Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Phys Chem B ; 122(49): 11591-11597, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30222355

ABSTRACT

Proteins that function in aqueous solution can be perturbed by the solvent. Here we present experimental studies on two such interactions in the hemoglobin molecule. (1) Hemoglobin's oxygen binding is altered by introduction of crowding species or osmoticants, such as sucrose, through the linked binding of ions such as Cl or CO2, but not otherwise. This rules out a significant role of buried surface in the allosteric energetics. (2) Sickle hemoglobin (HbS) polymerizes more readily in high concentrations of phosphate buffer. Such polymerization is analyzed quantitatively here for the first time in terms of the double nucleation mechanism. The changes in solubility are found to account for the increase in monomer addition rates and nucleation rates without requiring additional parameter adjustments. In the analysis, we also show how the analytical formulation of HbS nucleation may be adapted to include water that occupies the interstices between the assembled molecules. While such a "correction" has been applied to the equilibrium process, it has not previously been applied to the nucleation process.


Subject(s)
Carbon Dioxide/chemistry , Chlorides/chemistry , Hemoglobin, Sickle/chemistry , Polymerization , Water/chemistry , Carbon Dioxide/chemical synthesis , Chlorides/chemical synthesis , Hemoglobin, Sickle/chemical synthesis , Ions/chemical synthesis , Ions/chemistry , Solubility
2.
J Mol Biol ; 428(8): 1506-14, 2016 Apr 24.
Article in English | MEDLINE | ID: mdl-26975885

ABSTRACT

Sickle cell disease is fundamentally a kinetic disorder, in which cells containing the mutated hemoglobin (hemoglobin S; HbS) will cause occlusion if they sickle in the microvasculature, but have minimal (or no) consequences if they sickle in the venous return. Physiologically, sickling always occurs when some ligands are present; nonetheless, the kinetics in the presence of ligands are virtually unstudied. Sickling arises from nucleation-controlled polymer formation, triggered when the HbS loses ligands (e.g., oxygen). Thus, understanding how nucleation responds to the presence of oxygen is the key to understanding how sickling proceeds in a physiological context. We have measured the rate of nucleus formation in HbS partially liganded with NO or CO, which we find have equivalent effects in reducing the nucleation rates. We find that hemoglobin must be in the T (tense) quaternary structure for nucleation, but the presence of ligands inhibits nucleus formation even when the correct quaternary structure is present. From these results, we can predict the fraction of cells that will sickle at any given partial ligand saturations. The ability to make such predictions may prove especially useful in designing future therapies, particularly those where the oxygen affinity is perturbed.


Subject(s)
Anemia, Sickle Cell/diagnosis , Anemia, Sickle Cell/physiopathology , Hemoglobin, Sickle/chemistry , Hemoglobins/chemistry , Humans , Ligands , Microcirculation , Oxygen/chemistry , Oxygen Consumption , Polymers/chemistry , Probability , Protein Structure, Quaternary , Protein Structure, Tertiary
3.
Biophys J ; 101(11): 2790-7, 2011 Dec 07.
Article in English | MEDLINE | ID: mdl-22261068

ABSTRACT

Sickle hemoglobin (HbS) is a point mutation of the two ß subunits in normal Hb (HbA) that leads to nucleated polymerization and accompanying pathology. We measured the rates of homogeneous and heterogeneous nucleation of HbS in the presence of up to 50% HbA under conditions in which hybrid HbAS molecules will also form. The replacement of 50% of HbS by HbA slows polymerization by factors of ∼100 in the physiological range, which is substantially less than previously thought. To provide a theoretical description of these data, we extended the double nucleation model for HbS polymerization to conditions in which hybridized mixtures are present. Measurements of homogeneous nucleation and the theory agree only when at least one of the molecules in the nucleus is not a hybrid. We attribute this to the necessary presence in the nucleus of a molecule that utilizes both ß-subunit mutation sites in intermolecular contacts, whereas the remaining molecules engage only one of the mutation sites. Heterogeneous nucleation appears to require an even greater number of nonhybrid molecules, presumably because of the need for the nucleus to attach to the polymer as well as to form internal bonds. These results also provide insights into the pathophysiology of sickle cell disease, including the occasional severe events that strike persons in whom both HbS and HbA are expressed, a condition known as sickle trait. The studies reported here are necessary for understanding physiologically relevant polymerization in the presence of ligands as well as therapeutically relevant copolymerizing inhibitors.


Subject(s)
Hemoglobin A/metabolism , Hemoglobin, Sickle/metabolism , Models, Molecular , Humans , Temperature , Time Factors
4.
Biophys Chem ; 146(2-3): 55-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19880238

ABSTRACT

We have measured homogeneous and heterogeneous nucleation rates of sickle hemoglobin (HbS) in the presence of a strongly binding deletion mutant of the cytoplasmic domain of band 3 (cdb3), a membrane protein known to form dimers and to bind 2 HbS molecules to such a dimer, and we find that it accelerated both rates by a factor of 2. A weakly binding mutant, in contrast showed no impact on nucleation rates, contrary to naïve expectations of a slight enhancement based on the molecular crowding of the solution by the mutant. We find we can explain these phenomena by a model of HbS-cdb3 interaction in which the strong binding mutant, by stabilizing an HbS dimer, catalyzes the nucleation process, while the weak mutant binds only 1 HbS molecule, effectively inactivating it and thereby compensating for the crowding of the solution by the cdb3. The catalytic behavior we observe could play a role in intracellular processes.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Biocatalysis , Hemoglobin, Sickle/chemistry , Hemoglobin, Sickle/metabolism , Protein Multimerization , Anion Exchange Protein 1, Erythrocyte/chemistry , Anion Exchange Protein 1, Erythrocyte/genetics , Mutation , Protein Structure, Quaternary , Protein Structure, Tertiary
5.
J Mol Biol ; 369(5): 1170-4, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17493634

ABSTRACT

Sickle cell disease arises from a genetic mutation of one amino acid in each of the two hemoglobin beta chains, leading to the polymerization of hemoglobin in the red cell upon deoxygenation, and is characterized by vascular crises and tissue damage due to the obstruction of small vessels by sickled cells. It has been an untested assumption that, in red cells that sickle, the growing polymer mass would consume monomers until the thermodynamically well-described monomer solubility was reached. By photolysing droplets of sickle hemoglobin suspended in oil we find that polymerization does not exhaust the available store of monomers, but stops prematurely, leaving the solutions in a supersaturated, metastable state typically 20% above solubility at 37 degrees C, though the particular values depend on the details of the experiment. We propose that polymer growth stops because the growing ends reach the droplet edge, whereas new polymer formation is thwarted by long nucleation times, since the concentration of hemoglobin is lowered by depletion of monomers into the polymers that have formed. This finding suggests a new aspect to the pathophysiology of sickle cell disease; namely, that cells deoxygenated in the microcirculation are not merely undeformable, but will actively wedge themselves tightly against the walls of the microvasculature by a ratchet-like mechanism driven by the supersaturated solution.


Subject(s)
Hemoglobin, Sickle/chemistry , Hemoglobin, Sickle/metabolism , Molecular Biology/methods , Anemia, Sickle Cell/pathology , Humans , Lasers , Photolysis , Temperature
6.
J Mol Biol ; 362(3): 528-38, 2006 Sep 22.
Article in English | MEDLINE | ID: mdl-16926024

ABSTRACT

Polymerization of a 1:1 mixture of hemoglobin S (Hb S) and the artificial mutant HbAbeta73Leu produces a dramatic morphological change in the polymer domains in 1.0 M phosphate buffer that are a characteristic feature of polymer formation. Instead of feathery domains with quasi 2-fold symmetry that characterize polymerization of Hb S and all previously known mixtures such as Hb A/S and Hb F/S mixtures, these domains are compact structures of quasi-spherical symmetry. Solubility of Hb S/Abeta73Leu mixtures was similar to that of Hb S/F mixtures. Kinetics of polymerization indicated that homogeneous nucleation rates of Hb S/Abeta73Leu mixtures were the same as those of Hb S/F mixtures, while exponential polymer growth (B) of Hb S/Abeta73Leu mixtures were about three times slower than those of Hb S/F mixtures. Differential interference contrast (DIC) image analysis also showed that fibers in the mixture appear to elongate between three and five times more slowly than in equivalent Hb S/F mixtures by direct measurements of exponential growth of mass of polymer in a domain. We propose that these results of Hb S/Abeta73Leu mixtures arise from a non-productive binding of the hybrid species of this mixture to the end of the growing polymer. This "cap" prohibits growth of polymers, but by nature is temporary, so that the net effect is a lowered growth rate of polymers. Such a cap is consistent with known features of the structure of the Hb S polymer. Domains would be more spherulitic because slower growth provides more opportunity for fiber bending to spread domains from their initial 2-fold symmetry. Moreover, since monomer depletion proceeds more slowly in this mixture, more homogeneous nucleation events occur, and the resulting gel has a far more granular character than normally seen in mixtures of non-polymerizing hemoglobins with Hb S. This mixture is likely to be less stiff than polymerized mixtures of other hybrids such as Hb S with HbF, potentially providing a novel approach to therapy.


Subject(s)
Hemoglobin A/chemistry , Hemoglobin A/genetics , Hemoglobin, Sickle/chemistry , Amino Acid Substitution , Biopolymers/chemistry , Biopolymers/genetics , Biopolymers/metabolism , Hemoglobin A/metabolism , Hemoglobin, Sickle/genetics , Hemoglobin, Sickle/metabolism , Humans , In Vitro Techniques , Kinetics , Microscopy, Interference , Multiprotein Complexes , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Solubility
7.
Biophys J ; 89(4): 2677-84, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16055526

ABSTRACT

Sickle hemoglobin polymerizes by two types of nucleation: homogeneous nucleation of aggregates in solution, and heterogeneous nucleation on preexisting polymers. It has been proposed that the same contact that is made in the interior of the polymer between the mutant site beta6 and its receptor pocket on an adjacent molecule is the primary contact site for the heterogeneous nucleus. We have constructed cross-linked hybrid molecules in which one beta-subunit is from HbA with Glu at beta6, and the other is from HbS with a Val at beta6. We measured solubility (using sedimentation) and polymerization kinetics (using laser photolysis) on cross-linked hybrids, and cross-linked HbS as controls. We find approximately 4000 times less heterogeneous nucleation in the cross-linked AS molecules than in cross-linked HbS, in strong confirmation of the proposal. In addition, changes in stability of the nucleus support a further proposal that more than one beta6 contact is involved in the homogeneous nucleus.


Subject(s)
Crystallization/methods , Hemoglobin A/analysis , Hemoglobin A/chemistry , Hemoglobin, Sickle/analysis , Hemoglobin, Sickle/chemistry , Models, Chemical , Binding Sites , Computer Simulation , Dimerization , Multiprotein Complexes/analysis , Multiprotein Complexes/chemistry , Protein Binding , Protein Conformation , Solubility , Solutions , Viscosity
8.
J Mol Biol ; 347(5): 1015-23, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15784260

ABSTRACT

The dominant assumption central to most treatments for sickle cell anemia has been that replacement of sickle hemoglobin (HbS) by fetal hemoglobin (HbF) would have major clinical benefit. Using laser photolysis, we have measured polymerization kinetics including rates of homogeneous and heterogeneous nucleation on mixtures of 20% and 30% HbF with HbS. We find that the present model for polymerization, including molecular crowding, can accurately predict the rates of such mixtures, by using the single assumption that no significant amount of HbF enters the polymer. The effects of replacing HbS by HbF on the rates of polymer formation are found to be significantly lower than previous measurements appeared to indicate because the impact of the replacement is also highly dependent on the total hemoglobin concentration. This is because the molecular crowding of non-polymerizing HbF offsets substantially the effects of decreasing the concentration of HbS concentration, an effect that increases with concentration. Most strikingly, the demonstrated benefit of hydroxyurea therapy in slowing the kinetics of intracellular polymerization cannot be primarily due to enhanced HbF, but must have some other origin, which could itself represent a promising therapeutic approach.


Subject(s)
Anemia, Sickle Cell/metabolism , Anemia, Sickle Cell/therapy , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/therapeutic use , Anemia, Sickle Cell/pathology , Humans , Kinetics
9.
Biophys J ; 88(4): 2815-22, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15653736

ABSTRACT

Pathology in sickle cell disease begins with nucleation-dependent polymerization of deoxyhemoglobin S into stiff, rodlike fibers that deform and rigidify red cells. We have measured the effect of erythrocyte membranes on the rate of homogeneous nucleation in sickle hemoglobin, using preparations of open ghosts (OGs) with intact cytoskeletons from sickle (SS) and normal adult (AA) red cells. Nucleation rates were measured by inducing polymerization by laser photolysis of carboxy sickle hemoglobin and observing stochastic variation of replicate experiments of the time for the scattering signals to reach 10% of their respective maxima. By optical imaging of membrane fragments added to a hemoglobin solution we contrast the rate of nucleation immediately adjacent to membrane fragments with nucleation in a region of the same solution but devoid of membranes. From analysis of 29,272 kinetic curves obtained, we conclude that the effect of AA OGs is negligible (10% enhancement of nucleation rates +/-20%), whereas SS OGs caused 80% enhancement (+/-20%). In red cells, where more membrane surface is available to Hb, this implies enhancement of nucleation by a factor of 6. These experiments represent a 10-fold improvement in precision over previous approaches and are the first direct, quantitative measure of the impact of erythrocyte membranes on the homogeneous nucleation process that is responsible for polymer initiation in sickle cell disease.


Subject(s)
Anemia, Sickle Cell/blood , Erythrocyte Membrane/metabolism , Hemoglobin, Sickle/chemistry , Biophysics/methods , Erythrocytes, Abnormal/metabolism , Hemoglobins/chemistry , Humans , Kinetics , Lasers , Light , Microscopy, Confocal , Photolysis , Polymers/chemistry , Scattering, Radiation , Temperature , Time Factors
10.
J Mol Recognit ; 17(5): 497-504, 2004.
Article in English | MEDLINE | ID: mdl-15362110

ABSTRACT

Under physiological conditions, sickle hemoglobin, a natural mutant of human hemoglobin A with a surface hydrophobic valine in place of a negatively charged glutamic acid, polymerizes at high volume occupancy. Equilibrium solubility of sickle hemoglobin entails activity coefficients that can approach 10(3) at high concentrations. Polymerization occurs by homogeneous and heterogeneous nucleation mechanisms, which are both profoundly sensitive to crowding; homogeneous nucleation rates for example are enhanced by 10(10) when the initial concentration is augmented by 50% non-polymerizing hemoglobin. A molecular description of the reaction therefore entails substantial corrections for molecular crowding which are all very accurately described by excluded volume corrections, treating hemoglobin as a hard sphere with volume consistent with the molecular structure of the molecule, and involving no further adjustable parameters. These effects and the descriptions that rationalize this behavior are described.


Subject(s)
Biopolymers/chemistry , Hemoglobin, Sickle/chemistry , Anemia, Sickle Cell/metabolism , Biopolymers/metabolism , Hemoglobin, Sickle/metabolism , Humans , Kinetics , Solutions/chemistry , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL
...