Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Biochemistry ; 61(19): 2106-2117, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36099002

RESUMO

Ferritins are highly conserved supramolecular protein nanostructures that play a key role in iron homeostasis. Thousands of iron atoms can be stored inside their hollow cavity as a hydrated ferric oxyhydroxide mineral. Although phosphate associates with the ferritin iron nanoparticles, the effect of physiological concentrations on the kinetics, structure, and reactivity of ferritin iron cores has not yet been explored. Here, the iron loading and mobilization kinetics were studied in the presence of 1-10 mM phosphate using homopolymer and heteropolymer ferritins having different H to L subunit ratios. In the absence of ferritin, phosphate enhances the rate of ferrous ion oxidation and forms large and soluble polymeric Fe(III)-phosphate species. In the presence of phosphate, Fe(II) oxidation and core formation in ferritin is significantly accelerated with oxidation rates several-fold higher than with phosphate alone. High-angle annular dark-field scanning transmission electron microscopy measurements revealed a strong phosphate effect on both the size and morphology of the iron mineral in H-rich (but not L-rich) ferritins. While iron nanoparticles in L-rich ferritins have spherical shape in the absence and presence of phosphate, iron nanoparticles in H-rich ferritins change from irregular shapes in the absence of phosphate to spherical particles in the presence of phosphate with larger size distribution and smaller particle size. In the presence of phosphate, the kinetics of iron-reductive mobilization from ferritin releases twice as much iron than in its absence. Altogether, our results demonstrate an important role for phosphate, and the ferritin H and L subunit composition toward the kinetics of iron oxidation and removal from ferritin, as well as the structure and reactivity of the iron mineral, and may have an important implication on ferritin iron management in vivo.


Assuntos
Ferritinas , Ferro , Apoferritinas/metabolismo , Compostos Férricos/química , Ferritinas/química , Compostos Ferrosos/metabolismo , Humanos , Ferro/química , Cinética , Fosfatos/metabolismo
2.
Int J Mol Sci ; 23(11)2022 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-35682778

RESUMO

Most in vitro iron mobilization studies from ferritin have been performed in aqueous buffered solutions using a variety of reducing substances. The kinetics of iron mobilization from ferritin in a medium that resembles the complex milieu of cells could dramatically differ from those in aqueous solutions, and to our knowledge, no such studies have been performed. Here, we have studied the kinetics of iron release from ferritin in fresh yeast cell lysates and examined the effect of cellular metabolites on this process. Our results show that iron release from ferritin in buffer is extremely slow compared to cell lysate under identical experimental conditions, suggesting that certain cellular metabolites present in yeast cell lysate facilitate the reductive release of ferric iron from the ferritin core. Using filtration membranes with different molecular weight cut-offs (3, 10, 30, 50, and 100 kDa), we demonstrate that a cellular component >50 kDa is implicated in the reductive release of iron. When the cell lysate was washed three times with buffer, or when NADPH was omitted from the solution, a dramatic decrease in iron mobilization rates was observed. The addition of physiological concentrations of free flavins, such as FMN, FAD, and riboflavin showed about a two-fold increase in the amount of released iron. Notably, all iron release kinetics occurred while the solution oxygen level was still high. Altogether, our results indicate that in addition to ferritin proteolysis, there exists an auxiliary iron reductive mechanism that involves long-range electron transfer reactions facilitated by the ferritin shell. The physiological implications of such iron reductive mechanisms are discussed.


Assuntos
Ferritinas , Ferro , Transporte de Elétrons , Ferritinas/metabolismo , Ferro/metabolismo , Cinética , Riboflavina/metabolismo , Saccharomyces cerevisiae/metabolismo
3.
Biochemistry ; 59(29): 2707-2717, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32608971

RESUMO

Ferritinophagy is a ferritin autophagic degradation process mediated by the selective nuclear receptor coactivator-4 (NCOA4). NCOA4 binds to ferritin and delivers it to nascent autophagosomes, which then merge with the lysosomes for ferritin degradation and iron release. Earlier studies have demonstrated a specific association of NCOA4 with ferritin H-subunits, but not L-subunits. However, neither the thermodynamics of this interaction nor the effect of NCOA4 on iron oxidation, iron mineral core formation, or iron mobilization in ferritin has been explored. Using isothermal titration calorimetry, light absorption spectroscopy, and a soluble fragment (residues 383-522) of human NCOA4 expressed in Escherichia coli, we show that the NCOA4 fragment specifically binds H-rich ferritins with a binding stoichiometry of ∼8 NCOA4 molecules per ferritin shell, and Kd values of ∼0.4 and ∼2 µM for homopolymer H-chain ferritin and heteropolymer H-rich ferritin, respectively. The binding reaction was both enthalpically and entropically favored. Whereas the iron oxidation kinetics were not affected by the presence of NCOA4, iron mobilization from ferritin by two different reducing agents (FMN/NADH and sodium dithionite) showed a strong inhibitory effect that was dependent on the concentration of NCOA4 present in solution. Our results suggest that the binding of NCOA4 to ferritin may interfere in the electron transfer pathway through the ferritin shell and may have important biological implications on cellular iron homeostasis.


Assuntos
Apoferritinas/metabolismo , Ferritinas/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Oxirredutases/metabolismo , Apoferritinas/química , Sítios de Ligação , Ferritinas/química , Humanos , Cinética , Coativadores de Receptor Nuclear/química , Oxirredutases/química , Ligação Proteica , Mapas de Interação de Proteínas , Termodinâmica
4.
Biochemistry ; 56(30): 3900-3912, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28636371

RESUMO

In animals, the iron storage and detoxification protein, ferritin, is composed of two functionally and genetically distinct subunit types, H (heavy) and L (light), which co-assemble in various ratios with tissue specific distributions to form shell-like protein structures of 24 subunits within which a mineralized iron core is stored. The H-subunit possesses a ferroxidase center (FC) that catalyzes Fe(II) oxidation, whereas the L-subunit does not. To assess the role of the L-subunit in iron oxidation and core formation, two human recombinant heteropolymeric ferritins, designated H-rich and L-rich with ratios of ∼20H:4L and ∼22L:2H, respectively, were employed and compared to the human homopolymeric H-subunit ferritin (HuHF). These heteropolymeric ferritins have a composition similar to the composition of those found in hearts and brains (i.e., H-rich) and in livers and spleens (i.e., L-rich). As for HuHF, iron oxidation in H-rich ferritin was found to proceed with a 2:1 Fe(II):O2 stoichiometry at an iron level of 2 Fe(II) atoms/H-subunit with the generation of H2O2. The H2O2 reacted with additional Fe(II) in a 2:1 Fe(II):H2O2 ratio, thus avoiding the production of hydroxyl radical. A µ-1,2-peroxo-diFe(III) intermediate was observed at the FC of H-rich ferritin as for HuHF. Importantly, the H-rich protein regenerated full ferroxidase activity more rapidly than HuHF did and additionally formed larger iron cores, indicating dual roles for the L-subunit in facilitating iron turnover at the FC and in mineralization of the core. The L-rich ferritin, while also facilitating iron oxidation at the FC, additionally promoted oxidation at the mineral surface once the iron binding capacity of the FC was exceeded.


Assuntos
Apoferritinas/metabolismo , Coenzimas/metabolismo , Ferritinas/metabolismo , Heme/metabolismo , Ferro/metabolismo , Apoferritinas/química , Apoferritinas/genética , Domínio Catalítico , Coenzimas/química , Eletroforese Capilar , Ferritinas/química , Ferritinas/genética , Heme/química , Holoenzimas/química , Holoenzimas/genética , Holoenzimas/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Ferro/química , Cinética , Oxirredução , Oxirredutases/química , Oxirredutases/genética , Oxirredutases/metabolismo , Multimerização Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
5.
Biochim Biophys Acta ; 1860(5): 879-891, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26569121

RESUMO

BACKGROUND: The binding of metal ions to proteins is a crucial process required for their catalytic activity, structural stability and/or functional regulation. Isothermal titration calorimetry provides a wealth of fundamental information which when combined with structural data allow for a much deeper understanding of the underlying molecular mechanism. SCOPE OF REVIEW: A rigorous understanding of any molecular interaction requires in part an in-depth quantification of its thermodynamic properties. Here, we provide an overview of recent studies that have used ITC to quantify the interaction of essential first row transition metals with relevant proteins and highlight major findings from these thermodynamic studies. GENERAL SIGNIFICANCE: The thermodynamic characterization of metal ion-protein interactions is one important step to understanding the role that metal ions play in living systems. Such characterization has important implications not only to elucidating proteins' structure-function relationships and biological properties but also in the biotechnology sector, medicine and drug design particularly since a number of metal ions are involved in several neurodegenerative diseases. MAJOR CONCLUSIONS: Isothermal titration calorimetry measurements can provide complete thermodynamic profiles of any molecular interaction through the simultaneous determination of the reaction binding stoichiometry, binding affinity as well as the enthalpic and entropic contributions to the free energy change thus enabling a more in-depth understanding of the nature of these interactions.


Assuntos
Adenosina Trifosfatases/química , Anidrases Carbônicas/química , Proteínas de Transporte de Cátions/química , Metalochaperonas/química , Metais/química , Superóxido Dismutase/química , Elementos de Transição/química , Animais , Sítios de Ligação , Calorimetria , Bovinos , Proteínas de Transporte de Cobre , ATPases Transportadoras de Cobre , Ferritinas/química , Humanos , Cinética , Chaperonas Moleculares , Ligação Proteica , Superóxido Dismutase-1 , Termodinâmica , Transferrina/química
6.
Biochim Biophys Acta Gen Subj ; 1861(12): 3257-3262, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28943300

RESUMO

BACKGROUND: Ferritins are ubiquitous multi-subunit iron storage and detoxification proteins that play a critical role in iron homeostasis. Ferrous ions that enter the protein's shell through hydrophilic channels are rapidly oxidized at dinuclear centers on the H-subunit before transfer to the protein's cavity for storage. The mechanisms of iron loading have been extensively studied, but little is known about iron mobilization. Fe(III) reduction can occur via rapid reduction by suitable reducing agents followed by chelation of Fe(II) ions or via direct and slow Fe(III) chelation. Here, the iron release kinetics from ferritin by FMNH2 in the presence of various chaotropic agents are studied and their in-vivo physiological significance discussed. METHODS: The iron release kinetics from horse and human ferritins by FMNH2 were monitored at 522nm where the Fe(II)-bipyridine complex absorbs. The experiments were performed in the presence of different concentrations of three chaotropic agents, urea, guanidine HCl, and triton. RESULTS AND CONCLUSIONS: Under our experimental conditions, iron reductive mobilization by the non-enzymatic FMN/NAD(P)H system is limited by the concentration of FMNH2 and is independent on the type or amount of chaotropes present. Diffusion of FMNH2 through the ferritin pores is an unlikely mechanism for ferritin iron reduction. An iron mobilization mechanism involving rapid electron transfer through the protein shell is discussed. GENERAL SIGNIFICANCE: Caution must be exercised when interpreting the kinetics of iron mobilization from ferritin using the FMN/NAD(P)H system. The kinetics are highly dependent on the amount of dissolved oxygen and the concentration of reagents used.


Assuntos
Ferritinas/química , Mononucleotídeo de Flavina/farmacologia , Hidroquinonas/farmacologia , Ferro/química , Animais , Transporte de Elétrons , Guanidina/farmacologia , Cavalos , Humanos , Cinética , Octoxinol/farmacologia , Oxirredução , Ureia/farmacologia
7.
Biochim Biophys Acta ; 1850(9): 1930-41, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26093289

RESUMO

BACKGROUND: Iron is an essential element for bacterial growth and virulence. Because of its limited bioavailability in the host, bacteria have adapted several strategies to acquire iron during infection. In the human opportunistic bacteria Bacillus cereus, a surface protein IlsA is shown to be involved in iron acquisition from both ferritin and hemoproteins. IlsA has a modular structure consisting of a NEAT (Near Iron transporter) domain at the N-terminus, several LRR (Leucine Rich Repeat) motifs and a SLH (Surface Layer Homology) domain likely involved in anchoring the protein to the cell surface. METHODS: Isothermal titration calorimetry, UV-Vis spectrophotometry, affinity chromatography and rapid kinetics stopped-flow measurements were employed to probe the binding and transfer of hemin between two different B. cereus surface proteins (IlsA and IsdC). RESULTS: IlsA binds hemin via the NEAT domain and is able to extract heme from hemoglobin whereas the LRR domain alone is not involved in these processes. A rapid hemin transfer from hemin-containing IlsA (holo-IlsA) to hemin-free IsdC (apo-IsdC) is demonstrated. CONCLUSIONS: For the first time, it is shown that two different B. cereus surface proteins (IlsA and IsdC) can interact and transfer heme suggesting their involvement in B. cereus heme acquisition. GENERAL SIGNIFICANCE: An important role for the complete Isd system in heme-associated bacterial growth is demonstrated and new insights into the interplay between an Isd NEAT surface protein and an IlsA-NEAT-LRR protein, both of which appear to be involved in heme-iron acquisition in B. cereus are revealed.


Assuntos
Bacillus cereus/química , Proteínas de Bactérias/química , Heme/química , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Heme/metabolismo , Hemina/metabolismo , Ferro/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Análise de Sequência de Proteína , Termodinâmica
8.
PLoS Pathog ; 10(2): e1003935, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24550730

RESUMO

In host-pathogen interactions, the struggle for iron may have major consequences on the outcome of the disease. To overcome the low solubility and bio-availability of iron, bacteria have evolved multiple systems to acquire iron from various sources such as heme, hemoglobin and ferritin. The molecular basis of iron acquisition from heme and hemoglobin have been extensively studied; however, very little is known about iron acquisition from host ferritin, a 24-mer nanocage protein able to store thousands of iron atoms within its cavity. In the human opportunistic pathogen Bacillus cereus, a surface protein named IlsA (Iron-regulated leucine rich surface protein type A) binds heme, hemoglobin and ferritin in vitro and is involved in virulence. Here, we demonstrate that IlsA acts as a ferritin receptor causing ferritin aggregation on the bacterial surface. Isothermal titration calorimetry data indicate that IlsA binds several types of ferritins through direct interaction with the shell subunits. UV-vis kinetic data show a significant enhancement of iron release from ferritin in the presence of IlsA indicating for the first time that a bacterial protein might alter the stability of the ferritin iron core. Disruption of the siderophore bacillibactin production drastically reduces the ability of B. cereus to utilize ferritin for growth and results in attenuated bacterial virulence in insects. We propose a new model of iron acquisition in B. cereus that involves the binding of IlsA to host ferritin followed by siderophore assisted iron uptake. Our results highlight a possible interplay between a surface protein and a siderophore and provide new insights into host adaptation of B. cereus and general bacterial pathogenesis.


Assuntos
Bacillus cereus/patogenicidade , Ferritinas/metabolismo , Interações Hospedeiro-Patógeno/fisiologia , Ferro/metabolismo , Oligopeptídeos/metabolismo , Animais , Bacillus cereus/metabolismo , Proteínas de Bactérias/metabolismo , Imunofluorescência , Mariposas/metabolismo , Mariposas/microbiologia , Virulência/fisiologia
9.
Biochim Biophys Acta ; 1830(10): 4669-74, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23726988

RESUMO

BACKGROUND: Extensive in-vitro studies have focused on elucidating the mechanism of iron uptake and mineral core formation in ferritin. However, despite a plethora of studies attempting to characterize iron release under different experimental conditions, the in-vivo mobilization of iron from ferritin remains poorly understood. Several iron-reductive mobilization pathways have been proposed including, among others, flavin mononucleotides, ascorbate, glutathione, dithionite, and polyphenols. Here, we investigate the kinetics of iron release from ferritin by reduced flavin nucleotide, FMNH2, and discuss the physiological significance of this process in-vivo. METHODS: Iron release from horse spleen ferritin and recombinant human heteropolymer ferritin was followed by the change in optical density of the Fe(II)-bipyridine complex using a Cary 50 Bio UV-Vis spectrophotometer. Oxygen consumption curves were followed on a MI 730 Clark oxygen microelectrode. RESULTS: The reductive mobilization of iron from ferritin by the nonenzymatic FMN/NAD(P)H system is extremely slow in the presence of oxygen and might involve superoxide radicals, but not FMNH2. Under anaerobic conditions, a very rapid phase of iron mobilization by FMNH2 was observed. CONCLUSIONS: Under normoxic conditions, FMNH2 alone might not be a physiologically significant contributor to iron release from ferritin. GENERAL SIGNIFICANCE: There is no consensus on which iron release pathway is predominantly responsible for iron mobilization from ferritin under cellular conditions. While reduced flavin mononucleotide (FMNH2) is one likely candidate for in-vivo ferritin iron removal, its significance is confounded by the rapid oxidation of the latter by molecular oxygen.


Assuntos
Ferritinas/metabolismo , Mononucleotídeo de Flavina/metabolismo , Ferro/metabolismo , Animais , Cavalos , Humanos , NAD/metabolismo , Espectrofotometria Ultravioleta
10.
Int J Biol Macromol ; 278(Pt 4): 135044, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39182888

RESUMO

Ferritin is a 24-mer protein nanocage that stores iron and regulates intracellular iron homeostasis. The nuclear receptor coactivator-4 (NCOA4) binds specifically to ferritin H subunits and facilitates the autophagic trafficking of ferritin to the lysosome for degradation and iron release. Using isothermal titration calorimetry (ITC), we studied the thermodynamics of the interactions between ferritin and the soluble fragment of NCOA4 (residues 383-522), focusing on the effects of the recently identified FeS cluster bound to NCOA4, ferritin subunit composition, and ferritin-iron loading. Our findings show that in the presence of the FeS cluster, the binding is driven by a more favorable enthalpy change and a decrease in entropy change, indicating a key role for the FeS cluster in the structural organization and stability of the complex. The ferritin iron core further enhances this association, increasing binding enthalpy and stabilizing the NCOA4-ferritin complex. The ferritin subunit composition primarily affects binding stoichiometry of the reaction based on the number of H subunits in the ferritin H/L oligomer. Our results demonstrate that both the FeS cluster and the ferritin iron core significantly affect the binding thermodynamics of the NCOA4-ferritin interactions, suggesting regulatory roles for the FeS cluster and ferritin iron content in ferritinophagy.


Assuntos
Calorimetria , Ferritinas , Ferro , Coativadores de Receptor Nuclear , Ligação Proteica , Termodinâmica , Coativadores de Receptor Nuclear/metabolismo , Coativadores de Receptor Nuclear/química , Ferro/metabolismo , Ferro/química , Ferritinas/química , Ferritinas/metabolismo , Humanos , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/química , Subunidades Proteicas/metabolismo , Subunidades Proteicas/química
11.
Anal Methods ; 16(31): 5391-5398, 2024 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-38978467

RESUMO

We report a simple and highly sensitive colorimetric method for the detection and quantification of proteins, based on the aggregation of ascorbic acid (AA) capped gold nanoparticles (AuNPs) by proteins. The interactions between our AuNPs and nine different proteins of various sizes and shapes (cytochrome C (12 kDa), lysozyme (14.3 kDa), myoglobin (17 kDa), human serum albumin (66 kDa), bovine serum albumin (66.4 kDa), human transferrin (80 kDa), aldolase (160 kDa), catalase (240 kDa), and human H-ferritin (500 kDa)) generated similar AuNPs-protein absorption spectra in a concentration-dependent manner in the range of 1-15 nM. Upon the addition of a protein, the UV-visible spectra of AuNPs-protein conjugates shifted from 524 nm for the AuNps alone to longer wavelength (600-750 nm) due to the presence of one of these proteins. This bathochromic shift is accompanied by a color change from a cherry red, to dark purple, and then light grey or colorless if excess protein has been added, indicating the formation of AuNPs-protein conjugates followed by protein-induced aggregation of the AuNPs. High-resolution transmission electron microscopy images revealed uniformly distributed spherical nanoparticles with an average size of 27.5 ± 15.2 nm, increasing in size to 39.6 ± 12.9 nm upon the addition of a protein, indicating the formation of AuNPs-protein conjugates in solution. A general mechanism for the protein-induced aggregation of our AuNPs is proposed. The consistent behavior observed with the nine proteins tested in our study suggests that our assay can be universally applied for the quantification of pure proteins in a solution, regardless of size, shape, or molecular weight.


Assuntos
Ácido Ascórbico , Colorimetria , Ouro , Nanopartículas Metálicas , Proteínas , Ouro/química , Ácido Ascórbico/química , Colorimetria/métodos , Nanopartículas Metálicas/química , Humanos , Proteínas/química , Proteínas/análise , Animais , Bovinos , Espectrofotometria Ultravioleta/métodos
12.
Protein Sci ; 33(8): e5104, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38995055

RESUMO

Despite ferritin's critical role in regulating cellular and systemic iron levels, our understanding of the structure and assembly mechanism of isoferritins, discovered over eight decades ago, remains limited. Unveiling how the composition and molecular architecture of hetero-oligomeric ferritins confer distinct functionality to isoferritins is essential to understanding how the structural intricacies of H and L subunits influence their interactions with cellular machinery. In this study, ferritin heteropolymers with specific H to L subunit ratios were synthesized using a uniquely engineered plasmid design, followed by high-resolution cryo-electron microscopy analysis and deep learning-based amino acid modeling. Our structural examination revealed unique architectural features during the self-assembly mechanism of heteropolymer ferritins and demonstrated a significant preference for H-L heterodimer formation over H-H or L-L homodimers. Unexpectedly, while dimers seem essential building blocks in the protein self-assembly process, the overall mechanism of ferritin self-assembly is observed to proceed randomly through diverse pathways. The physiological significance of these findings is discussed including how ferritin microheterogeneity could represent a tissue-specific adaptation process that imparts distinctive tissue-specific functions to isoferritins.


Assuntos
Ferritinas , Multimerização Proteica , Humanos , Ferritinas/química , Ferritinas/metabolismo , Ferritinas/genética , Modelos Moleculares , Microscopia Crioeletrônica
13.
Nat Commun ; 15(1): 3802, 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38714719

RESUMO

The interaction between nuclear receptor coactivator 4 (NCOA4) and the iron storage protein ferritin is a crucial component of cellular iron homeostasis. The binding of NCOA4 to the FTH1 subunits of ferritin initiates ferritinophagy-a ferritin-specific autophagic pathway leading to the release of the iron stored inside ferritin. The dysregulation of NCOA4 is associated with several diseases, including neurodegenerative disorders and cancer, highlighting the NCOA4-ferritin interface as a prime target for drug development. Here, we present the cryo-EM structure of the NCOA4-FTH1 interface, resolving 16 amino acids of NCOA4 that are crucial for the interaction. The characterization of mutants, designed to modulate the NCOA4-FTH1 interaction, is used to validate the significance of the different features of the binding site. Our results explain the role of the large solvent-exposed hydrophobic patch found on the surface of FTH1 and pave the way for the rational development of ferritinophagy modulators.


Assuntos
Microscopia Crioeletrônica , Ferritinas , Coativadores de Receptor Nuclear , Ferritinas/metabolismo , Ferritinas/química , Ferritinas/genética , Humanos , Coativadores de Receptor Nuclear/metabolismo , Coativadores de Receptor Nuclear/química , Coativadores de Receptor Nuclear/genética , Ligação Proteica , Sítios de Ligação , Ferro/metabolismo , Autofagia , Modelos Moleculares , Células HEK293 , Oxirredutases/metabolismo , Oxirredutases/química , Oxirredutases/genética , Proteólise , Mutação
14.
Biochim Biophys Acta ; 1820(3): 318-25, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21843602

RESUMO

BACKGROUND: In mammals, serum-transferrins transport iron from the neutral environment of the blood to the cytoplasm by receptor-mediated endocytosis. Extensive in-vitro studies have focused on the thermodynamics and kinetics of Fe(3+) binding to a number of transferrins. However, little attention has been given to the thermodynamic characterization of the interaction of transferrin with its receptor. SCOPE OF REVIEW: Iron-loaded transferrin (Tf) binds with high affinity to the specific transferrin receptor (TfR) on the cell surface. The Tf-TfR complex is then internalized via receptor mediated endocytosis into an endosome where iron is released. Here, we provide an overview of recent studies that have used ITC to quantify the interaction of various metal ions with transferrin and highlight our current understanding of the thermodynamics of the transferrin-transferrin receptor system at physiological pH. GENERAL SIGNIFICANCE: The interaction of the iron-loaded transferrin with the transferrin receptor is a key cellular process that occurs during the normal course of iron metabolism. Understanding the thermodynamics of this interaction is important for iron homeostasis since the physiological requirement of iron must be appropriately maintained to avoid iron-related diseases. MAJOR CONCLUSIONS: The thermodynamic data revealed stoichiometric binding of all tested metal ions to transferrin with very high affinities ranging between 10(17) and 10(22)M(-1). Iron-loaded transferrin (monoferric or diferric) is shown to bind avidly (K~10(7)-10(8)M(-1)) to the receptor at neutral pH with a stoichiometry of one Tf molecule per TfR monomer. Significantly, both the N- and the C-lobe contribute to the binding interaction which is shown to be both enthalpically and entropically driven. This article is part of a Special Issue entitled Transferrins: Molecular mechanisms of iron transport and disorders.


Assuntos
Calorimetria/métodos , Receptores da Transferrina/metabolismo , Transferrinas/química , Transferrinas/metabolismo , Animais , Endocitose , Humanos , Ferro/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Receptores da Transferrina/química , Termodinâmica
15.
Protein Sci ; 32(1): e4543, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36519270

RESUMO

Mammalian ferritins are predominantly heteropolymeric species consisting of 2 structurally similar, but functionally and genetically distinct subunit types, called H (Heavy) and L (Light). The two subunits co-assemble in different H and L ratios to form 24-mer shell-like protein nanocages where thousands of iron atoms can be mineralized inside a hollow cavity. Here, we use differential scanning calorimetry (DSC) to study ferritin stability and understand how various combinations of H and L subunits confer aspects of protein structure-function relationships. Using a recently engineered plasmid design that enables the synthesis of complex ferritin nanostructures with specific H to L subunit ratios, we show that homopolymer L and heteropolymer L-rich ferritins have a remarkable hyperthermostability (Tm = 115 ± 1°C) compared to their H-ferritin homologues (Tm = 93 ± 1°C). Our data reveal a significant linear correlation between protein thermal stability and the number of L subunits present on the ferritin shell. A strong and unexpected iron-induced protein thermal destabilization effect (ΔTm up to 20°C) is observed. To our knowledge, this is the first report of recombinant human homo- and hetero-polymer ferritins that exhibit surprisingly high dissociation temperatures, the highest among all known ferritin species, including many known hyperthermophilic proteins and enzymes. This extreme thermostability of our L and L-rich ferritins may have great potential for biotechnological applications.


Assuntos
Ferritinas , Ferro , Animais , Humanos , Ferritinas/genética , Ferritinas/química , Ferro/metabolismo , Polímeros/metabolismo , Plasmídeos/genética , Proteínas Recombinantes/química , Mamíferos
16.
Biochim Biophys Acta Gen Subj ; 1867(3): 130288, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36470367

RESUMO

BACKGROUND: The development of safe and effective vaccines against SARS-CoV-2 and other viruses with high antigenic drift is of crucial importance to public health. Ferritin is a well characterized and ubiquitous iron storage protein that has emerged not only as a useful nanoreactor and nanocarrier, but more recently as an efficient platform for vaccine development. SCOPE OF REVIEW: This review discusses ferritin structure-function properties, self-assembly, and novel bioengineering strategies such as interior cavity and exterior surface modifications for cargo encapsulation and delivery. It also discusses the use of ferritin as a scaffold for biomedical applications, especially for vaccine development against influenza, Epstein-Barr, HIV, hepatitis-C, Lyme disease, and respiratory viruses such as SARS-CoV-2. The use of ferritin for the synthesis of mosaic vaccines to deliver a cocktail of antigens that elicit broad immune protection against different viral variants is also explored. MAJOR CONCLUSIONS: The remarkable stability, biocompatibility, surface functionalization, and self-assembly properties of ferritin nanoparticles make them very attractive platforms for a wide range of biomedical applications, including the development of vaccines. Strong immune responses have been observed in pre-clinical studies against a wide range of pathogens and have led to the exploration of ferritin nanoparticles-based vaccines in multiple phase I clinical trials. GENERAL SIGNIFICANCE: The broad protective antibody response of ferritin nanoparticles-based vaccines demonstrates the usefulness of ferritin as a highly promising and effective approaches for vaccine development.


Assuntos
COVID-19 , Vacinas contra Influenza , Humanos , Ferritinas , COVID-19/prevenção & controle , Vacinas contra COVID-19 , SARS-CoV-2 , Desenvolvimento de Vacinas
17.
Sci Rep ; 13(1): 19862, 2023 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-37963965

RESUMO

Ferritin is a ubiquitous intracellular iron storage protein that plays a crucial role in iron homeostasis. Animal tissue ferritins consist of multiple isoforms (or isoferritins) with different proportions of H and L subunits that contribute to their structural and compositional heterogeneity, and thus physiological functions. Using size exclusion and anion exchange chromatography, capillary isoelectric focusing (cIEF), and SDS-capillary gel electrophoresis (SDS-CGE), we reveal for the first time a significant variation in ferritin subunit composition and isoelectric points, in both recombinant and native ferritins extracted from animal organs. Our results indicate that subunits composition is the main determinant of the mean pI of recombinant ferritin heteropolymers, and that ferritin microheterogeneity is a common property of both natural and recombinant proteins and appears to be an intrinsic feature of the cellular machinery during ferritin expression, regulation, post-translational modifications, and post-subunits assembly. The functional significance and physiological implications of ferritin heterogeneity in terms of iron metabolism, response to oxidative stress, tissue-specific functions, and pathological processes are discussed.


Assuntos
Ferritinas , Ferro , Animais , Ferritinas/metabolismo , Focalização Isoelétrica , Eletroforese em Gel de Poliacrilamida , Ferro/metabolismo , Ponto Isoelétrico
19.
Nanoscale Adv ; 5(1): 208-219, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36605807

RESUMO

The physical properties of in vitro iron-reconstituted and genetically engineered human heteropolymer ferritins were investigated. High-angle annular dark-field scanning transmission electron microscopy (HAADF-STEM), electron energy-loss spectroscopy (EELS), and 57Fe Mössbauer spectroscopy were employed to ascertain (1) the microstructural, electronic, and micromagnetic properties of the nanosized iron cores, and (2) the effect of the H and L ferritin subunit ratios on these properties. Mössbauer spectroscopic signatures indicate that all iron within the core is in the high spin ferric state. Variable temperature Mössbauer spectroscopy for H-rich (H21/L3) and L-rich (H2/L22) ferritins reconstituted at 1000 57Fe/protein indicates superparamagnetic behavior with blocking temperatures of 19 K and 28 K, while HAADF-STEM measurements give average core diameters of (3.7 ± 0.6) nm and (5.9 ± 1.0) nm, respectively. Most significantly, H-rich proteins reveal elongated, dumbbell, and crescent-shaped cores, while L-rich proteins present spherical cores, pointing to a correlation between core shape and protein shell composition. Assuming an attempt time for spin reversal of τ 0 = 10-11 s, the Néel-Brown formula for spin-relaxation time predicts effective magnetic anisotropy energy densities of 6.83 × 104 J m-3 and 2.75 × 104 J m-3 for H-rich and L-rich proteins, respectively, due to differences in surface and shape contributions to magnetic anisotropy in the two heteropolymers. The observed differences in shape, size, and effective magnetic anisotropies of the derived biomineral cores are discussed in terms of the iron nucleation sites within the interior surface of the heteropolymer shells for H-rich and L-rich proteins. Overall, our results imply that site-directed nucleation and core growth within the protein cavity play a determinant role in the resulting core morphology. Our findings have relevance to iron biomineralization processes in nature and the growth of designer's magnetic nanoparticles within recombinant apoferritin nano-templates for nanotechnology.

20.
Biochim Biophys Acta ; 1800(8): 719-31, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20382203

RESUMO

BACKGROUND: Ferritins are ubiquitous and well-characterized iron storage and detoxification proteins. In bacteria and plants, ferritins are homopolymers composed of H-type subunits, while in vertebrates, they typically consist of 24 similar subunits of two types, H and L. The H-subunit is responsible for the rapid oxidation of Fe(II) to Fe(III) at a dinuclear center, whereas the L-subunit appears to help iron clearance from the ferroxidase center of the H-subunit and support iron nucleation and mineralization. SCOPE OF REVIEW: Despite their overall similar structures, ferritins from different origins markedly differ in their iron binding, oxidation, detoxification, and mineralization properties. This chapter provides a brief overview of the structure and function of ferritin, reviews our current knowledge of the process of iron uptake and mineral core formation, and highlights the similarities and differences of the iron oxidation and hydrolysis chemistry in a number of ferritins including those from archaea, bacteria, amphibians, and animals. GENERAL SIGNIFICANCE: Prokaryotic ferritins and ferritin-like proteins (Dps) appear to preferentially use H(2)O(2) over O(2) as the iron oxidant during ferritin core formation. While the product of iron oxidation at the ferroxidase centers of these and other ferritins is labile and is retained inside the protein cavity, the iron complex in the di-iron cofactor proteins is stable and remains at the catalytic site. Differences in the identity and affinity of the ferroxidase center ligands to iron have been suggested to influence the distinct reaction pathways in ferritins and the di-iron cofactor enzymes. MAJOR CONCLUSIONS: The ferritin 3-fold channels are shown to be flexible structures that allow the entry and exit of different ions and molecules through the protein shell. The H- and L-subunits are shown to have complementary roles in iron oxidation and mineralization, and hydrogen peroxide appears to be a by-product of oxygen reduction at the FC of most ferritins. The di-iron(III) complex at the FC of some ferritins acts as a stable cofactor during iron oxidation rather than a catalytic center where Fe(II) is oxidized at the FC followed by its translocation to the protein cavity.


Assuntos
Ferritinas/química , Ferritinas/metabolismo , Ferro/metabolismo , Animais , Domínio Catalítico , Humanos , Hidrólise , Ferro/química , Cinética , Modelos Biológicos , Modelos Moleculares , Oxirredução , Conformação Proteica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA