2676-83, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23363005

RESUMO

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.


Assuntos
Elementos da Série Actinoide/química , Cúrio/química , Transferrina/química , Elementos da Série Actinoide/metabolismo , Cromatografia Líquida de Alta Pressão , Complexos de Coordenação/química , Cúrio/metabolismo , Humanos , Luminescência , Termodinâmica , Transferrina/metabolismo
11.
Proc Natl Acad Sci U S A ; 107(18): 8123-8, 2010 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-20404192

RESUMO

The primary route of iron acquisition in vertebrates is the transferrin receptor (TfR) mediated endocytotic pathway, which provides cellular entry to the metal transporter serum transferrin (Tf). Despite extensive research efforts, complete understanding of Tf-TfR interaction mechanism is still lacking owing to the complexity of this system. Electrospray ionization mass spectrometry (ESI MS) is used in this study to monitor the protein/receptor interaction and demonstrate the ability of metal-free Tf to associate with TfR at neutral pH. A set of Tf variants is used in a series of competition and displacement experiments to bracket TfR affinity of apo-Tf at neutral pH (0.2-0.6 microM). Consistent with current models of endosomal iron release from Tf, acidification of the protein solution results in a dramatic change of binding preferences, with apo-Tf becoming a preferred receptor binder. Contrary to the current models implying that the apo-Tf/TfR complex dissociates almost immediately upon exposure to the neutral environment at the cell surface, our data indicate that this complex remains intact. Iron-loaded Tf displaces apo-Tf from TfR, making it available for the next cycle of iron binding, transport and delivery to tissues. However, apo-Tf may still interfere with the cellular uptake of engineered Tf molecules whose TfR affinity is affected by various modifications (e.g., conjugation to cytotoxic molecules). This work also highlights the great potential of ESI MS as a tool capable of providing precise details of complex protein-receptor interactions under conditions that closely mimic the environment in which these encounters occur in physiological systems.


Assuntos
Receptores da Transferrina/análise , Espectrometria de Massas por Ionização por Electrospray/métodos , Transferrina/análise , Humanos , Concentração de Íons de Hidrogênio , Ferro/metabolismo , Ligação Proteica , Receptores da Transferrina/metabolismo , Transferrina/metabolismo
12.
Biochemistry ; 51(10): 2113-21, 2012 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-22356162

RESUMO

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.


Assuntos
Receptores da Transferrina/química , Receptores da Transferrina/genética , Transferrina/química , Sítios de Ligação/genética , Cálcio/metabolismo , Dimerização , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Ferro/metabolismo , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Receptores da Transferrina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transferrina/genética , Transferrina/metabolismo
13.
Biochemistry ; 51(2): 686-94, 2012 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-22191507

RESUMO

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.


Assuntos
Ferro/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/química , Transferrina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Modelos Moleculares , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Receptores da Transferrina/química , Solubilidade , Transferrina/genética
14.
Curr Top Membr ; 69: 3-35, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23046645

RESUMO

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.


Assuntos
Ferro/metabolismo , Transferrina/metabolismo , Ânions/química , Ânions/metabolismo , Transporte Biológico , Humanos , Concentração de Íons de Hidrogênio , Cinética , Ligação Proteica , Estrutura Terciária de Proteína , Receptores da Transferrina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transferrina/química , Transferrina/genética
15.
J Biol Chem ; 285(36): 27609-13, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20615872

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Bacillus anthracis/efeitos dos fármacos , Soro/imunologia , Soro/microbiologia , Transferrina/farmacologia , Animais , Bacillus anthracis/crescimento & desenvolvimento , Feminino , Humanos , Imunidade Inata , Ferro/metabolismo , Camundongos
16.
Biochemistry ; 49(19): 4200-7, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20397659

RESUMO

Human serum transferrin (hTF) binds two ferric iron ions which are delivered to cells in a transferrin receptor (TFR) mediated process. Critical to the delivery of iron to cells is the binding of hTF to the TFR and the efficient release of iron orchestrated by the interaction. Within the endosome, iron release from hTF is also aided by lower pH, the presence of anions, and a chelator yet to be identified. We have recently shown that three of the four residues comprising a loop in the N-lobe (Pro142, Lys144, and Pro145) are critical to the high-affinity interaction of hTF with the TFR. In contrast, Arg143 in this loop does not participate in the binding isotherm. In the current study, the kinetics of iron release from alanine mutants of each of these four residues (placed into both diferric and monoferric N-lobe backgrounds) have been determined +/- the TFR. The R143A mutation greatly retards the rate of iron release from the N-lobe in the absence of the TFR but has considerably less of an effect in its presence. Our data definitively show that Arg143 serves as a kinetically significant anion binding (KISAB) site that is, by definition, sensitive to salt concentration and critical to the conformational change necessary to induce iron release from the N-lobe of hTF (in the absence of the TFR). This is the first identification of an authentic KISAB site in the N-lobe of hTF. The effect of the single R143A mutation on the kinetic profile of iron release provides a dramatic illustration of the dynamic nature of hTF.


Assuntos
Transferrina/química , Animais , Ânions/química , Sítios de Ligação , Células Cultivadas , Cricetinae , Cristalografia por Raios X , Humanos , Cinética , Mutação , Conformação Proteica , Transferrina/metabolismo
17.
Biochim Biophys Acta ; 1794(3): 532-40, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19103311

RESUMO

Human serum transferrin (hTF) is a bilobal glycoprotein that transports iron to cells. At neutral pH, diferric hTF binds with nM affinity to the transferrin receptor (TFR) on the cell surface. The complex is taken into the cell where, at the acidic pH of the endosome ( approximately pH 5.6), iron is released. Since iron coordination strongly quenches the intrinsic tryptophan fluorescence of hTF, the increase in the fluorescent signal reports the rate constant(s) of iron release. At pH 5.6, the TFR considerably enhances iron release from the C-lobe (with little effect on iron release from the N-lobe). The recombinant soluble TFR is a dimer with 11 tryptophan residues per monomer. In the hTF/TFR complex these residues could contribute to and compromise the readout ascribed to iron release from hTF. We report that compared to Fe(C) hTF alone, the increase in the fluorescent signal from the preformed complex of Fe(C) hTF and the TFR at pH 5.6 is significantly quenched (75%). To dissect the contributions of hTF and the TFR to the change in fluorescence, 5-hydroxytryptophan was incorporated into each using our mammalian expression system. Selective excitation of the samples at 280 or 315 nm shows that the TFR contributes little or nothing to the increase in fluorescence when ferric iron is released from Fe(C) hTF. Quantum yield determinations of TFR, Fe(C) hTF and the Fe(C) hTF/TFR complex strongly support our interpretation of the kinetic data.


Assuntos
5-Hidroxitriptofano/metabolismo , Ferro/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Dimerização , Humanos , Concentração de Íons de Hidrogênio , Cinética , Teoria Quântica , Receptores da Transferrina/química , Espectrometria de Fluorescência , Transferrina/química
18.
J Biol Inorg Chem ; 15(8): 1341-52, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20711621

RESUMO

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.


Assuntos
Histidina/química , Ferro/química , Receptores da Transferrina/química , Transferrina/química , Histidina/genética , Histidina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Ferro/metabolismo , Cinética , Ligação Proteica , Conformação Proteica , Receptores da Transferrina/metabolismo , Transferrina/genética , Transferrina/metabolismo
19.
Protein Expr Purif ; 72(1): 32-41, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20064616

RESUMO

Essential to iron transport and delivery, human serum transferrin (hTF) is a bilobal glycoprotein capable of reversibly binding one ferric ion in each lobe (the N- and C-lobes). A complete description of iron release from hTF, as well as insight into the physiological significance of the bilobal structure, demands characterization of the isolated lobes. Although production of large amounts of isolated N-lobe and full-length hTF has been well documented, attempts to produce the C-lobe (by recombinant and/or proteolytic approaches) have met with more limited success. Our new strategy involves replacing the hepta-peptide, PEAPTDE (comprising the bridge between the lobes) with the sequence ENLYFQ/G in a His-tagged non-glycosylated monoferric hTF construct, designated Fe(C)hTF. The new bridge sequence of this construct, designated Fe(C)TEV hTF, is readily cleaved by the tobacco etch virus (TEV) protease yielding non-glycosylated C-lobe. Following nickel column chromatography (to remove the N-lobe and the TEV protease which are both His tagged), the homogeneity of the C-lobe has been confirmed by mass spectroscopy. Differing reactivity with a monoclonal antibody specific to the C-lobe indicates that introduction of the TEV cleavage site into the bridge alters its conformation. The spectral and kinetic properties of the isolated C-lobe differ significantly from those of the isolated N-lobe.


Assuntos
Endopeptidases/metabolismo , Ferro/metabolismo , Transferrina/química , Transferrina/genética , Sequência de Aminoácidos , Expressão Gênica , Humanos , Ferro/química , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Análise Espectral , Transferrina/isolamento & purificação , Transferrina/metabolismo
20.
Biochemistry ; 48(13): 2858-67, 2009 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-19281173

RESUMO

Human serum transferrin (hTF), with two Fe3+ binding lobes, transports iron into cells. Diferric hTF preferentially binds to a specific receptor (TFR) on the surface of cells, and the complex undergoes clathrin dependent receptor-mediated endocytosis. The clathrin-coated vesicle fuses with an endosome where the pH is lowered, facilitating iron release from hTF. On a biologically relevant time scale (2-3 min), the factors critical to iron release include pH, anions, a chelator, and the interaction of hTF with the TFR. Previous work, in which the increase in the intrinsic fluorescence signal was used to monitor iron release from the hTF/TFR complex, established that the TFR significantly enhances the rate of iron release from the C-lobe of hTF. In the current study, the role of the five C-lobe Trp residues in reporting the fluorescence change has been evaluated (+/-sTFR). Only four of the five recombinant Trp --> Phe mutants produced well. A single slow rate constant for iron release is found for the monoferric C-lobe (FeC hTF) and the four Trp mutants in the FeC hTF background. The three Trp residues equivalent to those in the N-lobe differed from the N-lobe and each other in their contributions to the fluorescent signal. Two rate constants are observed for the FeC hTF control and the four Trp mutants in complex with the TFR: k(obsC1) reports conformational changes in the C-lobe initiated by the TFR, and k(obsC2) is ascribed to iron release. Excitation at 295 nm (Trp only) and at 280 nm (Trp and Tyr) reveals interesting and significant differences in the rate constants for the complex.


Assuntos
Ferro/metabolismo , Transferrina/química , Transferrina/metabolismo , Triptofano/metabolismo , Absorção , Cristalografia por Raios X , Fluorescência , Humanos , Cinética , Mutação Puntual/genética , Estrutura Secundária de Proteína , Receptores da Transferrina/metabolismo , Soro , Espectrometria de Fluorescência , Espectrofotometria Ultravioleta , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA