Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Inorg Biochem ; 233: 111839, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35599166

RESUMO

Globins play a key role in regulating nitric oxide (NO) levels in all forms of life. Five key reactions of NO with mammalian muscle myoglobin (Mb) and red blood cell hemoglobin (Hb) have been examined: (1) reversible NO binding to Fe(II) forms; (2) reversible NO binding to Fe(III) forms; (3) NO dioxygenation by Fe(II)O2 complexes; (4) autoxidation of Fe(II)NO complexes in the presence of O2; and (5) autoreduction of Fe(III)NO complexes. NO reacts rapidly and almost irreversibly with deoxyMb(FeII) in the absence of O2, whereas it reacts much more slowly and weakly with metMb(FeIII). The reaction of NO with Mb(FeII)O2 is very rapid and results in oxidation of the iron atom and dioxygenation of NO to nitrate. Autoxidation of Mb(FeII)NO in air is determined by the slow rate of NO dissociation from the Fe(II)NO complex, which is followed by rapid O2 binding to the newly formed deoxyMb(FeII) and dioxygenation of the displaced NO to generate NO3- and metMb(FeIII). MetMb(FeIII)NO autoreduces slowly by addition of a hydroxide ion to bound NO to generate nitrous acid and reduced deoxyMb(FeII), which immediately binds another NO to generate Mb(FeII)NO as the final product. The reverse of this process involves nitrite reduction to NO by deoxyMb(FeII), which can occur on physiological time scales when the globin concentration is in the millimolar range. The relevance of these processes to the regulation of NO metabolism by hemoglobins and myoglobins in humans and other organisms is discussed.


Assuntos
Hemoglobinas , Mamíferos , Mioglobina , Óxido Nítrico , Animais , Compostos Férricos/química , Compostos Férricos/metabolismo , Globinas/química , Globinas/metabolismo , Hemoglobinas/química , Hemoglobinas/metabolismo , Humanos , Mamíferos/metabolismo , Mioglobina/química , Mioglobina/metabolismo , Óxido Nítrico/química , Óxido Nítrico/metabolismo , Nitritos/química , Oxirredução
2.
Mol Aspects Med ; 84: 101024, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34544605

RESUMO

Antonini and Brunori's 1971 book "Hemoglobin and Myoglobin in Their Reactions with Ligands" was a truly remarkable publication that summarized almost 100 years of research on O2 binding to these globins. Over the ensuing 50 years, ultra-fast laser photolysis techniques, high-resolution and time resolved X-ray crystallography, molecular dynamics simulations, and libraries of recombinant myoglobin (Mb) and hemoglobin (Hb) variants have provided structural interpretations of O2 binding to these proteins. The resultant mechanisms provide quantitative descriptions of the stereochemical factors that govern overall affinity, including proximal and distal steric restrictions that affect iron reactivity and favorable positive electrostatic interactions that preferentially stabilize bound O2. The pathway for O2 uptake and release by Mb and subunits of Hb has been mapped by screening libraries of site-directed mutants in laser photolysis experiments. O2 enters mammalian Mb and the α and ß subunits of human HbA through a channel created by upward and outward rotation of the distal His at the E7 helical position, is non-covalently captured in the interior of the distal cavity, and then internally forms a bond with the heme Fe(II) atom. O2 dissociation is governed by disruption of hydrogen bonding interactions with His (E7), breakage of the Fe(II)-O2 bond, and then competition between rebinding and escape through the E7-gate. The structural features that govern the rates of both the individual steps and overall reactions have been determined and provide the framework for: (1) defining the physiological functions of specific globins and their evolution; (2) understanding the clinical features of hemoglobinopathies; and (3) designing safer and more efficient acellular hemoglobin-based oxygen carriers (HBOCs) for transfusion therapy, organ preservation, and other commercially relevant O2 transport and storage processes.


Assuntos
Monóxido de Carbono , Mioglobina , Animais , Monóxido de Carbono/metabolismo , Hemoglobinas/química , Humanos , Cinética , Ligantes , Mamíferos , Mioglobina/química , Mioglobina/metabolismo , Oxigênio
4.
Biophys J ; 118(6): 1381-1400, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32075750

RESUMO

Hemoglobin functions as a tetrameric oxygen transport protein, with each subunit containing a heme cofactor. Its denaturation, either in vivo or in vitro, involves autoxidation to methemoglobin, followed by cofactor loss and globin unfolding. We have proposed a global disassembly scheme for human methemoglobin, linking hemin (ferric protoporphyrin IX) disassociation and apoprotein unfolding pathways. The model is based on the evaluation of circular dichroism and visible absorbance measurements of guanidine-hydrochloride-induced disassembly of methemoglobin and previous measurements of apohemoglobin unfolding. The populations of holointermediates and equilibrium disassembly parameters were estimated quantitatively for adult and fetal hemoglobins. The key stages are characterized by hexacoordinated hemichrome intermediates, which are important for preventing hemin disassociation from partially unfolded, molten globular species during early disassembly and late-stage assembly events. Both unfolding experiments and independent small angle x-ray scattering measurements demonstrate that heme disassociation leads to the loss of tetrameric structural integrity. Our model predicts that after autoxidation, dimeric and monomeric hemichrome intermediates occur along the disassembly pathway inside red cells, where the hemoglobin concentration is very high. This prediction suggests why misassembled hemoglobins often get trapped as hemichromes that accumulate into insoluble Heinz bodies in the red cells of patients with unstable hemoglobinopathies. These Heinz bodies become deposited on the cell membranes and can lead to hemolysis. Alternatively, when acellular hemoglobin is diluted into blood plasma after red cell lysis, the disassembly pathway appears to be dominated by early hemin disassociation events, which leads to the generation of higher fractions of unfolded apo subunits and free hemin, which are known to damage the integrity of blood vessel walls. Thus, our model provides explanations of the pathophysiology of hemoglobinopathies and other disease states associated with unstable globins and red cell lysis and also insights into the factors governing hemoglobin assembly during erythropoiesis.


Assuntos
Heme , Hemoglobinas , Eritrócitos , Hemólise , Humanos , Metemoglobina
5.
Antioxid Redox Signal ; 32(4): 228-246, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31530172

RESUMO

Significance: Over the past 50 years, the mechanisms for O2 storage and transport have been determined quantitatively on distance scales from millimeters to tenths of nanometers and timescales from seconds to picoseconds. Recent Advances: In this review, I have described four key conclusions from work done by my group and our close colleagues. (i) O2 uptake by mammalian red cells is limited by diffusion through unstirred water layers adjacent to the cell surface and across cell-free layers adjacent to vessel walls. (ii) In most vertebrates, hemoglobins (Hbs) and myoglobins (Mbs), the distal histidine at the E7 helical position donates a strong hydrogen bond to bound O2, which selectively enhances O2 affinity, prevents carbon monoxide poisoning, and markedly slows autoxidation. (iii) O2 binding to mammalian Hbs and Mbs occurs by migration of the ligand through a channel created by upward rotation of the His(E7) side chain, capture in the empty space of the distal pocket, and then coordination with the ferroprotoporphyrin IX (heme) iron atom. (iv) The assembly of Mbs and Hbs occurs by formation of molten globule intermediates, in which the N- and C-terminal helices have almost fully formed secondary structures, but the heme pockets are disordered and followed by high-affinity binding of heme. Critical Issues: These conclusions indicate that there are often compromises between O2 transport function, holoprotein stability, and the efficiency of assembly. Future Directions: However, the biochemical mechanisms underlying these conclusions provide the framework for understanding globin evolution in greater detail and for engineering more efficient and stable globins.


Assuntos
Hemoglobinas/metabolismo , Mioglobina/metabolismo , Oxigênio/metabolismo , Animais , Hemoglobinas/química , Humanos , Modelos Moleculares , Mioglobina/química , Estabilidade Proteica , Estrutura Secundária de Proteína , Eletricidade Estática
6.
J Biol Chem ; 294(11): 4145-4159, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30630954

RESUMO

After reacting with hydrogen peroxide (H2O2), sickle-cell hemoglobin (HbS, ßE6V) remains longer in a highly oxidizing ferryl form (HbFe4+=O) and induces irreversible oxidation of "hot-spot" amino acids, including ßCys-93. To control the damaging ferryl heme, here we constructed three HbS variants. The first contained a redox-active Tyr in ß subunits (F41Y), a substitution present in Hb Mequon; the second contained the Asp (K82D) found in the ß cleft of Hb Providence; and the third had both of these ß substitutions. Both the single Tyr-41 and Asp-82 constructs lowered the oxygen affinity of HbS but had little or no effects on autoxidation or heme loss kinetics. In the presence of H2O2, both rHbS ßF41Y and ßF41Y/K82D enhanced ferryl Hb reduction by providing a pathway for electrons to reduce the heme via the Tyr-41 side chain. MS analysis of ßCys-93 revealed moderate inhibition of thiol oxidation in the HbS single F41Y variant and dramatic 3- to 8-fold inhibition of cysteic acid formation in rHbS ßK82D and ßF41Y/K82D, respectively. Under hypoxia, ßK82D and ßF41Y/K82D HbS substitutions increased the delay time by ∼250 and 600 s before the onset of polymerization compared with the rHbS control and rHbS ßF41Y, respectively. Moreover, at 60 °C, rHbS ßK82D exhibited superior structural stability. Asp-82 also enhanced the function of Tyr as a redox-active amino acid in the rHbS ßF41Y/K82D variant. We conclude that the ßK82D and ßF41Y substitutions add significant resistance to oxidative stress and anti-sickling properties to HbS and therefore could be potential genome-editing targets.


Assuntos
Anemia Falciforme/metabolismo , Hemoglobina Falciforme/metabolismo , Hemoglobina Falciforme/análise , Hemoglobina Falciforme/genética , Humanos , Cinética , Oxirredução , Estabilidade Proteica , Proteínas Recombinantes/análise , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Temperatura , Fatores de Tempo
7.
Shock ; 52(1S Suppl 1): 28-40, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-29112633

RESUMO

This article reviews the key biochemical mechanisms that govern O2 transport, NO scavenging, and oxidative degradation of acellular hemoglobin (Hb) and how these ideas have been used to try to develop strategies to engineer safer and more effective hemoglobin-based oxygen carriers (HBOCs). Significant toxicities due to acellular Hb have been observed after the administration of HBOCs or after the lysis of red cells, and include rapid clearance and kidney damage due to dissociation into dimers, haptoglobin binding, and macrophage activation; early O2 release leading to decreased tissue perfusion in capillary beds; interference with endothelial and smooth muscle signaling due to nitric oxide (NO) scavenging; autooxidization of heme iron followed by production of reactive oxygen species; and iron overload symptoms due to hemin loss, globin denaturation, iron accumulation, and further inflammation. Protein engineering can be used to mitigate some of these side effects, but requires an in-depth mechanistic understanding of the biochemical and biophysical features of Hb that regulate quaternary structure, O2 affinity, NO dioxygenation, and resistance to oxidation, hemin loss, and unfolding.


Assuntos
Hemoglobinas/metabolismo , Oxigênio/sangue , Engenharia de Proteínas/métodos , Animais , Substitutos Sanguíneos , Humanos
8.
J Biol Chem ; 293(18): 6942-6957, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29540481

RESUMO

Staphylococcus aureus is a leading cause of life-threatening infections in the United States. It actively acquires the essential nutrient iron from human hemoglobin (Hb) using the iron-regulated surface-determinant (Isd) system. This process is initiated when the closely related bacterial IsdB and IsdH receptors bind to Hb and extract its hemin through a conserved tri-domain unit that contains two NEAr iron Transporter (NEAT) domains that are connected by a helical linker domain. Previously, we demonstrated that the tri-domain unit within IsdH (IsdHN2N3) triggers hemin release by distorting Hb's F-helix. Here, we report that IsdHN2N3 promotes hemin release from both the α- and ß-subunits. Using a receptor mutant that only binds to the α-subunit of Hb and a stopped-flow transfer assay, we determined the energetics and micro-rate constants of hemin extraction from tetrameric Hb. We found that at 37 °C, the receptor accelerates hemin release from Hb up to 13,400-fold, with an activation enthalpy of 19.5 ± 1.1 kcal/mol. We propose that hemin removal requires the rate-limiting hydrolytic cleavage of the axial HisF8 Nϵ-Fe3+ bond, which, based on molecular dynamics simulations, may be facilitated by receptor-induced bond hydration. Isothermal titration calorimetry experiments revealed that two distinct IsdHN2N3·Hb protein·protein interfaces promote hemin release. A high-affinity receptor·Hb(A-helix) interface contributed ∼95% of the total binding standard free energy, enabling much weaker receptor interactions with Hb's F-helix that distort its hemin pocket and cause unfavorable changes in the binding enthalpy. We present a model indicating that receptor-introduced structural distortions and increased solvation underlie the IsdH-mediated hemin extraction mechanism.


Assuntos
Metabolismo Energético , Hemina/isolamento & purificação , Hemoglobinas/química , Staphylococcus aureus/metabolismo , Antígenos de Bactérias/metabolismo , Sítios de Ligação , Biopolímeros/química , Biopolímeros/metabolismo , Calorimetria , Proteínas de Transporte de Cátions/metabolismo , Hemina/metabolismo , Hemoglobinas/metabolismo , Humanos , Hidrólise , Cinética , Simulação de Dinâmica Molecular , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica , Receptores de Superfície Celular/metabolismo , Termodinâmica
9.
Biochem J ; 474(24): 4171-4192, 2017 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-29070524

RESUMO

Previous work suggested that hemoglobin (Hb) tetramer formation slows autoxidation and hemin loss and that the naturally occurring mutant, Hb Providence (HbProv; ßK82D), is much more resistant to degradation by H2O2 We have examined systematically the effects of genetic cross-linking of Hb tetramers with and without the HbProv mutation on autoxidation, hemin loss, and reactions with H2O2, using native HbA and various wild-type recombinant Hbs as controls. Genetically cross-linked Hb Presbyterian (ßN108K) was also examined as an example of a low oxygen affinity tetramer. Our conclusions are: (a) at low concentrations, all the cross-linked tetramers show smaller rates of autoxidation and hemin loss than HbA, which can dissociate into much less stable dimers and (b) the HbProv ßK82D mutation confers more resistance to degradation by H2O2, by markedly inhibiting oxidation of the ß93 cysteine side chain, particularly in cross-linked tetramers and even in the presence of the destabilizing Hb Presbyterian mutation. These results show that cross-linking and the ßK82D mutation do enhance the resistance of Hb to oxidative degradation, a critical element in the design of a safe and effective oxygen therapeutic.


Assuntos
Hemoglobinas/química , Hemoglobinas/genética , Mutação de Sentido Incorreto , Reagentes de Ligações Cruzadas/química , Dimerização , Hemoglobinas/metabolismo , Humanos , Peróxido de Hidrogênio/química , Oxirredução , Engenharia de Proteínas
10.
Biochemistry ; 56(10): 1444-1459, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28218841

RESUMO

Removal of heme from human hemoglobin (Hb) results in formation of an apoglobin heterodimer. Titration of this apodimer with guanidine hydrochloride (GdnHCl) leads to biphasic unfolding curves indicating two distinct steps. Initially, the heme pocket unfolds and generates a dimeric intermediate in which ∼50% of the original helicity is lost, but the α1ß1 interface is still intact. At higher GdnHCl concentrations, this intermediate dissociates into unfolded monomers. This structural interpretation was verified by comparing GdnHCl titrations for adult human hemoglobin A (HbA), recombinant fetal human hemoglobin (HbF), recombinant Hb cross-linked with a single glycine linker between the α chains, and recombinant Hbs with apolar heme pocket mutations that markedly stabilize native conformations in both subunits. The first phase of apoHb unfolding is independent of protein concentration, little affected by genetic cross-linking, but significantly shifted toward higher GdnHCl concentrations by the stabilizing distal pocket mutations. The second phase depends on protein concentration and is shifted to higher GdnHCl concentrations by genetic cross-linking. This model for apoHb unfolding allowed us to quantitate subtle differences in stability between apoHbA and apoHbF, which suggest that the ß and γ heme pockets have similar stabilities, whereas the α1γ1 interface is more resistant to dissociation than the α1ß1 interface.


Assuntos
Apoproteínas/química , Hemoglobina Fetal/química , Guanidina/química , Hemoglobina A/química , Hemoglobinas/química , Apoproteínas/genética , Apoproteínas/metabolismo , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Expressão Gênica , Glicina/química , Glicina/metabolismo , Heme/química , Heme/isolamento & purificação , Heme/metabolismo , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Cinética , Desnaturação Proteica , Domínios Proteicos , Dobramento de Proteína , Multimerização Proteica , Estabilidade Proteica , Estrutura Secundária de Proteína , Desdobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
11.
J Biol Chem ; 292(6): 2542-2555, 2017 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28011635

RESUMO

Mutations in hemoglobin can cause a wide range of phenotypic outcomes, including anemia due to protein instability and red cell lysis. Uncovering the biochemical basis for these phenotypes can provide new insights into hemoglobin structure and function as well as identify new therapeutic opportunities. We report here a new hemoglobin α chain variant in a female patient with mild anemia, whose father also carries the trait and is from the Turkish city of Kirklareli. Both the patient and her father had a His-58(E7) → Leu mutation in α1. Surprisingly, the patient's father is not anemic, but he is a smoker with high levels of HbCO (∼16%). To understand these phenotypes, we examined recombinant human Hb (rHb) Kirklareli containing the α H58L replacement. Mutant α subunits containing Leu-58(E7) autoxidize ∼8 times and lose hemin ∼200 times more rapidly than native α subunits, causing the oxygenated form of rHb Kirklareli to denature very rapidly under physiological conditions. The crystal structure of rHb Kirklareli shows that the α H58L replacement creates a completely apolar active site, which prevents electrostatic stabilization of bound O2, promotes autoxidation, and enhances hemin dissociation by inhibiting water coordination to the Fe(III) atom. At the same time, the mutant α subunit has an ∼80,000-fold higher affinity for CO than O2, causing it to rapidly take up and retain carbon monoxide, which prevents denaturation both in vitro and in vivo and explains the phenotypic differences between the father, who is a smoker, and his daughter.


Assuntos
Anemia Ferropriva/sangue , Monóxido de Carbono/metabolismo , Hemoglobinas Anormais/metabolismo , Adulto , Domínio Catalítico , Cromatografia Líquida de Alta Pressão , Cromatografia por Troca Iônica , Cromatografia de Fase Reversa , Cristalografia por Raios X , Feminino , Hemoglobinas Anormais/química , Humanos , Masculino , Espectrometria de Massas , Oxirredução , Oxigênio/metabolismo , Eletricidade Estática , Adulto Jovem
12.
Biochemistry ; 55(29): 4005-17, 2016 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-27355904

RESUMO

Water molecules can enter the heme pockets of unliganded myoglobins and hemoglobins, hydrogen bond with the distal histidine, and introduce steric barriers to ligand binding. The spectrokinetics of photodissociated CO complexes of human hemoglobin and its isolated α and ß chains were analyzed for the effect of heme hydration on ligand rebinding. A strong coupling was observed between heme hydration and quaternary state. This coupling may contribute significantly to the 20-60-fold difference between the R- and T-state bimolecular CO binding rate constants and thus to the modulation of ligand reactivity that is the hallmark of hemoglobin allostery. Heme hydration proceeded over the course of several kinetic phases in the tetramer, including the R to T quaternary transition. An initial 150 ns hydration phase increased the R-state distal pocket water occupancy, nw(R), to a level similar to that of the isolated α (∼60%) and ß (∼10%) chains, resulting in a modest barrier to ligand binding. A subsequent phase, concurrent with the first step of the R → T transition, further increased the level of heme hydration, increasing the barrier. The final phase, concurrent with the final step of the allosteric transition, brought the water occupancy of the T-state tetramer, nw(T), even higher and close to full occupancy in both the α and ß subunits (∼90%). This hydration level could present an even larger barrier to ligand binding and contribute significantly to the lower iron reactivity of the T state toward CO.


Assuntos
Hemoglobinas/química , Regulação Alostérica , Heme/química , Humanos , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular , Fotólise , Estrutura Quaternária de Proteína , Água/química , alfa-Globinas/química , Globinas beta/química
13.
J Mol Biol ; 428(6): 1107-1129, 2016 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-25687963

RESUMO

Staphylococcus aureus is a medically important bacterial pathogen that, during infections, acquires iron from human hemoglobin (Hb). It uses two closely related iron-regulated surface determinant (Isd) proteins to capture and extract the oxidized form of heme (hemin) from Hb, IsdH and IsdB. Both receptors rapidly extract hemin using a conserved tri-domain unit consisting of two NEAT (near iron transporter) domains connected by a helical linker domain. To gain insight into the mechanism of extraction, we used NMR to investigate the structure and dynamics of the 38.8-kDa tri-domain IsdH protein (IsdH(N2N3), A326-D660 with a Y642A mutation that prevents hemin binding). The structure was modeled using long-range paramagnetic relaxation enhancement (PRE) distance restraints, dihedral angle, small-angle X-ray scattering, residual dipolar coupling and inter-domain NOE nuclear Overhauser effect data. The receptor adopts an extended conformation wherein the linker and N3 domains pack against each other via a hydrophobic interface. In contrast, the N2 domain contacts the linker domain via a hydrophilic interface and, based on NMR relaxation data, undergoes inter-domain motions enabling it to reorient with respect to the body of the protein. Ensemble calculations were used to estimate the range of N2 domain positions compatible with the PRE data. A comparison of the Hb-free and Hb-bound forms reveals that Hb binding alters the positioning of the N2 domain. We propose that binding occurs through a combination of conformational selection and induced-fit mechanisms that may promote hemin release from Hb by altering the position of its F helix.


Assuntos
Antígenos de Bactérias/química , Antígenos de Bactérias/metabolismo , Hemoglobinas/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Ligação Proteica , Conformação Proteica
14.
Blood ; 126(20): 2338-41, 2015 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-26307534

RESUMO

Intravascular hemolysis occurs in patients on extracorporeal membrane oxygenation. High levels of free acellular adult hemoglobin (free HbA) are associated with clotting in this mechanical device that can result in thrombotic complications. Adsorption of fibrinogen onto the surface of biomaterial correlates with platelet adhesion, which is mediated by von Willebrand factor (VWF). Because free Hb interacts with VWF, we studied the effect of hemoglobin (Hb) on platelet adhesion to fibrin(ogen) under conditions of different hydrodynamic forces. This effect was investigated using purified human HbA and fibrinogen, extracellular matrix, collagen, or purified plasma VWF as surface-coated substrates to examine flow-dependent platelet adhesion. Antibodies and VWF-deficient plasma were also used. Free Hb (≥50 mg/dL) effectively augmented platelet adhesion, and microthrombi formation on fibrin(ogen), extracellular matrix, and collagen at high shear stress. The effect of free Hb was effectively blocked by anti-glycoprotein Ibα (GPIbα) antibodies or depletion of VWF. Unexpectedly, free Hb also promoted firm platelet adhesion and stable microthrombi on VWF. Lastly, we determined that Hb interacts directly with the A1 domain. This study is the first to demonstrate that extracellular Hb directly affects the GPIbα-VWF interaction in thrombosis, and describes another mechanism by which hemolysis is connected to thrombotic events.


Assuntos
Plaquetas/metabolismo , Hemodinâmica , Hemoglobinas/metabolismo , Adesividade Plaquetária , Trombose/metabolismo , Fator de von Willebrand/metabolismo , Anticorpos/química , Plaquetas/química , Plaquetas/patologia , Hemoglobinas/química , Humanos , Integrina alfa2/química , Integrina alfa2/metabolismo , Estrutura Terciária de Proteína , Trombose/patologia , Fator de von Willebrand/química
15.
J Biol Chem ; 290(39): 23479-95, 2015 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-26205820

RESUMO

Expression levels in animal muscle tissues and in Escherichia coli vary widely for naturally occurring mammalian myoglobins (Mb). To explore this variation, we developed an in vitro transcription and wheat germ extract-based translation assay to examine quantitatively the factors that govern expression of holoMb. We constructed a library of naturally occurring Mbs from two terrestrial and four deep-diving aquatic mammals and three distal histidine mutants designed to enhance apoglobin stability but decrease hemin affinity. A strong linear correlation is observed between cell-free expression levels of holo-metMb variants and their corresponding apoglobin stabilities, which were measured independently by guanidine HCl-induced unfolding titrations using purified proteins. In contrast, there is little dependence of expression on hemin affinity. Our results confirm quantitatively that deep diving mammals have highly stable Mbs that express to higher levels in animal myocytes, E. coli, and the wheat germ cell-free system than Mbs from terrestrial mammals. Our theoretical analyses show that the rate of aggregation of unfolded apoMb is very large, and as a result, the key factor for high level expression of holoMb, and presumably other heme proteins, is an ultra high fraction of folded, native apoglobin that is capable of rapidly binding hemin. This fraction is determined by the overall equilibrium folding constant and not hemin affinity. These results also demonstrate that the cell-free transcription/translation system can be used as a high throughput platform to screen for apoglobin stability without the need to generate large amounts of protein for in vitro unfolding measurements.


Assuntos
Mioglobina/metabolismo , Isoformas de Proteínas/metabolismo , Animais , Sistema Livre de Células , Escherichia coli/genética , Mioglobina/química , Mioglobina/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Estabilidade Proteica
16.
J Biol Chem ; 289(32): 22342-57, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-24939847

RESUMO

A pathogenic V67M mutation occurs at the E11 helical position within the heme pockets of variant human fetal and adult hemoglobins (Hb). Subsequent post-translational modification of Met to Asp was reported in γ subunits of human fetal Hb Toms River (γ67(E11)Val → Met) and ß subunits of adult Hb (HbA) Bristol-Alesha (ß67(E11)Val → Met) that were associated with hemolytic anemia. Using kinetic, proteomic, and crystal structural analysis, we were able to show that the Met → Asp transformation involves heme cycling through its oxoferryl state in the recombinant versions of both proteins. The conversion to Met and Asp enhanced the spontaneous autoxidation of the mutants relative to wild-type HbA and human fetal Hb, and the levels of Asp were elevated with increasing levels of hydrogen peroxide (H2O2). Using H2(18)O2, we verified incorporation of (18)O into the Asp carboxyl side chain confirming the role of H2O2 in the oxidation of the Met side chain. Under similar experimental conditions, there was no conversion to Asp at the αMet(E11) position in the corresponding HbA Evans (α62(E11)Val → Met). The crystal structures of the three recombinant Met(E11) mutants revealed similar thioether side chain orientations. However, as in the solution experiments, autoxidation of the Hb mutant crystals leads to electron density maps indicative of Asp(E11) formation in ß subunits but not in α subunits. This novel post-translational modification highlights the nonequivalence of human Hb α, ß, and γ subunits with respect to redox reactivity and may have direct implications to α/ß hemoglobinopathies and design of oxidatively stable Hb-based oxygen therapeutics.


Assuntos
Heme/metabolismo , Hemoglobinas/química , Hemoglobinas/metabolismo , Ferro/metabolismo , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Ácido Aspártico/química , Cristalografia por Raios X , Hemoglobina Fetal/química , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Heme/química , Hemoglobina A/química , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobinas/genética , Hemoglobinas Anormais/química , Hemoglobinas Anormais/genética , Hemoglobinas Anormais/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Ferro/química , Metionina/química , Modelos Moleculares , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Oxirredução , Processamento de Proteína Pós-Traducional , Subunidades Proteicas , Proteômica , Eletricidade Estática
17.
Free Radic Biol Med ; 69: 265-77, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24486321

RESUMO

Haptoglobin (Hp) is an abundant and conserved plasma glycoprotein, which binds acellular adult hemoglobin (Hb) dimers with high affinity and facilitates their rapid clearance from circulation after hemolysis. Humans possess three main phenotypes of Hp, designated Hp 1-1, Hp 2-1, and Hp 2-2. These variants exhibit diverse structural configurations and have been reported to be functionally nonequivalent. We have investigated the functional and redox properties of Hb-Hp complexes prepared using commercially fractionated Hp and found that all forms exhibit similar behavior. The rate of Hb dimer binding to Hp occurs with bimolecular rate constants of ~0.9 µM(-1) s(-1), irrespective of the type of Hp assayed. Although Hp binding does accelerate the observed rate of HbO2 autoxidation by dissociating Hb tetramers into dimers, the rate observed for these bound dimers is three- to fourfold slower than that of Hb dimers free in solution. Co-incubation of ferric Hb with any form of Hp inhibits heme loss to below detectable levels. Intrinsic redox potentials (E1/2) of the ferric/ferrous pair of each Hb-Hp complex are similar, varying from +54 to +59 mV (vs NHE), and are essentially the same as reported by us previously for Hb-Hp complexes prepared from unfractionated Hp. All Hb-Hp complexes generate similar high amounts of ferryl Hb after exposure to hydrogen peroxide. Electron paramagnetic resonance data indicate that the yields of protein-based radicals during this process are approximately 4 to 5% and are unaffected by the variant of Hp assayed. These data indicate that the Hp fractions examined are equivalent to one another with respect to Hb binding and associated stability and redox properties and that this result should be taken into account in the design of phenotype-specific Hp therapeutics aimed at countering Hb-mediated vascular disease.


Assuntos
Dimerização , Haptoglobinas/genética , Hemoglobinas/genética , Oxirredução , Espectroscopia de Ressonância de Spin Eletrônica , Haptoglobinas/química , Hemoglobinas/química , Humanos , Peróxido de Hidrogênio/química , Cinética , Complexos Multiproteicos/química , Complexos Multiproteicos/genética , Fenótipo , Polímeros/química
18.
Proc Natl Acad Sci U S A ; 111(3): 900-5, 2014 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-24379385

RESUMO

Successful diagnosis, screening, and elimination of malaria critically depend on rapid and sensitive detection of this dangerous infection, preferably transdermally and without sophisticated reagents or blood drawing. Such diagnostic methods are not currently available. Here we show that the high optical absorbance and nanosize of endogenous heme nanoparticles called "hemozoin," a unique component of all blood-stage malaria parasites, generates a transient vapor nanobubble around hemozoin in response to a short and safe near-infrared picosecond laser pulse. The acoustic signals of these malaria-specific nanobubbles provided transdermal noninvasive and rapid detection of a malaria infection as low as 0.00034% in animals without using any reagents or drawing blood. These on-demand transient events have no analogs among current malaria markers and probes, can detect and screen malaria in seconds, and can be realized as a compact, easy-to-use, inexpensive, and safe field technology.


Assuntos
Eritrócitos/parasitologia , Malária/diagnóstico , Administração Cutânea , Animais , Eritrócitos/metabolismo , Feminino , Gases , Heme/química , Hemeproteínas/química , Humanos , Lasers , Malária/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/química , Agulhas , Plasmodium falciparum
19.
J Biol Chem ; 288(27): 19986-20001, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23696640

RESUMO

α-Hemoglobin (αHb)-stabilizing protein (AHSP) is a molecular chaperone that assists hemoglobin assembly. AHSP induces changes in αHb heme coordination, but how these changes are facilitated by interactions at the αHb·AHSP interface is not well understood. To address this question we have used NMR, x-ray absorption spectroscopy, and ligand binding measurements to probe αHb conformational changes induced by AHSP binding. NMR chemical shift analyses of free CO-αHb and CO-αHb·AHSP indicated that the seven helical elements of the native αHb structure are retained and that the heme Fe(II) remains coordinated to the proximal His-87 side chain. However, chemical shift differences revealed alterations of the F, G, and H helices and the heme pocket of CO-αHb bound to AHSP. Comparisons of iron-ligand geometry using extended x-ray absorption fine structure spectroscopy showed that AHSP binding induces a small 0.03 Å lengthening of the Fe-O2 bond, explaining previous reports that AHSP decreases αHb O2 affinity roughly 4-fold and promotes autooxidation due primarily to a 3-4-fold increase in the rate of O2 dissociation. Pro-30 mutations diminished NMR chemical shift changes in the proximal heme pocket, restored normal O2 dissociation rate and equilibrium constants, and reduced O2-αHb autooxidation rates. Thus, the contacts mediated by Pro-30 in wild-type AHSP promote αHb autooxidation by introducing strain into the proximal heme pocket. As a chaperone, AHSP facilitates rapid assembly of αHb into Hb when ßHb is abundant but diverts αHb to a redox resistant holding state when ßHb is limiting.


Assuntos
Proteínas Sanguíneas/química , Hemoglobina A/química , Ferro/química , Chaperonas Moleculares/química , Oxigênio/química , Oxiemoglobinas/química , Sítios de Ligação , Proteínas Sanguíneas/metabolismo , Hemoglobina A/metabolismo , Humanos , Ferro/metabolismo , Chaperonas Moleculares/metabolismo , Ressonância Magnética Nuclear Biomolecular , Oxirredução , Oxigênio/metabolismo , Oxiemoglobinas/metabolismo , Estrutura Secundária de Proteína
20.
Biochim Biophys Acta ; 1834(9): 1824-31, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23459127

RESUMO

We have investigated CO migration and binding in CuBMb, a copper-binding myoglobin double mutant (L29H-F43H), by using Fourier transform infrared spectroscopy and flash photolysis over a wide temperature range. This mutant was originally engineered with the aim to mimic the catalytic site of heme-copper oxidases. Comparison of the wild-type protein Mb and CuBMb shows that the copper ion in the distal pocket gives rise to significant effects on ligand binding to the heme iron. In Mb and copper-free CuBMb, primary and secondary ligand docking sites are accessible upon photodissociation. In copper-bound CuBMb, ligands do not migrate to secondary docking sites but rather coordinate to the copper ion. Ligands entering the heme pocket from the outside normally would not be captured efficiently by the tight distal pocket housing the two additional large imidazole rings. Binding at the Cu ion, however, ensures efficient trapping in CuBMb. The Cu ion also restricts the motions of the His64 side chain, which is the entry/exit door for ligand movement into the active site, and this restriction results in enhanced geminate and slow bimolecular CO rebinding. These results support current mechanistic views of ligand binding in hemoglobins and the role of the CuB in the active of heme-copper oxidases. This article is part of a Special Issue entitled: Oxygen Binding and Sensing Proteins.


Assuntos
Monóxido de Carbono/metabolismo , Cobre/metabolismo , Heme/metabolismo , Mioglobina/metabolismo , Animais , Sítios de Ligação , Monóxido de Carbono/química , Domínio Catalítico , Cobre/química , Heme/química , Cinética , Masculino , Modelos Moleculares , Mutação/genética , Mioglobina/química , Mioglobina/genética , Fotólise , Ligação Proteica , Espectroscopia de Infravermelho com Transformada de Fourier , Temperatura , Baleias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...