Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Biopolymers ; 112(6): e23427, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33792032

ABSTRACT

Hemoglobin III (HbIII) is one of the two oxygen reactive hemoproteins present in the bivalve, Lucina pectinata. The clam inhabits a sulfur-rich environment and HbIII is the only hemoprotein present in the system which does not yet have a structure described elsewhere. It is known that HbIII exists as a heterodimer with hemoglobin II (HbII) to generate the stable Oxy(HbII-HbIII) complex but it remains unknown if HbIII can form a homodimeric species. Here, a new chromatographic methodology to separate OxyHbIII from the HbII-HbIII dimer has been developed, employing a fast performance liquid chromatography and ionic exchange chromatography column. The nature of OxyHbIII in solution at concentrations from 1.6 mg/mL to 20.4 mg/mL was studied using small angle X-ray scattering (SAXS). The results show that at all concentrations, the Oxy(HbIII-HbIII) dimer dominates in solution. However, as the concentration increases to nonphysiological values, 20.4 mg/mL, HbIII forms a 30% tetrameric fraction. Thus, there is a direct relationship between the Oxy(HbIII-HbIII) oligomeric form and hemoglobin concentration. We suggest it is likely that the OxyHbIII dimer contributes to active oxygen transport in tissues of L pectinata, where the Oxy(HbII-HbIII) complex is not present.


Subject(s)
Bivalvia/metabolism , Oxyhemoglobins/chemistry , Protein Multimerization , Scattering, Small Angle , X-Ray Diffraction/methods , Amino Acid Sequence , Animals , Bivalvia/genetics , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Heme/chemistry , Heme/metabolism , Hydrogen Sulfide/metabolism , Oxyhemoglobins/genetics , Oxyhemoglobins/metabolism , Protein Conformation , Sequence Homology, Amino Acid , Tandem Mass Spectrometry/methods
2.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33753505

ABSTRACT

Dive capacities of air-breathing vertebrates are dictated by onboard O2 stores, suggesting that physiologic specialization of diving birds such as penguins may have involved adaptive changes in convective O2 transport. It has been hypothesized that increased hemoglobin (Hb)-O2 affinity improves pulmonary O2 extraction and enhances the capacity for breath-hold diving. To investigate evolved changes in Hb function associated with the aquatic specialization of penguins, we integrated comparative measurements of whole-blood and purified native Hb with protein engineering experiments based on site-directed mutagenesis. We reconstructed and resurrected ancestral Hb representing the common ancestor of penguins and the more ancient ancestor shared by penguins and their closest nondiving relatives (order Procellariiformes, which includes albatrosses, shearwaters, petrels, and storm petrels). These two ancestors bracket the phylogenetic interval in which penguin-specific changes in Hb function would have evolved. The experiments revealed that penguins evolved a derived increase in Hb-O2 affinity and a greatly augmented Bohr effect (i.e., reduced Hb-O2 affinity at low pH). Although an increased Hb-O2 affinity reduces the gradient for O2 diffusion from systemic capillaries to metabolizing cells, this can be compensated by a concomitant enhancement of the Bohr effect, thereby promoting O2 unloading in acidified tissues. We suggest that the evolved increase in Hb-O2 affinity in combination with the augmented Bohr effect maximizes both O2 extraction from the lungs and O2 unloading from the blood, allowing penguins to fully utilize their onboard O2 stores and maximize underwater foraging time.


Subject(s)
Adaptation, Physiological , Oxygen/metabolism , Oxyhemoglobins/metabolism , Spheniscidae/physiology , Amino Acid Substitution , Animals , Oxyhemoglobins/chemistry , Oxyhemoglobins/genetics , Phylogeny , Protein Conformation , Protein Engineering , Spheniscidae/blood , Spheniscidae/classification
3.
mSphere ; 5(6)2020 12 09.
Article in English | MEDLINE | ID: mdl-33298575

ABSTRACT

Streptococcus pneumoniae and other streptococci produce a greenish halo on blood agar plates referred to as alpha-hemolysis. This phenotype is utilized by clinical microbiology laboratories to report culture findings of alpha-hemolytic streptococci, including S. pneumoniae, and other bacteria. The alpha-hemolysis halo on blood agar plates has been related to the hemolytic activity of pneumococcal pneumolysin (Ply) or, to a lesser extent, to lysis of erythrocytes by S. pneumoniae-produced hydrogen peroxide. We investigated the molecular basis of the alpha-hemolysis halo produced by S. pneumoniae Wild-type strains TIGR4, D39, R6, and EF3030 and isogenic derivative Δply mutants produced similar alpha-hemolytic halos on blood agar plates, while cultures of hydrogen peroxide knockout ΔspxB ΔlctO mutants lacked this characteristic halo. Moreover, in the presence of catalase, the alpha-hemolysis halo was absent in cultures of the wild-type (wt) and Δply mutant strains. Spectroscopic studies demonstrated that culture supernatants of TIGR4 released hemoglobin-bound heme (heme-hemoglobin) from erythrocytes and oxidized oxy-hemoglobin to met-hemoglobin within 30 min of incubation. As expected, given Ply hemolytic activity and that hydrogen peroxide contributes to the release of Ply, TIGR4Δply and ΔspxB ΔlctO isogenic mutants had significantly decreased release of heme-hemoglobin from erythrocytes. However, TIGR4Δply that produces hydrogen peroxide oxidized oxy-hemoglobin to met-hemoglobin, whereas TIGR4ΔspxB ΔlctO failed to produce oxidation of oxy-hemoglobin. Studies conducted with all other wt strains and isogenic mutants resulted in similar findings. We demonstrated that the so-called alpha-hemolysis halo is caused by the oxidation of oxy-hemoglobin (Fe+2) to a non-oxygen-binding met-hemoglobin (Fe+3) by S. pneumoniae-produced hydrogen peroxide.IMPORTANCE There is a misconception that alpha-hemolysis observed on blood agar plate cultures of Streptococcus pneumoniae and other alpha-hemolytic streptococci is produced by a hemolysin or, alternatively, by lysis of erythrocytes caused by hydrogen peroxide. We noticed in the course of our investigations that wild-type S. pneumoniae strains and hemolysin (e.g., pneumolysin) knockout mutants produced the alpha-hemolytic halo on blood agar plates. In contrast, hydrogen peroxide-defective mutants prepared in four different strains lacked the characteristic alpha-hemolysis halo. We also demonstrated that wild-type strains and pneumolysin mutants oxidized oxy-hemoglobin to met-hemoglobin. Hydrogen peroxide knockout mutants, however, failed to oxidize oxy-hemoglobin. Therefore, the greenish halo formed on cultures of S. pneumoniae and other so-called alpha-hemolytic streptococci is caused by the oxidation of oxy-hemoglobin produced by hydrogen peroxide. Oxidation of oxy-hemoglobin to the nonbinding oxygen form, met-hemoglobin, might occur in the lungs during pneumococcal pneumonia.


Subject(s)
Hydrogen Peroxide/metabolism , Pneumococcal Infections/microbiology , Streptococcus pneumoniae/physiology , Streptolysins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Humans , Lung/microbiology , Oxyhemoglobins/genetics , Oxyhemoglobins/metabolism , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/growth & development , Streptolysins/genetics
4.
Am J Hum Genet ; 105(5): 1057-1068, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31668705

ABSTRACT

Average arterial oxyhemoglobin saturation during sleep (AvSpO2S) is a clinically relevant measure of physiological stress associated with sleep-disordered breathing, and this measure predicts incident cardiovascular disease and mortality. Using high-depth whole-genome sequencing data from the National Heart, Lung, and Blood Institute (NHLBI) Trans-Omics for Precision Medicine (TOPMed) project and focusing on genes with linkage evidence on chromosome 8p23,1,2 we observed that six coding and 51 noncoding variants in a gene that encodes the GTPase-activating protein (DLC1) are significantly associated with AvSpO2S and replicated in independent subjects. The combined DLC1 association evidence of discovery and replication cohorts reaches genome-wide significance in European Americans (p = 7.9 × 10-7). A risk score for these variants, built on an independent dataset, explains 0.97% of the AvSpO2S variation and contributes to the linkage evidence. The 51 noncoding variants are enriched in regulatory features in a human lung fibroblast cell line and contribute to DLC1 expression variation. Mendelian randomization analysis using these variants indicates a significant causal effect of DLC1 expression in fibroblasts on AvSpO2S. Multiple sources of information, including genetic variants, gene expression, and methylation, consistently suggest that DLC1 is a gene associated with AvSpO2S.


Subject(s)
Chromosomes, Human, Pair 8/genetics , GTPase-Activating Proteins/genetics , Oxyhemoglobins/genetics , Sleep/genetics , Tumor Suppressor Proteins/genetics , Genetic Linkage/genetics , Genome-Wide Association Study , Humans , Whole Genome Sequencing/methods
5.
Adv Exp Med Biol ; 1072: 221-225, 2018.
Article in English | MEDLINE | ID: mdl-30178349

ABSTRACT

Heme mediated oxidative toxicity has been linked to adverse side effects in Hemoglobin Based Oxygen Carriers (HBOC), initiated by reactive ferryl (FeIV) iron and globin based free radical species. We recently showed that the addition of a redox active tyrosine residue in the beta subunit (ßF41Y) of recombinant hemoglobin had the capability to decrease lipid peroxidation by facilitating the reduction of FeIV iron by plasma antioxidants such as ascorbate. In order to explore this functionality further we created a suite of tyrosine mutants designed to be accessible for both reductant access at the protein surface, yet close enough to the heme cofactor to enable efficient electron transfer to the FeIV. The residues chosen were: ßF41Y; ßK66Y; ßF71Y; ßT84Y; ßF85Y; and ßL96Y. As with ßF41Y, all mutants significantly enhanced the rate of ferryl (FeIV) to ferric (FeIII) reduction by ascorbate. However, surprisingly a subset of these mutations (ßT84Y, and ßF85Y) also enhanced the further reduction of ferric (FeIII) to ferrous (FeII) heme, regenerating functional oxyhemoglobin. The largest increase was seen in ßT84Y with the percentage of oxyhemoglobin formed from ferric hemoglobin in the presence of 100 µM ascorbate over a time period of 60 min increasing from 10% in ßF41Y to over 50% in ßT84Y. This increase was accompanied by an increased rate of ascorbate consumption. We conclude that the insertion of novel redox active tyrosine residues may be a useful component of any recombinant HBOC designed for longer functional activity without oxidative side effects.


Subject(s)
Blood Substitutes/chemistry , Blood Substitutes/metabolism , Methemoglobin/metabolism , Oxyhemoglobins/metabolism , Tyrosine/metabolism , Drug Design , Humans , Methemoglobin/genetics , Mutation , Oxidation-Reduction , Oxyhemoglobins/genetics , Tyrosine/genetics
6.
Hum Mol Genet ; 25(23): 5244-5253, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27798093

ABSTRACT

Genetic determinants of sleep-disordered breathing (SDB), a common set of disorders that contribute to significant cardiovascular and neuropsychiatric morbidity, are not clear. Overnight nocturnal oxygen saturation (SaO2) is a clinically relevant and easily measured indicator of SDB severity but its genetic contribution has never been studied. Our recent study suggests nocturnal SaO2 is heritable. We performed linkage analysis, association analysis and haplotype analysis of average nocturnal oxyhaemoglobin saturation in participants in the Cleveland Family Study (CFS), followed by gene-based association and additional tests in four independent samples. Linkage analysis identified a peak (LOD = 4.29) on chromosome 8p23. Follow-up association analysis identified two haplotypes in angiopoietin-2 (ANGPT2) that significantly contributed to the variation of SaO2 (P = 8 × 10-5) and accounted for a portion of the linkage evidence. Gene-based association analysis replicated the association of ANGPT2 and nocturnal SaO2. A rare missense SNP rs200291021 in ANGPT2 was associated with serum angiopoietin-2 level (P = 1.29 × 10-4), which was associated with SaO2 (P = 0.002). Our study provides the first evidence for the association of ANGPT2, a gene previously implicated in acute lung injury syndromes, with nocturnal SaO2, suggesting that this gene has a broad range of effects on gas exchange, including influencing oxygenation during sleep.


Subject(s)
Angiopoietin-2/genetics , Oxygen Consumption/genetics , Oxyhemoglobins/genetics , Sleep Apnea Syndromes/genetics , Adult , Female , Genetic Association Studies , Genetic Linkage , Genetic Predisposition to Disease , Haplotypes/genetics , Humans , Male , Oxygen/metabolism , Polymorphism, Single Nucleotide , Respiration/genetics , Sleep/genetics , Sleep Apnea Syndromes/metabolism , Sleep Apnea Syndromes/pathology
7.
Blood ; 128(23): 2708-2716, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27688804

ABSTRACT

Functional studies have shown that the oxygenation state of the erythrocyte regulates many important pathways, including glucose metabolism, membrane mechanical stability, and cellular adenosine triphosphate (ATP) release. Deoxyhemoglobin (deoxyHb), but not oxyhemoglobin, binds avidly and reversibly to band 3, the major erythrocyte membrane protein. Because band 3 associates with multiple metabolic, solute transport, signal transduction, and structural proteins, the hypothesis naturally arises that the O2-dependent regulation of erythrocyte properties might be mediated by the reversible association of deoxyHb with band 3. To explore whether the band 3-deoxyHb interaction constitutes a "molecular switch" for regulating erythrocyte biology, we have generated transgenic mice with mutations in the deoxyHb-binding domain of band 3. One strain of mouse contains a "humanized" band 3 in which the N-terminal 45 residues of mouse band 3 are replaced by the homologous sequence from human band 3, including the normal human band 3 deoxyHb-binding site. The second mouse contains the same substitution as the first, except the deoxyHb site on band 3 (residues 12-23) has been deleted. Comparison of these animals with wild-type mice demonstrates that the following erythrocyte properties are controlled by the O2-dependent association of hemoglobin with band 3: (1) assembly of a glycolytic enzyme complex on the erythrocyte membrane which is associated with a shift in glucose metabolism between the pentose phosphate pathway and glycolysis, (2) interaction of ankyrin with band 3 and the concomitant regulation of erythrocyte membrane stability, and (3) release of ATP from the red cell which has been linked to vasodilation.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Erythrocyte Membrane/metabolism , Oxygen/metabolism , Oxyhemoglobins/metabolism , Animals , Anion Exchange Protein 1, Erythrocyte/genetics , Erythrocyte Membrane/genetics , Glycolysis/physiology , Mice , Mice, Transgenic , Oxyhemoglobins/genetics , Pentose Phosphate Pathway/physiology
8.
J Am Soc Mass Spectrom ; 24(7): 997-1005, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23666601

ABSTRACT

The function of hemoglobin (Hb) as oxygen transporter is mediated by reversible O2 binding to Fe(2+) heme in each of the α and ß subunits. X-ray crystallography revealed different subunit arrangements in oxy-Hb and deoxy-Hb. The deoxy state is stabilized by additional contacts, causing a rigidification that results in strong protection against hydrogen/deuterium exchange (HDX). Aquomet-Hb is a dysfunctional degradation product with four water-bound Fe(3+) centers. Heme release from aquomet-Hb is relatively facile, triggering oxidative damage of membrane lipids. Aquomet-Hb crystallizes in virtually the same conformation as oxy-Hb. Hence, it is commonly implied that the solution-phase properties of aquomet-Hb should resemble those of the oxy state. This work compares the structural dynamics of oxy-Hb and aquomet-Hb by HDX mass spectrometry (MS). It is found that the aquomet state exhibits a solution-phase structure that is significantly more dynamic, as manifested by elevated HDX levels. These enhanced dynamics affect the aquomet α and ß subunits in a different fashion. The latter undergoes global destabilization, whereas the former shows elevated HDX levels only in the heme binding region. It is proposed that these enhanced dynamics play a role in facilitating heme release from aquomet-Hb. Our findings should be of particular interest to the MS community because oxy-Hb and aquomet-Hb serve as widely used test analytes for probing the relationship between biomolecular structure in solution and in the gas phase. We are not aware of any prior comparative HDX/MS experiments on oxy-Hb and aquomet-Hb.


Subject(s)
Deuterium Exchange Measurement/methods , Mass Spectrometry/methods , Methemoglobin/chemistry , Oxyhemoglobins/chemistry , Amino Acid Sequence , Animals , Cattle , Methemoglobin/genetics , Models, Molecular , Molecular Dynamics Simulation , Molecular Sequence Data , Oxyhemoglobins/genetics , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Conformation , Protein Stability , Protein Structure, Quaternary , Protein Subunits
9.
Biochemistry ; 49(50): 10606-15, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21077639

ABSTRACT

On the basis of X-ray crystal structures and electron paramagnetic resonance (EPR) measurements, it has been inferred that the O(2) binding to hemoglobin is stabilized by the hydrogen bonds between the oxygen ligands and the distal histidines. Our previous study by multinuclear nuclear magnetic resonance (NMR) spectroscopy has provided the first direct evidence of such H-bonds in human normal adult oxyhemoglobin (HbO(2) A) in solution. Here, the NMR spectra of uniformly (15)N-labeled recombinant human Hb A (rHb A) and five mutant rHbs in the oxy form have been studied under various experimental conditions of pH and temperature and also in the presence of an organic phosphate, inositol hexaphosphate (IHP). We have found significant effects of pH and temperature on the strength of the H-bond markers, i.e., the cross-peaks for the side chains of the two distal histidyl residues, α58His and ß63His, which form H-bonds with the O(2) ligands. At lower pH and/or higher temperature, the side chains of the distal histidines appear to be more mobile, and the exchange with water molecules in the distal heme pockets is faster. These changes in the stability of the H-bonds with pH and temperature are consistent with the changes in the O(2) affinity of Hb as a function of pH and temperature and are clearly illustrated by our NMR experiments. Our NMR results have also confirmed that this H-bond in the ß-chain is weaker than that in the α-chain and is more sensitive to changes in pH and temperature. IHP has only a minor effect on these H-bond markers compared to the effects of pH and temperature. These H-bonds are sensitive to mutations in the distal heme pockets but not affected directly by the mutations in the quaternary interfaces, i.e., α(1)ß(1) and/or α(1)ß(2) subunit interface. These findings provide new insights regarding the roles of temperature, hydrogen ion, and organic phosphate in modulating the structure and function of hemoglobin in solution.


Subject(s)
Oxyhemoglobins/chemistry , Phytic Acid/chemistry , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Models, Molecular , Mutation , Oxyhemoglobins/genetics , Temperature
10.
Comp Biochem Physiol A Mol Integr Physiol ; 148(3): 498-503, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17689995

ABSTRACT

The hemoglobins contained within the red blood cells of the adult brushtail possum exhibited cooperative (n=2.6) oxygen binding curves with an associated p50 of 38 mm Hg at pH 7.4 and a large Bohr effect (-0.60). Stripped hemolysate showed a Bohr effect of -0.27, and was sensitive to added DPG (K=56 micromol L(-1)), ATP (K=130 micromol L(-1)), and chloride ions. Four isoforms of hemoglobin were identified using isoelectric focusing. Mass spectrometry indicated that all four isoforms most likely represent the same gene products which have differentially undergone post-translational deamidation and glutathionylation. The oxygen binding characteristics of three isolated isohemoglobins have been determined.


Subject(s)
Erythrocytes/metabolism , Hemoglobins/metabolism , Oxygen/blood , Oxyhemoglobins/metabolism , Protein Processing, Post-Translational , Trichosurus/blood , 2,3-Diphosphoglycerate/metabolism , Adenosine Triphosphate/metabolism , Amides/metabolism , Animals , Chlorides/metabolism , Erythrocytes/chemistry , Glutathione/metabolism , Hemoglobins/chemistry , Hemoglobins/genetics , Hemoglobins/isolation & purification , Hydrogen-Ion Concentration , Isoelectric Focusing , Mass Spectrometry , Molecular Weight , Oxyhemoglobins/chemistry , Oxyhemoglobins/genetics , Oxyhemoglobins/isolation & purification , Protein Binding , Protein Isoforms/metabolism , Protein Subunits
11.
Wien Med Wochenschr ; 155(7-8): 171-5, 2005 Apr.
Article in German | MEDLINE | ID: mdl-15966263

ABSTRACT

Many studies have concentrated on investigating the effects of short and medium term hypobaric conditions in people living at low altitudes. In contrast, only little is known about long term or genetic adaptations to chronic hypobaric conditions in people living at high altitudes. A small number of phenotypic characteristics have been defined in these people so far, comprising hormonal functions, oxygen saturation of hemoglobin, skeletal growth, muscular morphology and function, cardiovascular functions, and fluid and electrolyte changes. Several of these characteristics have been attributed to genetic adaptations. However, so far, no specific genes coding for any of the specific phenotypes found in high altitude dwellers have been detected.


Subject(s)
Adaptation, Physiological/physiology , Altitude Sickness/physiopathology , Bone and Bones/physiopathology , Hormones/blood , Muscle, Skeletal/physiopathology , Phenotype , Adaptation, Physiological/genetics , Altitude Sickness/diagnosis , Altitude Sickness/genetics , Animals , Atmospheric Pressure , Blood Pressure/genetics , Blood Pressure/physiology , Body Height , Brain/physiopathology , Cardiovascular System/physiopathology , Female , Genotype , Humans , Infant, Newborn , Male , Oxyhemoglobins/genetics , Pregnancy , Sexual Maturation/genetics , Sexual Maturation/physiology , Water-Electrolyte Balance/physiology
12.
Am J Hematol ; 77(3): 268-76, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15495251

ABSTRACT

Hemoglobin (Hb) Bassett, an abnormal Hb variant with a markedly reduced oxygen affinity, was discovered in a Caucasian (Anglo-Saxon) male child who experienced episodes of cyanosis. Cation-exchange and reversed-phase (RP) high-performance liquid chromatography (HPLC) showed that the patient has an abnormal Hb, with a mutation in the alpha-globin. Tryptic peptide digest of the abnormal alpha-globin with subsequent HPLC analysis revealed abnormal elution of the alpha-T11 peptide. Further studies with Edman sequencing and electrospray mass spectrometry of tryptic peptide alpha-T11, as well as structural analysis by X-ray crystallography revealed an Asp-->Ala substitution at the alpha94 (G1) position, a match for Hb Bassett. Detailed functional studies showed that this Hb variant had a markedly reduced oxygen affinity (P(50) at pH 7.0 = 22 mmHg; Hb A P(50) = 10.5 mmHg), reduced Bohr effect (-0.26 compared to - 0.54 in Hb A), and low subunit cooperativity (n = 1.4, compared to 2.6 in Hb A). X-ray crystallography results explain the probable effects of the structural modification on the oxygen-binding properties of this Hb variant.


Subject(s)
Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Oxygen/blood , Oxyhemoglobins/genetics , Oxyhemoglobins/metabolism , Alanine/genetics , Amino Acid Substitution , Aspartic Acid/genetics , Child , Chromatography, Ion Exchange/methods , Crystallography, X-Ray , Hemoglobin A/metabolism , Hemoglobins, Abnormal/chemistry , Humans , Hydrogen-Ion Concentration , Male , Models, Molecular , Mutation , Oxygen/chemistry , Oxyhemoglobins/chemistry , Partial Pressure , Protein Subunits/chemistry , Protein Subunits/metabolism
14.
Am J Physiol Heart Circ Physiol ; 285(2): H549-61, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12689854

ABSTRACT

With the objective of developing a recombinant oxygen carrier suitable for therapeutic applications, we have employed an Escherichia coli expression system to synthesize in high-yield hemoglobin (Hb) Minotaur, containing alpha-human and beta-bovine chains. Polymerization of Hb Minotaur through S-S intermolecular cross-linking was obtained by introducing a Cys at position beta9 and substituting the naturally occurring Cys. This homogeneous polymer, Hb Polytaur, has a molecular mass of approximately 500 kDa and was resistant toward reducing agents present in blood. In mice, the circulating half-time (3 h) was fivefold greater than adult human Hb (HbA). The half-time of autooxidation measured in blood (46 h) exceeded the circulating retention time. Hypervolemic exchange transfusion resulted in increased arterial blood pressure similar to that with albumin. The increase in pressure was less than that obtained by transfusion of cross-linked tetrameric Hb known to undergo renovascular extravasation. The nitric oxide reactivity of Hb Polytaur was similar to HbA, suggesting that the diminished pressor response to Hb Polytaur was probably related to diminished extravasation. Transfusion of 3% Hb Polytaur during focal cerebral ischemia reduced infarct volume by 22%. Therefore, site-specific Cys insertion on the Hb surface results in uniform size polymers that do not produce the large pressor response seen with tetrameric Hb. Polymerization maintains physiologically relevant oxygen and heme affinity, stability toward denaturation and oxidation, and effective oxygen delivery as indicated by reduced cerebral ischemic damage.


Subject(s)
Hemoglobins/metabolism , Ischemic Attack, Transient/drug therapy , Oxygen/metabolism , Recombinant Proteins/metabolism , Animals , Base Sequence , Blood Substitutes/chemistry , Blood Transfusion , Cattle , Heme/metabolism , Hemoglobins/chemistry , Hemoglobins/genetics , Humans , Ischemic Attack, Transient/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nitric Oxide/metabolism , Oxidation-Reduction , Oxyhemoglobins/chemistry , Oxyhemoglobins/genetics , Oxyhemoglobins/metabolism , Polymers , Protein Conformation , Protein Denaturation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
15.
Acta Crystallogr D Biol Crystallogr ; 57(Pt 12): 1850-6, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11717498

ABSTRACT

The greylag goose (Anser anser), which lives on lowlands and cannot tolerate hypoxic conditions, presents a striking contrast to its close relative the bar-headed goose (A. indicus), which lives at high altitude and possesses high-altitude hypoxia adaptation. There are only four amino-acid residue differences at alpha18, alpha63, alpha119 and beta125 between the haemoglobins of the two species. The crystal structure of greylag goose oxy haemoglobin was determined at 3.09 A resolution. Its quaternary structure is slightly different from that of the bar-headed goose oxy haemoglobin, with a rotation of 2.8 degrees in relative orientation of the two dimers. Of the four mutations, those at alpha119 and beta125 produce contact changes in the alpha(1)beta(1) interface and may be responsible for the differences in intrinsic oxygen affinity between the two species; those at alpha18 and alpha63 may be responsible for the differences in quaternary structure between the two species.


Subject(s)
Oxyhemoglobins/chemistry , Animals , Crystallization , Crystallography, X-Ray , Geese , Heme/metabolism , Inositol Phosphates/metabolism , Models, Molecular , Mutation , Oxyhemoglobins/genetics , Protein Conformation , Quality Control
16.
Proteins ; 42(3): 355-77, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11151007

ABSTRACT

A practical computational method for the molecular modeling of free-energy changes associated with protein mutations is reported. The de novo generation, optimization, and thermodynamic analysis of a wide variety of deoxy and oxy hemoglobin mutants are described in detail. Hemoglobin is shown to be an ideal candidate protein for study because both the native deoxy and oxy states have been crystallographically determined, and a large and diverse population of its mutants has been thermodynamically characterized. Noncovalent interactions for all computationally generated hemoglobin mutants are quantitatively examined with the molecular modeling program HINT (Hydropathic INTeractions). HINT scores all biomolecular noncovalent interactions, including hydrogen bonding, acid-base, hydrophobic-hydrophobic, acid-acid, base-base, and hydrophobic-polar, to generate dimer-dimer interface "scores" that are translated into free-energy estimates. Analysis of 23 hemoglobin mutants, in both deoxy and oxy states, indicates that the effects of mutant residues on structurally bound waters (and visa versa) are important for generating accurate free-energy estimates. For several mutants, the addition/elimination of structural waters is key to understanding the thermodynamic consequences of residue mutation. Good agreement is found between calculated and experimental data for deoxy hemoglobin mutants (r = 0.79, slope = 0.78, standard error = 1.4 kcal mol(-1), n = 23). Less accurate estimates were initially obtained for oxy hemoglobin mutants (r = 0.48, slope = 0.47, standard error = 1.4 kcal mol(-1), n = 23). However, the elimination of three outliers from this data set results in a better correlation of r = 0.87 (slope = 0.72, standard error = 0.75, n = 20). These three mutations may significantly perturb the hemoglobin quaternary structure beyond the scope of our structural optimization procedure. The method described is also useful in the examination of residue ionization states in protein structures. Specifically, we find an acidic residue within the native deoxy hemoglobin dimer-dimer interface that may be protonated at physiological pH. The final analysis is a model design of novel hemoglobin mutants that modify cooperative free energy (deltaGc)--the energy barrier between the allosteric transition from deoxy to oxy hemoglobin.


Subject(s)
Computational Biology , Hemoglobins/chemistry , Oxyhemoglobins/chemistry , Dimerization , Energy Metabolism , Hemoglobins/genetics , Models, Molecular , Mutagenesis, Site-Directed , Oxyhemoglobins/genetics , Protein Conformation , Software , Thermodynamics , Water/chemistry
17.
Protein Eng ; 13(2): 113-20, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10708650

ABSTRACT

The cysteine residue at F9(93) of the human hemoglobin (Hb A) beta chain, conserved in mammalian and avian hemoglobins, is located near the functionally important alpha1-beta2 interface and C-terminal region of the beta chain and is reactive to sulfhydryl reagents. The functional roles of this residue are still unclear, although regulation of local blood flow through allosteric S-nitrosylation of this residue is proposed. To clarify the role of this residue and its functional homology to F9(88) of the alpha chain, we measured oxygen equilibrium curves, UV-region derivative spectra, Soret-band absorption spectra, the number of titratable -SH groups with p-mercuribenzoate and the rate of reaction of these groups with 4, 4'-dipyridine disulfide for three recombinant mutant Hbs with single amino acid substitutions: Ala-->Cys at 88alpha (rHb A88alphaC), Cys-->Ala at 93beta (rHb C93betaA) and Cys-->Thr at 93beta (rHb C93betaT). These Hbs showed increased oxygen affinities and impaired allosteric effects. The spectral data indicated that the R to T transition upon deoxygenation was partially restricted in these Hbs. The number of titratable -SH groups of liganded form was 3.2-3.5 for rHb A88alphaC compared with 2.2 for Hb A, whereas those for rHb C93betaA and rHb C93betaT were negligibly small. The reduction of rate of reaction with 4,4'-dipyridine disulfide upon deoxygenation in rHb A88alphaC was smaller than that in Hb A. Our experimental data have shown that the residues at 88alpha and 93beta have definite roles but they have no functional homology. Structure-function relationships in our mutant Hbs are discussed.


Subject(s)
Hemoglobin A/chemistry , Hemoglobin A/genetics , Mutagenesis, Site-Directed , Allosteric Regulation , Amino Acid Substitution , Amino Acids/chemistry , Carboxyhemoglobin/chemistry , Cysteine/chemistry , Humans , Hydrogen-Ion Concentration , Kinetics , Mass Spectrometry , Mercuribenzoates/chemistry , Mercuribenzoates/metabolism , Oxygen/chemistry , Oxygen/metabolism , Oxyhemoglobins/chemistry , Oxyhemoglobins/genetics , Recombinant Proteins , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Sulfhydryl Reagents/chemistry , Sulfhydryl Reagents/metabolism , Titrimetry
18.
Biochemistry ; 38(27): 8751-61, 1999 Jul 06.
Article in English | MEDLINE | ID: mdl-10393550

ABSTRACT

Using our Escherichia coli expression system, we have produced five mutant recombinant (r) hemoglobins (Hbs): r Hb (alpha V96 W), r Hb Presbyterian (beta N108K), r Hb Yoshizuka (beta N108D), r Hb (alpha V96W, beta N108K), and r Hb (alpha V96W, beta N108D). These r Hbs allow us to investigate the effect on the structure-function relationship of Hb of replacing beta 108Asn by either a positively charged Lys or a negatively charged Asp as well as the effect of replacing alpha 96Val by a bulky, nonpolar Trp. We have conducted oxygen-binding studies to investigate the effect of several allosteric effectors on the oxygenation properties and the Bohr effects of these r Hbs. The oxygen affinity of these mutants is lower than that of human normal adult hemoglobin (Hb A) under various experimental conditions. The oxygen affinity of r Hb Yoshizuka is insensitive to changes in chloride concentration, whereas the oxygen affinity of r Hb Presbyterian exhibits a pronounced chloride effect. r Hb Presbyterian has the largest Bohr effect, followed by Hb A, r Hb (alpha V96W), and r Hb Yoshizuka. Thus, the amino acid substitution in the central cavity that increases the net positive charge enhances the Bohr effect. Proton nuclear magnetic resonance studies demonstrate that these r Hbs can switch from the R quaternary structure to the T quaternary structure without changing their ligation states upon the addition of an allosteric effector, inositol hexaphosphate, and/or by reducing the temperature. r Hb (alpha V96W, beta N108K), which has the lowest oxygen affinity among the hemoglobins studied, has the greatest tendency to switch to the T quaternary structure. The following conclusions can be derived from our results: First, if we can stabilize the deoxy (T) quaternary structure of a hemoglobin molecule without perturbing its oxy (R) quaternary structure, we will have a hemoglobin with low oxygen affinity and high cooperativity. Second, an alteration of the charge distribution by amino acid substitutions in the alpha 1 beta 1 subunit interface and in the central cavity of the hemoglobin molecule can influence the Bohr effect. Third, an amino acid substitution in the alpha 1 beta 1 subunit interface can affect both the oxygen affinity and cooperativity of the oxygenation process. There is communication between the alpha 1 beta 1 and alpha 1 beta 2 subunit interfaces during the oxygenation process. Fourth, there is considerable cooperativity in the oxygenation process in the T-state of the hemoglobin molecule.


Subject(s)
Amino Acid Substitution/genetics , Hemoglobin A/chemistry , Oxygen/blood , 2,3-Diphosphoglycerate , Adult , Asparagine/genetics , Aspartic Acid/genetics , Buffers , Chlorides , HEPES , Hemoglobin A/genetics , Hemoglobins/chemistry , Hemoglobins/genetics , Hemoglobins, Abnormal/chemistry , Humans , Lysine/genetics , Nuclear Magnetic Resonance, Biomolecular , Oxyhemoglobins/chemistry , Oxyhemoglobins/genetics , Phosphates , Protein Binding/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Structure-Activity Relationship , Tryptophan/genetics , Valine/genetics
19.
Clin Lab Haematol ; 18(4): 237-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9054694

ABSTRACT

A new high oxygen affinity haemoglobin with the beta chain mutation beta146 HIS --> TYR is described. This variant was detected in a fit 34-year-old man with true erythrocytosis. The abnormal haemoglobin was identified as an extra band on cellulose acetate electrophoresis at pH 6.3 and was later confirmed by beta globin gene sequencing and oxygen dissociation studies. Whole blood containing Haemoglobin Hallamshire has a P50 of 18 mmHg. This newly described haemoglobin variant was also responsible for erythrocytosis in the mother and maternal half cousin of the index case. The identification of Haemoglobin Hallamshire provides confirmatory evidence of the important role of the C-terminal end of the chain in haemoglobin function.


Subject(s)
Hemoglobins, Abnormal/analysis , Polycythemia/blood , Adult , Blood Protein Electrophoresis , Family Health , Female , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Heterozygote , Histidine/genetics , Histidine/physiology , Humans , Hydrogen-Ion Concentration , Male , Oxyhemoglobins/genetics , Oxyhemoglobins/metabolism , Point Mutation/genetics , Polycythemia/genetics , Polycythemia/physiopathology , Tyrosine/genetics , Tyrosine/physiology
20.
J Biol Chem ; 270(45): 26857-62, 1995 Nov 10.
Article in English | MEDLINE | ID: mdl-7592928

ABSTRACT

In order to investigate the role of the R (relaxed) to T (tense) structural transition in facilitating polymerization of deoxy-Hb S, we have engineered and expressed two Hb S variants which destabilize either T state (Hb S-Kempsey, alpha 2 beta 2 Val-6,Asn-99) or R state structures (Hb S-Kansas, alpha 2 beta 2 Val-6, Thr-102). Polymerization of deoxy-Hb S-Kempsey, which shows high oxygen affinity and increased dimer dissociation, required about 2- and 6-fold higher hemoglobin concentrations than deoxy-Hb S for polymerization in low and high phosphate concentrations, and its kinetic pattern of polymerization was biphasic. In contrast, oxy- or CO Hb S-Kansas, which shows low oxygen affinity and increased dimer dissociation, polymerized at a slightly higher critical concentration than that required for polymerization of deoxy-Hb S in both low and high phosphate buffers. Polymerization of oxy- and CO Hb S-Kansas was linear and showed no delay time, which is similar to oversaturated oxy- or CO Hb S. These results suggest that nuclei formation, which occurs during the delay time prior to deoxy-Hb S polymerization, does not occur in T state oxy-Hb S-Kansas, even though the critical concentration for polymerization of T state oxy-Hb S-Kansas is similar to that of T state deoxy-Hb S.


Subject(s)
Hemoglobins, Abnormal/chemistry , Carboxyhemoglobin/chemistry , Carboxyhemoglobin/genetics , Genetic Variation , Hemoglobins/chemistry , Hemoglobins/genetics , Hemoglobins, Abnormal/genetics , Humans , Kinetics , Molecular Structure , Oxidation-Reduction , Oxyhemoglobins/chemistry , Oxyhemoglobins/genetics , Polymers/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Saccharomyces cerevisiae/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...