Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 289(32): 22342-57, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24939847

ABSTRACT

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.


Subject(s)
Heme/metabolism , Hemoglobins/chemistry , Hemoglobins/metabolism , Iron/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Aspartic Acid/chemistry , Crystallography, X-Ray , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Heme/chemistry , Hemoglobin A/chemistry , Hemoglobin A/genetics , Hemoglobin A/metabolism , Hemoglobins/genetics , Hemoglobins, Abnormal/chemistry , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Humans , Hydrogen Peroxide/metabolism , Iron/chemistry , Methionine/chemistry , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Oxidation-Reduction , Protein Processing, Post-Translational , Protein Subunits , Proteomics , Static Electricity
2.
Free Radic Biol Med ; 69: 265-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24486321

ABSTRACT

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.


Subject(s)
Dimerization , Haptoglobins/genetics , Hemoglobins/genetics , Oxidation-Reduction , Electron Spin Resonance Spectroscopy , Haptoglobins/chemistry , Hemoglobins/chemistry , Humans , Hydrogen Peroxide/chemistry , Kinetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Phenotype , Polymers/chemistry
3.
J Biol Chem ; 288(27): 19986-20001, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23696640

ABSTRACT

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.


Subject(s)
Blood Proteins/chemistry , Hemoglobin A/chemistry , Iron/chemistry , Molecular Chaperones/chemistry , Oxygen/chemistry , Oxyhemoglobins/chemistry , Binding Sites , Blood Proteins/metabolism , Hemoglobin A/metabolism , Humans , Iron/metabolism , Molecular Chaperones/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Oxygen/metabolism , Oxyhemoglobins/metabolism , Protein Structure, Secondary
4.
Antioxid Redox Signal ; 18(17): 2251-3, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23330885

ABSTRACT

In the last several years, significant work has been done studying hemoglobin (Hb) oxidative reactions and clearance mechanisms using both in vitro and in vivo model systems. One active research area involves the study of molecular chaperones and other proteins that are thought to mitigate the toxicity of acellular Hb. For example, the plasma protein haptoglobin (Hp) and the pre-erythroid protein alpha-hemoglobin-stabilizing protein (AHSP) bind to acellular Hb and alpha-subunits of Hb, respectively, to reduce these adverse effects. Moreover, there has been significant work studying hemopexin and alpha-1 microglobulin, both of which are thought to be involved with hemin degradation. These studies have coincided with the timely publication of the first crystal structure of the Hb-Hp complex. In constructing this Forum, we have invited a number of researchers in the area of Hb and myoglobin (Mb) redox biochemistry, as well as those who have contributed fundamentally to our knowledge of Hp function. Our goal has been to update this critically important research area, because we believe that it will ultimately impact the practice of transfusion medicine in a number of important ways.


Subject(s)
Hemoglobins/physiology , Animals , Hemoglobins/chemistry , Humans , Oxidation-Reduction
5.
J Biol Chem ; 288(6): 4288-98, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23264625

ABSTRACT

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.


Subject(s)
Blood Proteins/chemistry , Hemoglobin A/chemistry , Hydrogen Peroxide/chemistry , Methemoglobin/chemistry , Molecular Chaperones/chemistry , Multiprotein Complexes/chemistry , Blood Proteins/metabolism , Hemoglobin A/metabolism , Humans , Hydrogen Peroxide/pharmacology , Hydrogen-Ion Concentration , Methemoglobin/metabolism , Molecular Chaperones/metabolism , Multiprotein Complexes/metabolism , Oxidants/chemistry , Oxidants/pharmacology , Oxidation-Reduction/drug effects
6.
Antioxid Redox Signal ; 18(17): 2314-28, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23025383

ABSTRACT

SIGNIFICANCE: The worldwide blood shortage has generated a significant demand for alternatives to whole blood and packed red blood cells for use in transfusion therapy. One such alternative involves the use of acellular recombinant hemoglobin (Hb) as an oxygen carrier. RECENT ADVANCES: Large amounts of recombinant human Hb can be expressed and purified from transgenic Escherichia coli. The physiological suitability of this material can be enhanced using protein-engineering strategies to address specific efficacy and toxicity issues. Mutagenesis of Hb can (i) adjust dioxygen affinity over a 100-fold range, (ii) reduce nitric oxide (NO) scavenging over 30-fold without compromising dioxygen binding, (iii) slow the rate of autooxidation, (iv) slow the rate of hemin loss, (v) impede subunit dissociation, and (vi) diminish irreversible subunit denaturation. Recombinant Hb production is potentially unlimited and readily subjected to current good manufacturing practices, but may be restricted by cost. Acellular Hb-based O(2) carriers have superior shelf-life compared to red blood cells, are universally compatible, and provide an alternative for patients for whom no other alternative blood products are available or acceptable. CRITICAL ISSUES: Remaining objectives include increasing Hb stability, mitigating iron-catalyzed and iron-centered oxidative reactivity, lowering the rate of hemin loss, and lowering the costs of expression and purification. Although many mutations and chemical modifications have been proposed to address these issues, the precise ensemble of mutations has not yet been identified. FUTURE DIRECTIONS: Future studies are aimed at selecting various combinations of mutations that can reduce NO scavenging, autooxidation, oxidative degradation, and denaturation without compromising O(2) delivery, and then investigating their suitability and safety in vivo.


Subject(s)
Blood Substitutes , Hemoglobins/genetics , Hemoglobins/metabolism , Oxygen/blood , Recombinant Proteins , Animals , Biological Transport , Heme/metabolism , Hemoglobins/chemistry , Humans , Nitric Oxide/metabolism , Oxidation-Reduction , Protein Binding , Protein Stability
7.
Protein Sci ; 21(10): 1444-55, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22821886

ABSTRACT

Hemoglobin Brigham (ß Pro100 to Leu) was first reported in a patient with familial erythrocytosis. Erythrocytes of an affected individual from the same family contain both HbA and Hb Brigham and exhibit elevated O2 affinity compared with normal cells (P50 = 23 mm Hg vs. 31 mmHg at pH 7.4 at 37°C). O2 affinities measured for hemolysates were sensitive to changes in pH or chloride concentrations, indicating little change in the Bohr and Chloride effects. Hb Brigham was separated from normal HbA by nondenaturing cation exchange liquid chromatography, and the amino acid substitution was verified by mass spectrometry. The properties of Hb Brigham isolated from the patient's blood were then compared with those of recombinant Hb Brigham expressed in Escherichia coli. Kinetic experiments suggest that the rate constants for ligand binding and release in the high (R) and low (T) affinity quaternary states of Hb Brigham are similar to those of native hemoglobin. However, the Brigham mutation decreases the T to R equilibrium constant (L) which accelerates the switch to the R state during ligand binding to deoxy-Hb, increasing the rate of association by approximately twofold, and decelerates the switch during ligand dissociation from HbO2, decreasing the rate approximately twofold. These kinetic data help explain the high O2 affinity characteristics of Hb Brigham and provide further evidence for the importance of the contribution of Pro100 to intersubunit contacts and stabilization of the T quaternary structure.


Subject(s)
Hemoglobins, Abnormal/metabolism , Oxygen/metabolism , Polycythemia/metabolism , Adult , Chromatography, Reverse-Phase , Female , Hemoglobins/chemistry , Hemoglobins/isolation & purification , Hemoglobins, Abnormal/chemistry , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/isolation & purification , Humans , Kinetics , Polycythemia/blood , Polycythemia/congenital , Polycythemia/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
J Biol Chem ; 287(14): 11338-50, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22298770

ABSTRACT

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.


Subject(s)
Blood Proteins/metabolism , Hemeproteins/chemistry , Molecular Chaperones/metabolism , Protein Folding , alpha-Globins/chemistry , alpha-Globins/metabolism , Blood Proteins/chemistry , Blood Proteins/genetics , Hemoglobin A/chemistry , Hemoglobin A/metabolism , Humans , Kinetics , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Mutagenesis, Site-Directed , Mutation , Oxidation-Reduction , Protein Binding , Protein Stability , Protein Structure, Secondary , Static Electricity , Substrate Specificity
9.
J Biol Chem ; 287(14): 11325-37, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22287545

ABSTRACT

α-Hemoglobin stabilizing protein (AHSP) is believed to facilitate adult Hemoglobin A assembly and protect against toxic free α-globin subunits. Recombinant AHSP binds multiple forms of free α-globin to stabilize their structures and inhibit precipitation. However, AHSP also stimulates autooxidation of αO(2) subunit and its rapid conversion to a partially unfolded bishistidyl hemichrome structure. To investigate these biochemical properties, we altered the evolutionarily conserved AHSP proline 30 in recombinantly expressed proteins and introduced identical mutations into the endogenous murine Ahsp gene. In vitro, the P30W AHSP variant bound oxygenated α chains with 30-fold increased affinity. Both P30W and P30A mutant proteins also caused decreased rates of αO(2) autooxidation as compared with wild-type AHSP. Despite these abnormalities, mice harboring P30A or P30W Ahsp mutations exhibited no detectable defects in erythropoiesis at steady state or during induced stresses. Further biochemical studies revealed that the AHSP P30A and P30W substitutions had minimal effects on AHSP interactions with ferric α subunits. Together, our findings indicate that the ability of AHSP to stabilize nascent α chain folding intermediates prior to hemin reduction and incorporation into adult Hemoglobin A is physiologically more important than AHSP interactions with ferrous αO(2) subunits.


Subject(s)
Hemoglobin A/chemistry , Molecular Chaperones/metabolism , Mutagenesis , Mutant Proteins/metabolism , Protein Multimerization , Amino Acid Sequence , Amino Acid Substitution , Animals , Erythropoiesis , Gene Knock-In Techniques , Hemin/metabolism , Hemoglobin A/metabolism , Humans , Methemoglobin/chemistry , Methemoglobin/metabolism , Mice , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutation , Oxidation-Reduction , Protein Conformation , Protein Multimerization/genetics , beta-Thalassemia/genetics
10.
N Engl J Med ; 364(19): 1837-43, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21561349

ABSTRACT

Globin-gene mutations are a rare but important cause of cyanosis. We identified a missense mutation in the fetal Gγ-globin gene (HBG2) in a father and daughter with transient neonatal cyanosis and anemia. This new mutation modifies the ligand-binding pocket of fetal hemoglobin by means of two mechanisms. First, the relatively large side chain of methionine decreases both the affinity of oxygen for binding to the mutant hemoglobin subunit and the rate at which it does so. Second, the mutant methionine is converted to aspartic acid post-translationally, probably through oxidative mechanisms. The presence of this polar amino acid in the heme pocket is predicted to enhance hemoglobin denaturation, causing anemia.


Subject(s)
Fetal Hemoglobin/genetics , Hemoglobins, Abnormal/genetics , Mutation, Missense , gamma-Globins/genetics , Anemia/genetics , Cyanosis/genetics , Female , Humans , Infant, Newborn , Male , Methemoglobin/biosynthesis , Oxygen/blood , Protein Conformation , Sequence Analysis, DNA
11.
Antioxid Redox Signal ; 12(2): 219-31, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19659437

ABSTRACT

Hemoglobin biosynthesis in erythrocyte precursors involves several steps. The correct ratios and concentrations of normal alpha (alpha) and beta (beta) globin proteins must be expressed; apoproteins must be folded correctly; heme must be synthesized and incorporated into these globins rapidly; and the individual alpha and beta subunits must be rapidly and correctly assembled into heterotetramers. These events occur on a large scale in vivo, and dysregulation causes serious clinical disorders such as thalassemia syndromes. Recent work has implicated a conserved erythroid protein known as Alpha-Hemoglobin Stabilizing Protein (AHSP) as a participant in these events. Current evidence suggests that AHSP enhances alpha subunit stability and diminishes its participation in harmful redox chemistry. There is also evidence that AHSP facilitates one or more early-stage post-translational hemoglobin biosynthetic events. In this review, recent experimental results are discussed in light of several current models describing globin subunit folding, heme uptake, assembly, and denaturation during hemoglobin synthesis. Particular attention is devoted to molecular interactions with AHSP that relate to alpha chain oxidation and the ability of alpha chains to associate with partner beta chains.


Subject(s)
Blood Proteins/metabolism , Hemoglobins/metabolism , Molecular Chaperones/metabolism , Animals , Blood Proteins/chemistry , Humans , Models, Biological , Molecular Chaperones/chemistry , Oxidation-Reduction , Protein Binding , Protein Structure, Secondary
12.
Blood ; 113(23): 5961-9, 2009 Jun 04.
Article in English | MEDLINE | ID: mdl-19349619

ABSTRACT

Alpha hemoglobin stabilizing protein (AHSP) reversibly binds nascent alpha globin to maintain its native structure and facilitate its incorporation into hemoglobin A. Previous studies indicate that some naturally occurring human alpha globin mutations may destabilize the protein by inhibiting its interactions with AHSP. However, these mutations could also affect hemoglobin A production through AHSP-independent effects, including reduced binding to beta globin. We analyzed 6 human alpha globin variants with altered AHSP contact surfaces. Alpha globin amino acid substitutions H103Y, H103R, F117S, and P119S impaired interactions with both AHSP and beta globin. These mutations are destabilizing in biochemical assays and are associated with microcytosis and anemia in humans. By contrast, K99E and K99N alpha globins bind beta globin normally but exhibit attenuated binding to AHSP. These mutations impair protein folding and expression in vitro and appear to be mildly destabilizing in vivo. In Escherichia coli and erythroid cells, alpha globin K99E stability is rescued on coexpression with AHSP mutants in which binding to the abnormal globin chain is restored. Our results better define the biochemical properties of some alpha globin variants and support the hypothesis that AHSP promotes alpha globin chain stability during human erythropoiesis.


Subject(s)
Blood Proteins/metabolism , DNA/analysis , DNA/genetics , Molecular Chaperones/metabolism , alpha-Globins/analysis , alpha-Globins/metabolism , Amino Acid Sequence , Blood Proteins/chemistry , DNA Mutational Analysis , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Models, Molecular , Molecular Chaperones/chemistry , Molecular Sequence Data , Mutation/genetics , Protein Folding , Protein Structure, Quaternary , alpha-Globins/chemistry , alpha-Globins/genetics , beta-Globins/genetics , beta-Globins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...