Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 371
Filtrar
1.
Int J Mol Sci ; 20(22)2019 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-31744112

RESUMEN

The presence of hemoglobin A-S (HbAS) in erythrocytes has been related to the high production of reactive oxygen species (ROS) and an increased in intracellular oxidative stress that affects the progress of Plasmodium erythrocytic cycle life and attenuates its serious clinical symptoms. Nevertheless, oxidative effects on P. falciparum proteome across the intraerythrocytic cycle in the presence of HbAS traits have not been described yet. Here, an immune dot-blot assay was used to quantify the carbonyl index (C.I) on P. falciparum 3D7 proteome at the different asexual erythrocytic stages. Protein carbonylation on parasites cultivated in erythrocytes from two donors with HbAS increased 5.34 ± 1.42 folds at the ring stage compared to control grown in hemoglobin A-A (HbAA) red blood cells. Whereas at trophozoites and schizonts stages were augmented 2.80 ± 0.52 and 3.05 ± 0.75 folds, respectively. Besides proteins involved in processes of the stress response, recognition and invasion were identified from schizonts carbonylated bands by combining SDS-PAGE with MALDI-TOF-TOF analysis. Our results reinforce the hypothesis that such oxidative modifications do not appear to happen randomly, and the sickle cell trait affects mainly a small fraction of parasite proteins particularly sensitive to ROS.


Asunto(s)
Eritrocitos/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Proteoma/análisis , Rasgo Drepanocítico/patología , Electroforesis en Gel de Poliacrilamida , Eritrocitos/parasitología , Hemoglobina A/química , Hemoglobina A/metabolismo , Hemoglobina Falciforme/química , Hemoglobina Falciforme/metabolismo , Humanos , Estadios del Ciclo de Vida , Estrés Oxidativo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Carbonilación Proteica , Proteoma/metabolismo , Proteínas Protozoarias/análisis , Proteínas Protozoarias/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
2.
Biochemistry ; 56(10): 1444-1459, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28218841

RESUMEN

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.


Asunto(s)
Apoproteínas/química , Hemoglobina Fetal/química , Guanidina/química , Hemoglobina A/química , Hemoglobinas/química , Apoproteínas/genética , Apoproteínas/metabolismo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Expresión Génica , Glicina/química , Glicina/metabolismo , Hemo/química , Hemo/aislamiento & purificación , Hemo/metabolismo , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Cinética , Desnaturalización Proteica , Dominios Proteicos , Pliegue de Proteína , Multimerización de Proteína , Estabilidad Proteica , Estructura Secundaria de Proteína , Desplegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
3.
Proc Natl Acad Sci U S A ; 111(35): 12758-63, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25139985

RESUMEN

Monod, Wyman, and Changeux (MWC) explained allostery in multisubunit proteins with a widely applied theoretical model in which binding of small molecules, so-called allosteric effectors, affects reactivity by altering the equilibrium between more reactive (R) and less reactive (T) quaternary structures. In their model, each quaternary structure has a single reactivity. Here, we use silica gels to trap protein conformations and a new kind of laser photolysis experiment to show that hemoglobin, the paradigm of allostery, exhibits two ligand binding phases with the same fast and slow rates in both R and T quaternary structures. Allosteric effectors change the fraction of each phase but not the rates. These surprising results are readily explained by the simplest possible extension of the MWC model to include a preequilibrium between two tertiary conformations that have the same functional properties within each quaternary structure. They also have important implications for the long-standing question of a structural explanation for the difference in hemoglobin oxygen affinity of the two quaternary structures.


Asunto(s)
Hemoglobina A/química , Hemoglobina A/metabolismo , Hemoglobinas/química , Hemoglobinas/metabolismo , Modelos Químicos , Regulación Alostérica , Sitio Alostérico , Humanos , Rayos Láser , Ligandos , Oxígeno/química , Oxígeno/metabolismo , Fotólisis , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Gel de Sílice/química , Gel de Sílice/metabolismo
4.
J Biol Chem ; 290(1): 99-117, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25371199

RESUMEN

The giant extracellular hemoglobin (erythrocruorin) from the earth worm (Lumbricus terrestris) has shown promise as a potential hemoglobin-based oxygen carrier (HBOC) in in vivo animal studies. An important beneficial characteristic of this hemoglobin (LtHb) is the large number of heme-based oxygen transport sites that helps overcome issues of osmotic stress when attempting to provide enough material for efficient oxygen delivery. A potentially important additional property is the capacity of the HBOC either to generate nitric oxide (NO) or to preserve NO bioactivity to compensate for decreased levels of NO in the circulation. The present study compares the NO-generating and NO bioactivity-preserving capability of LtHb with that of human adult hemoglobin (HbA) through several reactions including the nitrite reductase, reductive nitrosylation, and still controversial nitrite anhydrase reactions. An assignment of a heme-bound dinitrogen trioxide as the stable intermediate associated with the nitrite anhydrase reaction in both LtHb and HbA is supported based on functional and EPR spectroscopic studies. The role of the redox potential as a factor contributing to the NO-generating activity of these two proteins is evaluated. The results show that LtHb undergoes the same reactions as HbA and that the reduced efficacy for these reactions for LtHb relative to HbA is consistent with the much higher redox potential of LtHb. Evidence of functional heterogeneity in LtHb is explained in terms of the large difference in the redox potential of the isolated subunits.


Asunto(s)
Sustitutos Sanguíneos/química , Hemoglobinas/química , Óxido Nítrico/química , Nitritos/química , Subunidades de Proteína/química , Animales , Sustitutos Sanguíneos/aislamiento & purificación , Hemoglobina A/química , Hemoglobina A/aislamiento & purificación , Hemoglobinas/aislamiento & purificación , Humanos , Cinética , Nitrito Reductasas/química , Óxidos de Nitrógeno/química , Oligoquetos/química , Oxidación-Reducción , Unión Proteica , Subunidades de Proteína/aislamiento & purificación , Soluciones
5.
Int J Mol Sci ; 17(4): 519, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27070578

RESUMEN

In this paper we review recent advances in computational chemistry and specifically focus on the chemical description of heme proteins and synthetic porphyrins that act as both mimics of natural processes and technological uses. These are challenging biochemical systems involved in electron transfer as well as biocatalysis processes. In recent years computational tools have improved considerably and now can reproduce experimental spectroscopic and reactivity studies within a reasonable error margin (several kcal·mol(-1)). This paper gives recent examples from our groups, where we investigated heme and synthetic metal-porphyrin systems. The four case studies highlight how computational modelling can correctly reproduce experimental product distributions, predicted reactivity trends and guide interpretation of electronic structures of complex systems. The case studies focus on the calculations of a variety of spectroscopic features of porphyrins and show how computational modelling gives important insight that explains the experimental spectra and can lead to the design of porphyrins with tuned properties.


Asunto(s)
Hemo/química , Porfirinas/química , Animales , Sistema Enzimático del Citocromo P-450/química , Citocromo-c Peroxidasa/química , Complejo IV de Transporte de Electrones/química , Electrones , Hemoproteínas/química , Hemoglobina A/química , Humanos , Metaloporfirinas/química , Modelos Moleculares , Conformación Proteica , Teoría Cuántica
6.
Biochemistry ; 54(34): 5268-78, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26226318

RESUMEN

Atomistic molecular dynamics simulations of diffusion of O2 from the hemes to the external solvent in the α- and ß-subunits of the human hemoglobin (HbA) tetramer reveal transient gas tunnels that are not seen in crystal structures. We find here that the tunnel topology, which encompasses the reported experimental Xe binding cavities, is identical in HbA's T, R, and R2 quaternary states. However, the O2 population in the cavities and the preferred O2 escape portals vary significantly with quaternary structure. For example, most O2 molecules escape from the T ß-subunit via the cavity at the center of the tetramer, but direct exit from the distal heme pocket dominates in the R2 ß-subunit. To understand what triggers the quaternary-linked redistribution of O2 within its tunnels, we examined how the simulated tertiary structure and dynamics of each subunit differs among T, R, and R2 and report that minor adjustments in α-chain dynamics and ß-heme position modulate O2 distribution and escape in HbA. Coupled to the ß-heme position, residue ßF71 undergoes quaternary-linked conformations that strongly regulate O2 migration between the ß-subunit and HbA's central cavity. Remarkably, the distal histidine (HisE7) remains in a closed conformation near the α- and ß-hemes in all states, but this does not prevent an average of 23, 31, and 46% of O2 escapes from the distal heme pockets of T, R, and R2, respectively, via several distal portals, with the balance of escapes occurring via the interior tunnels. Furthermore, preventing or restricting the access of O2 to selected cavities by mutating HisE7 and other heme pocket residues to tryptophan reveals how O2 migration adjusts to the bulky indole ring and sheds light on the experimental ligand binding kinetics of these variants. Overall, our simulations underscore the high gas porosity of HbA in its T, R, and R2 quaternary states and provide new mechanistic insights into why undergoing transitions among these states likely ensures effective O2 delivery by this tetrameric protein.


Asunto(s)
Hemoglobina A/química , Hemoglobina A/metabolismo , Oxígeno/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Difusión , Hemo/química , Humanos , Simulación de Dinámica Molecular , Análisis de Componente Principal , Conformación Proteica , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Subunidades de Proteína
7.
Biochemistry ; 54(34): 5279-89, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26226401

RESUMEN

Hemoglobin transports O2 by binding the gas at its four hemes. Hydrogen bonding between the distal histidine (HisE7) and heme-bound O2 significantly increases the affinity of human hemoglobin (HbA) for this ligand. HisE7 is also proposed to regulate the release of O2 to the solvent via a transient E7 channel. To reveal the O2 escape routes controlled by HisE7 and to evaluate its role in gating heme access, we compare simulations of O2 diffusion from the distal heme pockets of the T and R states of HbA performed with HisE7 in its open (protonated) and closed (neutral) conformations. Irrespective of HisE7's conformation, we observe the same four or five escape routes leading directly from the α- or ß-distal heme pockets to the solvent. Only 21-53% of O2 escapes occur via these routes, with the remainder escaping through routes that encompass multiple internal cavities in HbA. The conformation of the distal HisE7 controls the escape of O2 from the heme by altering the distal pocket architecture in a pH-dependent manner, not by gating the E7 channel. Removal of the HisE7 side chain in the GlyE7 variant exposes the distal pockets to the solvent, and the percentage of O2 escapes to the solvent directly from the α- or ß-distal pockets of the mutant increases to 70-88%. In contrast to O2, the dominant water route from the bulk solvent is gated by HisE7 because protonation and opening of this residue dramatically increase the rate of influx of water into the empty distal heme pockets. The occupancy of the distal heme site by a water molecule, which functions as an additional nonprotein barrier to binding of the ligand to the heme, is also controlled by HisE7. Overall, analysis of gas and water diffusion routes in the subunits of HbA and its GlyE7 variant sheds light on the contribution of distal HisE7 in controlling polar and nonpolar ligand movement between the solvent and the hemes.


Asunto(s)
Hemoglobina A/química , Hemoglobina A/metabolismo , Sustitución de Aminoácidos , Sitios de Unión , Difusión , Hemo/química , Hemoglobina A/genética , Histidina/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Oxígeno/metabolismo , Conformación Proteica , Subunidades de Proteína , Solventes , Agua/metabolismo
8.
J Biol Chem ; 289(32): 22342-57, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24939847

RESUMEN

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.


Asunto(s)
Hemo/metabolismo , Hemoglobinas/química , Hemoglobinas/metabolismo , Hierro/metabolismo , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Ácido Aspártico/química , Cristalografía por Rayos X , Hemoglobina Fetal/química , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Hemo/química , Hemoglobina A/química , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobinas/genética , Hemoglobinas Anormales/química , Hemoglobinas Anormales/genética , Hemoglobinas Anormales/metabolismo , Humanos , Peróxido de Hidrógeno/metabolismo , Hierro/química , Metionina/química , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Oxidación-Reducción , Procesamiento Proteico-Postraduccional , Subunidades de Proteína , Proteómica , Electricidad Estática
9.
Biochim Biophys Acta ; 1844(7): 1201-7, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24747784

RESUMEN

As a potential hemoglobin (Hb)-based oxygen carrier (HBOC), the PEGylated Hb has received much attention for its non-nephrotoxicity. However, PEGylation can adversely alter the structural and functional properties of Hb. The site of PEGylation is an important factor to determine the structure and function of the PEGylated Hb. Thus, protection of some sensitive residues of Hb from PEGylation is of great significance to develop the PEGylated Hb as HBOC. Here, Cys-93(ß) of Hb was conjugated with 20 kDa polyethylene glycol (PEG20K) through hydrazone and disulfide bonds. Then, the conjugate was modified with PEG5K succinimidyl carbonate (PEG5K-SC) using acylation chemistry, followed by removal of PEG20K Hb with hydrazone hydrolysis and disulfide reduction. Reversible conjugation of PEG20K at Cys-93(ß) can protect Lys-95(ß), Val-1(α) and Lys-16(α) of Hb from PEGylation with PEG5K-SC. The autoxidation rate, oxygen affinity, structural perturbation and tetramer instability of the PEGylated Hb were significantly decreased upon protection with PEG20K. The present study is expected to improve the efficacy of the PEGylated Hb as an oxygen therapeutic.


Asunto(s)
Cisteína/química , Hemoglobina A/química , Hemoglobina A/metabolismo , Oxígeno/metabolismo , Polietilenglicoles/química , Cromatografía en Gel , Cromatografía Líquida de Alta Presión , Dicroismo Circular , Ultracentrifugación
10.
Biochim Biophys Acta ; 1840(1): 277-87, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24060751

RESUMEN

BACKGROUND: AHSP is an erythroid molecular chaperone of the α-hemoglobin chains (α-Hb). Upon AHSP binding, native ferric α-Hb undergoes an unprecedented structural rearrangement at the heme site giving rise to a 6th coordination bond with His(E7). METHODS: Recombinant AHSP, WT α-Hb:AHSP and α-Hb(HE7Q):AHSP complexes were expressed in Escherichia coli. Thermal denaturation curves were measured by circular dichroism for the isolated α-Hb and bound to AHSP. Kinetics of ligand binding and redox reactions of α-Hb bound to AHSP as well as α-Hb release from the α-Hb:AHSP complex were measured by time-resolved absorption spectroscopy. RESULTS: AHSP binding to α-Hb is kinetically controlled to prevail over direct binding with ß-chains and is also thermodynamically controlled by the α-Hb redox state and not the liganded state of the ferrous α-Hb. The dramatic instability of isolated ferric α-Hb is greatly decreased upon AHSP binding. Removing the bis-histidyl hexacoordination in α-HbH58(E7)Q:AHSP complex reduces the stabilizing effect of AHSP binding. Once the ferric α-Hb is bound to AHSP, the globin can be more easily reduced by several chemical and enzymatic systems compared to α-Hb within the Hb-tetramer. CONCLUSION: α-Hb reduction could trigger its release from AHSP toward its final Hb ß-chain partner producing functional ferrous Hb-tetramers. This work indicates a preferred kinetic pathway for Hb-synthesis. GENERAL SIGNIFICANCE: The cellular redox balance in Hb-synthesis should be considered as important as the relative proportional synthesis of both Hb-subunits and their heme cofactor. The in vivo role of AHSP is discussed in the context of the molecular disorders observed in thalassemia.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Hemo/metabolismo , Hemoglobina A/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Monóxido de Carbono/metabolismo , Dicroismo Circular , Hemo/química , Hemoglobina A/química , Humanos , Cinética , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Oxígeno/metabolismo , Unión Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
11.
Lasers Med Sci ; 30(1): 247-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25217409

RESUMEN

Raman spectroscopy has been proposed as a tool for diagnosis of human blood diseases aiming a quick and accurate diagnosis. Sickle cell disease arises in infancy and causes a severe anemia; thus, an early diagnosis may avoid pathological complications such as vasoocclusion, hemolytic anemia, retinopathy, cardiovascular disease, and infections. This work evaluated spectral differences between hemoglobin S (HbS) and hemoglobin A (HbA) to be used in a diagnostic model based on principal components analysis. Blood samples of patients with a previous diagnosis of sickle cell disease were hemolyzed with water, centrifuged, and the pellet was collected with a pipette. Near-infrared Raman spectra (830 nm, 200 mW) were obtained from these samples, and a model based on principal components analysis and Mahalanobis distance were used to discriminate HbA from HbS. Differences were found in the spectra of HbS and HbA, mainly in the 882 and 1,373 cm(-1) (valine, HbA) and 1,547 and 1,622 cm(-1) (glutamic acid, HbS). The spectral model could correctly discriminate 100% of the samples in the correspondent groups. Raman spectroscopy was able to detect the subtle changes in the polypeptide chain (valine and glutamic acid substitution) due to the sickle cell disease and could be used to discriminate blood samples with HbS from HbA with minimum sample preparations (hemolysis with water and centrifugation).


Asunto(s)
Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/diagnóstico , Hemoglobina A/química , Hemoglobina Falciforme/química , Espectrometría Raman , Adulto , Interpretación Estadística de Datos , Femenino , Hemólisis , Humanos , Masculino , Análisis de Componente Principal , Reproducibilidad de los Resultados , Adulto Joven
12.
Biochemistry ; 53(43): 6793-9, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25325574

RESUMEN

Cross-linked human hemoglobins have been evaluated for clinical use as circulating oxygen carriers. However, their induction of vasoactivity was sufficiently problematic to lead to the cessation of clinical trials. The source of vasoactivity is likely to be endothelial extravasation causing the scavenging of endogenous nitric oxide. It was recently shown that species that consist of two coupled hemoglobin tetramers do not evoke vasoactivity in a sensitive murine model. Presumably these materials are too large to extravasate. In order to make this class of material more readily available, there is a need for improved methods that can form a cross-linked bis-tetramer without producing smaller species at the same time. A potentially efficient route to cross-linking and coupling two Hb tetramers is through phase-directed copper-catalyzed azide alkyne cycloaddition (PDCuAAC). However, introduction of the necessary azide-containing cross-link gives mixtures of tetrameric and bis-tetrameric proteins, as the PDCuAAC process appears to be limited to only those proteins where the cross-link containing the azide is exclusively within the ß-subunits. In order to block formation of the azide cross-link within the α-subunits, subunit-specific introduction of the azide is necessary. This is achieved by blocking reaction at the reactive amino groups of the ß-subunits in the site that binds the allosteric activator 2,3-diphosphoglycerate (DPG) with inositol hexaphosphate (IHP), permitting α-selective acetylation with acetyl 3,5-dibromosalicylate. After removal of IHP, reaction with an anionic cross-linker containing an azide group occurs within the ß-subunits. The resulting α-acetylated ß-ß'-cross-linked hemoglobin azide (acHb>-N3) undergoes efficient PDCuAAC with bis-alkynes to produce cross-linked bis-tetramers. Analysis of circular dichroism spectra of the modified species shows that there is little change in the structure of the globin chains as a result of the chemical modifications. The oxygenation properties are consistent with those needed for effective oxygenation in circulation, while the bis-tetrameric structure is sufficiently large to avoid extravasation and depletion of nitric oxide.


Asunto(s)
2,3-Difosfoglicerato/química , Hemoglobina A/química , Ácido Fítico/química , Multimerización de Proteína , Acetilación , Humanos , Estructura Cuaternaria de Proteína
13.
Biochemistry ; 53(15): 2474-82, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24670063

RESUMEN

Dehaloperoxidase hemoglobin A (DHP A) is a multifunctional hemoglobin that appears to have evolved oxidative pathways for the degradation of xenobiotics as a protective function that complements the oxygen transport function. DHP A possesses at least two internal binding sites, one for substrates and one for inhibitors, which include various halogenated phenols and indoles. Herein, we report the X-ray crystallographic structure of the carbonmonoxy complex (DHPCO). Unlike other DHP structures with 6-coordinated heme, the conformation of the distal histidine (H55) in DHPCO is primarily external or solvent exposed, despite the fact that the heme Fe is 6-coordinated. As observed generally in globins, DHP exhibits two distal histidine conformations (one internal and one external). In previous structural studies, we have shown that the distribution of H55 conformations is weighted strongly toward the external position when the DHP heme Fe is 5-coordinated. The large population of the external conformation of the distal histidine observed in DHPCO crystals at pH 6.0 indicates that some structural factor in DHP must account for the difference from other globins, which exhibit a significant external conformation only when pH < 4.5. While the original hypothesis suggested that interaction with a heme-Fe-bound ligand was the determinant of H55 conformation, the current study forces a refinement of that hypothesis. The external or open conformation of H55 is observed to have interactions with two propionate groups in heme, at distances of 3.82 and 2.73 Å, respectively. A relatively weak hydrogen bonding interaction between H55 and CO, combined with strong interactions with heme propionate (position 6), is hypothesized to strengthen the external conformation of H55. Density function theory (DFT) calculations were conducted to test whether there is a weaker hydrogen bond interaction between H55 and heme bonded CO or O2. Molecular dynamics simulations were conducted to examine how the tautomeric forms of H55 affect the dynamic motions of the distal histidine that govern the switching between open and closed conformations. The calculations support the modified hypothesis suggesting a competition between the strength of interactions with heme ligand and the heme propionates as the factors that determine the conformation of the distal histidine.


Asunto(s)
Monóxido de Carbono/química , Hemoglobina A/química , Hemoglobinas/química , Histidina/química , Peroxidasas/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Datos de Secuencia Molecular , Conformación Proteica
14.
J Biol Chem ; 288(27): 19986-20001, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23696640

RESUMEN

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.


Asunto(s)
Proteínas Sanguíneas/química , Hemoglobina A/química , Hierro/química , Chaperonas Moleculares/química , Oxígeno/química , Oxihemoglobinas/química , Sitios de Unión , Proteínas Sanguíneas/metabolismo , Hemoglobina A/metabolismo , Humanos , Hierro/metabolismo , Chaperonas Moleculares/metabolismo , Resonancia Magnética Nuclear Biomolecular , Oxidación-Reducción , Oxígeno/metabolismo , Oxihemoglobinas/metabolismo , Estructura Secundaria de Proteína
15.
J Biol Chem ; 288(6): 4288-98, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23264625

RESUMEN

α-Hemoglobin stabilizing protein (AHSP) is a molecular chaperone that binds monomeric α-subunits of human hemoglobin A (HbA) and modulates heme iron oxidation and subunit folding states. Although AHSP·αHb complexes autoxidize more rapidly than HbA, the redox mechanisms appear to be similar. Both metHbA and isolated met-ß-subunits undergo further oxidation in the presence of hydrogen peroxide (H(2)O(2)) to form ferryl heme species. Surprisingly, much lower levels of H(2)O(2)-induced ferryl heme are produced by free met-α-subunits as compared with met-ß-subunits, and no ferryl heme is detected in H(2)O(2)-treated AHSP·met-α-complex at pH values from 5.0 to 9.0 at 23 °C. Ferryl heme species were similarly not detected in AHSP·met-α Pro-30 mutants known to exhibit different rates of autoxidation and hemin loss. EPR data suggest that protein-based radicals associated with the ferryl oxidation state exist within HbA α- and ß-subunits. In contrast, treatment of free α-subunits with H(2)O(2) yields much smaller radical signals, and no radicals are detected when H(2)O(2) is added to AHSP·α-complexes. AHSP binding also dramatically reduces the redox potential of α-subunits, from +40 to -78 mV in 1 m glycine buffer, pH 6.0, at 8 °C, demonstrating independently that AHSP has a much higher affinity for Fe(III) versus Fe(II) α-subunits. Hexacoordination in the AHSP·met-α complex markedly decreases the rate of the initial H(2)O(2) reaction with iron and thus provides α-subunits protection against damaging oxidative reactions.


Asunto(s)
Proteínas Sanguíneas/química , Hemoglobina A/química , Peróxido de Hidrógeno/química , Metahemoglobina/química , Chaperonas Moleculares/química , Complejos Multiproteicos/química , Proteínas Sanguíneas/metabolismo , Hemoglobina A/metabolismo , Humanos , Peróxido de Hidrógeno/farmacología , Concentración de Iones de Hidrógeno , Metahemoglobina/metabolismo , Chaperonas Moleculares/metabolismo , Complejos Multiproteicos/metabolismo , Oxidantes/química , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos
16.
J Am Chem Soc ; 136(29): 10325-39, 2014 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-24991732

RESUMEN

The use of hybrid hemoglobin (Hb), with mesoheme substituted for protoheme, allows separate monitoring of the α or ß hemes along the allosteric pathway. Using resonance Raman (rR) spectroscopy in silica gel, which greatly slows protein motions, we have observed that the Fe-histidine stretching frequency, νFeHis, which is a monitor of heme reactivity, evolves between frequencies characteristic of the R and T states, for both α or ß chains, prior to the quaternary R-T and T-R shifts. Computation of νFeHis, using QM/MM and the conformational search program PELE, produced remarkable agreement with experiment. Analysis of the PELE structures showed that the νFeHis shifts resulted from heme distortion and, in the α chain, Fe-His bond tilting. These results support the tertiary two-state model of ligand binding (Henry et al., Biophys. Chem. 2002, 98, 149). Experimentally, the νFeHis evolution is faster for ß than for α chains, and pump-probe rR spectroscopy in solution reveals an inflection in the νFeHis time course at 3 µs for ß but not for α hemes, an interval previously shown to be the first step in the R-T transition. In the α chain νFeHis dropped sharply at 20 µs, the final step in the R-T transition. The time courses are fully consistent with recent computational mapping of the R-T transition via conjugate peak refinement by Karplus and co-workers (Fischer et al., Proc. Natl. Acad. Sci. U. S. A. 2011, 108, 5608). The effector molecule IHP was found to lower νFeHis selectively for α chains within the R state, and a binding site in the α1α2 cleft is suggested.


Asunto(s)
Biología Computacional/métodos , Hemo/química , Hemoglobina A/química , Espectrometría Raman/métodos , Globinas alfa/química , Globinas beta/química , Regulación Alostérica , Mesoporfirinas/química , Modelos Moleculares , Estructura Cuaternaria de Proteína
17.
Biochemistry ; 52(12): 2108-17, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23458680

RESUMEN

The clam Scapharca inaequivalvis possesses two cooperative oxygen binding hemoglobins in its red cells: a homodimeric HbI and a heterotetrameric A2B2 HbII. Each AB dimeric half of HbII is assembled in a manner very similar to that of the well-studied HbI. This study presents crystal structures of HbII along with oxygen binding data both in the crystalline state and in wet nanoporous silica gels. Despite very similar ligand-linked structural transitions observed in HbI and HbII crystals, HbII in the crystal or encapsulated in silica gels apparently exhibits minimal cooperativity in oxygen binding, in contrast with the full cooperativity exhibited by HbI crystals. However, oxygen binding curves in the crystal indicate the presence of a significant functional inequivalence of A and B chains. When this inequivalence is taken into account, both crystal and R state gel functional data are consistent with the conservation of a tertiary contribution to cooperative oxygen binding, quantitatively similar to that measured for HbI, and are in keeping with the structural information. Furthermore, our results indicate that to fully express cooperative ligand binding, HbII requires quaternary transitions hampered by crystal lattice and gel encapsulation, revealing greater complexity in cooperative function than the direct communication across a dimeric interface observed in HbI.


Asunto(s)
Hemoglobinas/química , Hemoglobinas/metabolismo , Scapharca/metabolismo , Animales , Monóxido de Carbono/metabolismo , Cristalografía por Rayos X , Hemoglobina A/química , Humanos , Cinética , Ligandos , Modelos Moleculares , Oxígeno/metabolismo , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
18.
Biochemistry ; 52(34): 5809-20, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23901897

RESUMEN

The solution structure of human adult carbonmonoxy hemoglobin (HbCO A) was refined using stereospecifically assigned methyl groups and residual dipolar couplings based on our previous nuclear magnetic resonance structure. The tertiary structures of individual chains were found to be very similar to the X-ray structures, while the quaternary structures in solution at low salt concentrations resembled the X-ray R structure more than the R2 structure. On the basis of chemical shift perturbation by inositol hexaphosphate (IHP) titration and docking, we identified five possible IHP binding sites in HbCO A. Amide-water proton exchange experiments demonstrated that αThr38 located in the α1ß2 interface and several loop regions in both α- and ß-chains were dynamic on the subsecond time scale. Side chain methyl dynamics revealed that methyl groups in the α1ß2 interface were dynamic, but those in the α1ß1 interface were quite rigid on the nanosecond to picosecond and millisecond to microsecond time scales. All the data strongly suggest a dynamic α1ß2 interface that allows conformational changes among different forms (like T, R, and R2) easily in solution. Binding of IHP to HbCO A induced small structural and dynamic changes in the α1ß2 interface and the regions around the hemes but did not increase the conformational entropy of HbCO A. The binding also caused conformational changes on the millisecond time scale, very likely arising from the relative motion of the α1ß1 dimer with respect to the α2ß2 dimer. Heterotropic effectors like IHP may change the oxygen affinity of Hb through modulating the relative motion of the two dimers and then further altering the structure of heme binding regions.


Asunto(s)
Carboxihemoglobina/química , Hemoglobina A/química , Adulto , Carboxihemoglobina/efectos de los fármacos , Hemo/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Ácido Fítico/metabolismo , Ácido Fítico/farmacología , Conformación Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Soluciones
19.
Biochemistry ; 52(47): 8539-55, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24224786

RESUMEN

The Bohr effect in hemoglobin, which refers to the dependence of the oxygen affinity on the pH, plays an important role in its cooperativity and physiological function. The dominant contribution to the Bohr effect arises from the difference in the pKa values of His residues of the unliganded (deoxy) and liganded (carbonmonoxy) structures. Using recent high resolution structures, the residue pKa values corresponding to the two structures are calculated. The method is based on determining the electrostatic interactions between residues in the protein, relative to those of the residue in solution, by use of the linearized finite difference Poisson-Boltzmann equation and Monte Carlo sampling of protonation states. Given that good agreement is obtained with the available experimental values for the contribution of His residues in HbA to the Bohr effect, the calculated results are used to determine the atomic origin of the pKa shift between deoxy and carbonmonoxy HbA. The contributions to the pKa shift calculated by means of the linear response approximation show that the salt bridge involving His146 plays an important role in the alkaline Bohr effect, as suggested by Perutz but that other interactions are significant as well. A corresponding analysis is made for the contribution of His143 to the acid Bohr effect for which there is no proposed explanation. The method used is summarized and the program by which it is implemented is described in the Appendix .


Asunto(s)
Hemoglobina A/metabolismo , Histidina/metabolismo , Oxihemoglobinas/metabolismo , Carboxihemoglobina/química , Carboxihemoglobina/metabolismo , Biología Computacional/métodos , Bases de Datos de Proteínas , Hemoglobina A/química , Subunidades de Hemoglobina/química , Subunidades de Hemoglobina/metabolismo , Hemoglobinas/química , Hemoglobinas/metabolismo , Histidina/química , Humanos , Concentración de Iones de Hidrógeno , Cinética , Método de Montecarlo , Oxihemoglobinas/química , Distribución de Poisson , Conformación Proteica
20.
J Biol Chem ; 287(14): 11338-50, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22298770

RESUMEN

Human α-hemoglobin stabilizing protein (AHSP) is a conserved mammalian erythroid protein that facilitates the production of Hemoglobin A by stabilizing free α-globin. AHSP rapidly binds to ferrous α with association (k'(AHSP)) and dissociation (k(AHSP)) rate constants of ≈10 µm(-1) s(-1) and 0.2 s(-1), respectively, at pH 7.4 at 22 °C. A small slow phase was observed when AHSP binds to excess ferrous αCO. This slow phase appears to be due to cis to trans prolyl isomerization of the Asp(29)-Pro(30) peptide bond in wild-type AHSP because it was absent when αCO was mixed with P30A and P30W AHSP, which are fixed in the trans conformation. This slow phase was also absent when met(Fe(3+))-α reacted with wild-type AHSP, suggesting that met-α is capable of rapidly binding to either Pro(30) conformer. Both wild-type and Pro(30)-substituted AHSPs drive the formation of a met-α hemichrome conformation following binding to either met- or oxy(Fe(2+))-α. The dissociation rate of the met-α·AHSP complex (k(AHSP) ≈ 0.002 s(-1)) is ∼100-fold slower than that for ferrous α·AHSP complexes, resulting in a much higher affinity of AHSP for met-α. Thus, in vivo, AHSP acts as a molecular chaperone by rapidly binding and stabilizing met-α hemichrome folding intermediates. The low rate of met-α dissociation also allows AHSP to have a quality control function by kinetically trapping ferric α and preventing its incorporation into less stable mixed valence Hemoglobin A tetramers. Reduction of AHSP-bound met-α allows more rapid release to ß subunits to form stable fully, reduced hemoglobin dimers and tetramers.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Hemoproteínas/química , Chaperonas Moleculares/metabolismo , Pliegue de Proteína , Globinas alfa/química , Globinas alfa/metabolismo , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Hemoglobina A/química , Hemoglobina A/metabolismo , Humanos , Cinética , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Mutagénesis Sitio-Dirigida , Mutación , Oxidación-Reducción , Unión Proteica , Estabilidad Proteica , Estructura Secundaria de Proteína , Electricidad Estática , Especificidad por Sustrato
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda