Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
PLoS One ; 13(9): e0203955, 2018.
Article in English | MEDLINE | ID: mdl-30235277

ABSTRACT

Nitric oxide (NO) can be generated from nitrite by reductase activity of deoxygenated hemoglobin (deoxyHb) apparently to facilitate tissue perfusion under hypoxic condition. Although hemoglobin E (HbE) solutions have been shown to exhibit decreased rate of nitrite reduction to NO, this observation has never been reported in erythrocytes from subjects with hemoglobin E/ß-thalassemia (HbE/ß-thal). In this study, we investigated the nitrite reductase activity of deoxyHb dialysates from 58 non-splenectomized and 23 splenectomized HbE/ß-thal subjects compared to 47 age- and sex-matched normal subjects, and examined its correlation with platelet activity. Iron-nitrosyl-hemoglobin (HbNO) was measured by tri-iodide reductive chemiluminescence as a marker of NO generation. HbNO produced from the reaction of nitrite with deoxyHb dialysate from both non-splenectomized and splenectomized HbE/ß-thal subjects was lower than that of normal (AA) hemoglobin subjects. P-selectin expression, a marker of platelet activation, at baseline and in reactivity to stimulation by adenosine diphosphate (ADP), were higher in HbE/ß-thal subjects than normal subjects. HbNO formation from the reactions of nitrite and deoxyHb inversely correlated with baseline platelet P-selectin expression, HbE levels, and tricuspid regurgitant velocity (TRV). Nitrite plus deoxygenated erythrocytes from HbE/ß-thal subjects had a lower ability to inhibit ADP-induced P-selectin expression on platelets than erythrocytes from normal subjects. We conclude that deoxyHb in erythrocytes from HbE/ß-thal subjects has a decreased ability to reduce nitrite to NO, which is correlated with increased platelet activity in these individuals.


Subject(s)
Hemoglobin E/metabolism , Hemoglobins/metabolism , Nitrite Reductases/metabolism , Platelet Activation/physiology , beta-Thalassemia/metabolism , Adult , Blood Platelets/metabolism , Female , Humans , Male , P-Selectin/metabolism
2.
Antioxid Redox Signal ; 26(14): 794-813, 2017 05 10.
Article in English | MEDLINE | ID: mdl-27650096

ABSTRACT

SIGNIFICANCE: Oxidative stress and generation of free radicals are fundamental in initiating pathophysiological mechanisms leading to an inflammatory cascade resulting in high rates of morbidity and death from many inherited point mutation-derived hemoglobinopathies. Hemoglobin (Hb)E is the most common point mutation worldwide. The ßE-globin gene is found in greatest frequency in Southeast Asia, including Thailand, Malaysia, Indonesia, Vietnam, Cambodia, and Laos. With the wave of worldwide migration, it is entering the gene pool of diverse populations with greater consequences than expected. CRITICAL ISSUES: While HbE by itself presents as a mild anemia and a single gene for ß-thalassemia is not serious, it remains unexplained why HbE/ß-thalassemia (HbE/ß-thal) is a grave disease with high morbidity and mortality. Patients often exhibit defective physical development, severe chronic anemia, and often die of cardiovascular disease and severe infections. Recent Advances: This article presents an overview of HbE/ß-thal disease with an emphasis on new findings pointing to pathophysiological mechanisms derived from and initiated by the dysfunctional property of HbE as a reduced nitrite reductase concomitant with excess α-chains exacerbating unstable HbE, leading to a combination of nitric oxide imbalance, oxidative stress, and proinflammatory events. FUTURE DIRECTIONS: Additionally, we present new therapeutic strategies that are based on the emerging molecular-level understanding of the pathophysiology of this and other hemoglobinopathies. These strategies are designed to short-circuit the inflammatory cascade leading to devastating chronic morbidity and fatal consequences. Antioxid. Redox Signal. 26, 794-813.


Subject(s)
Hemoglobin E/metabolism , Hemoglobinopathies/drug therapy , Hemoglobinopathies/physiopathology , Oxidative Stress , beta-Thalassemia/metabolism , Animals , Hemoglobin E/genetics , Hemoglobinopathies/metabolism , Humans , Point Mutation , beta-Thalassemia/genetics
3.
Redox Biol ; 8: 363-74, 2016 08.
Article in English | MEDLINE | ID: mdl-26995402

ABSTRACT

When adding peroxide (H2O2), ß subunits of hemoglobin (Hb) bear the burden of oxidative changes due in part to the direct oxidation of its Cys93. The presence of unpaired α subunits within red cells and/or co-inheritance of another ß subunit mutant, HbE (ß26 Glu→Lys) have been implicated in the pathogenesis and severity of ß thalassemia. We have found that although both HbA and HbE autoxidize at initially comparable rates, HbE loses heme at a rate almost 2 fold higher than HbA due to unfolding of the protein. Using mass spectrometry and the spin trap, DMPO, we were able to quantify irreversible oxidization of ßCys93 to reflect oxidative instability of ß subunits. In the presence of free α subunits and H2O2, both HbA and HbE showed ßCys93 oxidation which increased with higher H2O2 concentrations. In the presence of Alpha-hemoglobin stabilizing protein (AHSP), which stabilizes the α-subunit in a redox inactive hexacoordinate conformation (thus unable to undergo the redox ferric/ferryl transition), Cys93 oxidation was substantially reduced in both proteins. These experiments establish two important features that may have relevance to the mechanistic understanding of these two inherited hemoglobinopathies, i.e. HbE/ß thalassemia: First, a persistent ferryl/ferryl radical in HbE is more damaging to its own ß subunit (i.e., ßCys93) than HbA. Secondly, in the presence of excess free α-subunit and under the same oxidative conditions, these events are substantially increased for HbE compared to HbA, and may therefore create an oxidative milieu affecting the already unstable HbE.


Subject(s)
Blood Proteins/metabolism , Hemoglobin E/metabolism , Molecular Chaperones/metabolism , Oxidative Stress/genetics , beta-Thalassemia/metabolism , Blood Proteins/chemistry , Erythrocytes/metabolism , Erythrocytes/pathology , Heme/chemistry , Heme/metabolism , Hemoglobin E/chemistry , Humans , Hydrogen Peroxide/toxicity , Molecular Chaperones/chemistry , Oxidative Stress/drug effects , beta-Thalassemia/pathology
4.
J Biol Chem ; 288(31): 22408-25, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23775069

ABSTRACT

In vitro, ferrous deoxy-hemes in hemoglobin (Hb) react with nitrite to generate nitric oxide (NO) through a nitrite reductase reaction. In vivo studies indicate Hb with nitrite can be a source of NO bioactivity. The nitrite reductase reaction does not appear to account fully for this activity because free NO is short lived especially within the red blood cell. Thus, the exporting of NO bioactivity both out of the RBC and over a large distance requires an additional mechanism. A nitrite anhydrase (NA) reaction in which N2O3, a potent S-nitrosating agent, is produced through the reaction of NO with ferric heme-bound nitrite has been proposed (Basu, S., Grubina, R., Huang, J., Conradie, J., Huang, Z., Jeffers, A., Jiang, A., He, X., Azarov, I., Seibert, R., Mehta, A., Patel, R., King, S. B., Hogg, N., Ghosh, A., Gladwin, M. T., and Kim-Shapiro, D. B. (2007) Nat. Chem. Biol. 3, 785-794) as a possible mechanism. Legitimate concerns, including physiological relevance and the nature of the mechanism, have been raised concerning the NA reaction. This study addresses these concerns demonstrating NO and nitrite with ferric hemes under near physiological conditions yield an intermediate having the properties of the purported NA heme-bound N2O3 intermediate. The results indicate that ferric heme sites, traditionally viewed as a source of potential toxicity, can be functionally significant, especially for partially oxygenated/partially met-R state Hb that arises from the NO dioxygenation reaction. In the presence of low levels of nitrite and either NO or a suitable reductant such as L-cysteine, these ferric heme sites can function as a generator for the formation of S-nitrosothiols such as S-nitrosoglutathione and, as such, should be considered as a source of RBC-derived and exportable bioactive NO.


Subject(s)
Hemoglobins/metabolism , S-Nitrosothiols/metabolism , Chromatography, High Pressure Liquid , Fluorescence , Hemoglobins/chemistry , Humans , Mass Spectrometry , Molecular Conformation , S-Nitrosothiols/chemistry
5.
Blood Cells Mol Dis ; 48(2): 91-101, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22260787

ABSTRACT

Hemoglobin (Hb) E (ß26 Glu→Lys) is the most common abnormal hemoglobin (Hb) variant in the world. Homozygotes for HbE are mildly thalassemic as a result of the alternate splice mutation and present with a benign clinical picture (microcytic and mildly anemic) with rare clinical symptoms. Given that the human red blood cell (RBC) contains both HbE and excess α-chains along with minor hemoglobins, the consequence of HbE alone on RBC pathophysiology has not been elucidated. This becomes critical for the highly morbid ß(E)-thalassemia disease. We have generated transgenic mice exclusively expressing human HbE (HbEKO) that exhibit the known aberrant splicing of ß(E) globin mRNA, but are essentially non-thalassemic as demonstrated by RBC α/ß (human) globin chain synthesis. These mice exhibit hematological characteristics similar to presentations in human EE individuals: microcytic RBC with low MCV and MCH but normal MCHC; target RBC; mild anemia with low Hb, HCT and mildly elevated reticulocyte levels and decreased osmotic fragility, indicating altered RBC surface area to volume ratio. These alterations are correlated with a mild RBC oxidative stress indicated by enhanced membrane lipid peroxidation, elevated zinc protoporphyrin levels, and by small but significant changes in cardiac function. The C57 (background) mouse and full KO mouse models expressing HbE with the presence of HbS or HbA are used as controls. In select cases, the HbA full KO mouse model is compared but found to be limited due to its RBC thalassemic characteristics. Since the HbEKO mouse RBC lacks an abundance of excess α-chains that would approximate a mouse thalassemia (or a human thalassemia), the results indicate that the observed in vivo RBC mild oxidative stress arises, at least in part, from the molecular consequences of the HbE mutation.


Subject(s)
Hemoglobin E/genetics , Hemoglobin E/metabolism , Mice, Transgenic , Oxidative Stress , Animals , Breeding , Erythrocyte Indices , Erythrocytes/metabolism , Female , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osmotic Fragility , alpha-Globins/biosynthesis , beta-Globins/biosynthesis
6.
J Biol Chem ; 286(26): 23452-66, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21531715

ABSTRACT

Hemoglobin (Hb) E (ß-Glu26Lys) remains an enigma in terms of its contributions to red blood cell (RBC) pathophysiological mechanisms; for example, EE individuals exhibit a mild chronic anemia, and HbE/ß-thalassemia individuals show a range of clinical manifestations, including high morbidity and death, often resulting from cardiac dysfunction. The purpose of this study was to determine and evaluate structural and functional consequences of the HbE mutation that might account for the pathophysiology. Functional studies indicate minimal allosteric consequence to both oxygen and carbon monoxide binding properties of the ferrous derivatives of HbE. In contrast, redox-sensitive reactions are clearly impacted as seen in the following: 1) the ∼2.5 times decrease in the rate at which HbE catalyzes nitrite reduction to nitric oxide (NO) relative to HbA, and 2) the accelerated rate of reduction of aquometHbE by L-cysteine (L-Cys). Sol-gel encapsulation studies imply a shift toward a higher redox potential for both the T and R HbE structures that can explain the origin of the reduced nitrite reductase activity of deoxyHbE and the accelerated rate of reduction of aquometHbE by cysteine. Deoxy- and CO HbE crystal structures (derived from crystals grown at or near physiological pH) show loss of hydrogen bonds in the microenvironment of ßLys-26 and no significant tertiary conformational perturbations at the allosteric transition sites in the R and T states. Together, these data suggest a model in which the HbE mutation, as a consequence of a relative change in redox properties, decreases the overall rate of Hb-mediated production of bioactive NO.


Subject(s)
Hemoglobin E/chemistry , Models, Molecular , Nitric Oxide/chemistry , Oxygen/chemistry , Allosteric Regulation/physiology , Catalysis , Crystallography, X-Ray , Hemoglobin E/genetics , Hemoglobin E/metabolism , Humans , Mutation , Nitric Oxide/metabolism , Oxidation-Reduction , Oxygen/metabolism , Structure-Activity Relationship
7.
Photosynth Res ; 106(3): 191-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21161598

ABSTRACT

We honor Steve Brody, a dear friend and a mentor on what would have been his 83rd birthday (November 29, 2010). Steve was a pioneer of chlorophyll structure and function, an outstanding biophysicist, an innovator, an artist and an adventurer, a true renaissance man. We present here first his first-of-a-kind contributions on the primary processes of photosynthesis at the University of Illinois at Urbana-Champaign, and then review his research on the interactions of chlorophyll monolayers and various photosynthetic electron donors and acceptors in artificial membrane systems at New York University. We highlight significant research contributions of interest to the reader and conclude with biographical notes.


Subject(s)
Biology/history , History, 20th Century , History, 21st Century , United States
8.
Biophys J ; 95(8): 4025-33, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18621841

ABSTRACT

Individuals expressing hemoglobin C (beta6 Glu-->Lys) present red blood cells (RBC) with intraerythrocytic crystals that form when hemoglobin (Hb) is oxygenated. Our earlier in vitro liquid-liquid (L-L) phase separation studies demonstrated that liganded HbC exhibits a stronger net intermolecular attraction with a longer range than liganded HbS or HbA, and that L-L phase separation preceded and enhanced crystallization. We now present evidence for the role of phase separation in HbC crystallization in the RBC, and the role of the RBC membrane as a nucleation center. RBC obtained from both human homozygous HbC patients and transgenic mice expressing only human HbC were studied by bright-field and differential interference contrast video-enhanced microscopy. RBC were exposed to hypertonic NaCl solution (1.5-3%) to induce crystallization within an appropriate experimental time frame. L-L phase separation occurred inside the RBC, which in turn enhanced the formation of intraerythrocytic crystals. RBC L-L phase separation and crystallization comply with the thermodynamic and kinetics laws established through in vitro studies of phase transformations. This is the first report, to the best of our knowledge, to capture a temporal view of intraerythrocytic HbC phase separation, crystal formation, and dissolution.


Subject(s)
Erythrocytes/chemistry , Hemoglobin C/chemistry , Hemoglobin C/isolation & purification , Animals , Crystallization , Cytosol , Erythrocyte Membrane/metabolism , Humans , Mice , Temperature , Time Factors
9.
J Cell Biol ; 180(5): 897-904, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18332216

ABSTRACT

Human embryonic stem cells (hESCs) are potential therapeutic tools and models of human development. With a growing interest in primary cilia in signal transduction pathways that are crucial for embryological development and tissue differentiation and interest in mechanisms regulating human hESC differentiation, demonstrating the existence of primary cilia and the localization of signaling components in undifferentiated hESCs establishes a mechanistic basis for the regulation of hESC differentiation. Using electron microscopy (EM), immunofluorescence, and confocal microscopies, we show that primary cilia are present in three undifferentiated hESC lines. EM reveals the characteristic 9 + 0 axoneme. The number and length of cilia increase after serum starvation. Important components of the hedgehog (Hh) pathway, including smoothened, patched 1 (Ptc1), and Gli1 and 2, are present in the cilia. Stimulation of the pathway results in the concerted movement of Ptc1 out of, and smoothened into, the primary cilium as well as up-regulation of GLI1 and PTC1. These findings show that hESCs contain primary cilia associated with working Hh machinery.


Subject(s)
Cell Differentiation/genetics , Cilia/ultrastructure , Embryonic Stem Cells/ultrastructure , Hedgehog Proteins/metabolism , Signal Transduction/genetics , Axoneme/genetics , Axoneme/metabolism , Axoneme/ultrastructure , Cell Line , Cell Lineage/genetics , Cilia/genetics , Cilia/metabolism , Culture Media, Serum-Free/pharmacology , Embryonic Stem Cells/metabolism , Female , Fluorescent Antibody Technique , Hedgehog Proteins/genetics , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
10.
Biophys J ; 93(3): 902-13, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17449671

ABSTRACT

Sickle cell anemia is a debilitating genetic disease that affects hundreds of thousands of babies born each year worldwide. Its primary pathogenic event is the polymerization of a mutant, sickle cell, hemoglobin (HbS); and this is one of a line of diseases (Alzheimer's, Huntington's, prion, etc.) in which nucleation initiates pathophysiology. We show that the homogeneous nucleation of HbS polymers follows a two-step mechanism with metastable dense liquid clusters serving as precursor to the ordered nuclei of the HbS polymer. The evidence comes from data on the rates of fiber nucleation and growth and nucleation delay times, the interaction of fibers with polarized light, and mesoscopic metastable HbS clusters in solution. The presence of a precursor in the HbS nucleation mechanism potentially allows low-concentration solution components to strongly affect the nucleation kinetics. The variations of these concentrations in patients might account for the high variability of the disease in genetically identical patients. In addition, these components can potentially be utilized for control of HbS polymerization and treatment of the disease.


Subject(s)
Hemoglobin, Sickle/chemistry , Anemia, Sickle Cell/blood , Erythrocytes/chemistry , Hemoglobin, Sickle/isolation & purification , Humans , Kinetics , Macromolecular Substances/chemistry , Models, Molecular , Protein Conformation
11.
Article in English | MEDLINE | ID: mdl-17364471

ABSTRACT

Natural acellular polymeric hemoglobins (Hb) provide oxygen transport and delivery within many terrestrial and marine invertebrate organisms. It has been our premise that these natural acellular Hbs may serve as models of therapeutic hemoglobin-based oxygen carriers (HBOC). Our attention has focused on the acellular Hb from the terrestrial invertebrate, Lumbricus terrestris (Lt), which possesses a unique hierarchical structure and a unique ability to function extracellularly without oxidative damage. Lumbricus Hb and Arenicola Hb are resistant to autoxidation, chemical oxidation by potassium ferricyanide, and have little or no capacity to transfer electrons to Fe(+3)-complexes at 37 degrees C. An understanding of how these invertebrate acellular oxygen carriers maintain their structural integrity and redox stability in vivo is vital for the design of a safe and effective red cell substitute. We report here a positive redox potential for these giant hemoglobins that may lie at the basis for its resistance to oxidation.


Subject(s)
Blood Substitutes/metabolism , Hemoglobins/metabolism , Oxygen/metabolism , Animals , Blood Substitutes/chemistry , Drug Stability , Electrochemistry , Hemoglobins/chemistry , Invertebrates , Models, Biological , Oligochaeta , Oxidation-Reduction
12.
Exp Hematol ; 34(12): 1635-42, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157159

ABSTRACT

OBJECTIVE: To develop a method to produce in culture large number of erythroid cells from human embryonic stem cells. MATERIALS AND METHODS: Human H1 embryonic stem cells were differentiated into hematopoietic cells by coculture with a human fetal liver cell line, and the resulting CD34-positive cells were expanded in vitro in liquid culture using a three-step method. The erythroid cells produced were then analyzed by light microscopy and flow cytometry. Globin expression was characterized by quantitative reverse-transcriptase polymerase chain reaction and by high-performance liquid chromatography. RESULTS: CD34-positive cells produced from human embryonic stem cells could be efficiently differentiated into erythroid cells in liquid culture leading to a more than 5000-fold increase in cell number. The erythroid cells produced are similar to primitive erythroid cells present in the yolk sac of early human embryos and did not enucleate. They are fully hemoglobinized and express a mixture of embryonic and fetal globins but no beta-globin. CONCLUSIONS: We have developed an experimental protocol to produce large numbers of primitive erythroid cells starting from undifferentiated human embryonic stem cells. As the earliest human erythroid cells, the nucleated primitive erythroblasts, are not very well characterized because experimental material at this stage of development is very difficult to obtain, this system should prove useful to answer a number of experimental questions regarding the biology of these cells. In addition, production of mature red blood cells from human embryonic stem cells is of great potential practical importance because it could eventually become an alternate source of cell for transfusion.


Subject(s)
Erythrocytes/cytology , Stem Cells/cytology , Animals , Antigens, CD34/analysis , Cell Culture Techniques/methods , Cell Differentiation , Cell Line , Cells, Cultured , Coculture Techniques , Erythrocytes/physiology , Fetal Blood/cytology , Fetal Blood/physiology , Gene Expression Profiling , Globins/genetics , Humans , Liver/cytology , Liver/physiology , Mice , Reverse Transcriptase Polymerase Chain Reaction/methods , Stem Cells/physiology
13.
FEBS Lett ; 580(18): 4485-90, 2006 Aug 07.
Article in English | MEDLINE | ID: mdl-16860794

ABSTRACT

Transgenic mouse models of hemoglobinopathies unravel pathophysiological mechanisms; yet the validity of the red blood cell (RBC) model of human hemoglobin (hHb) enveloped by a mouse (m) membrane has been questioned. Isoelectric focusing of hHb and mHb from transgenic mRBC shows a greater association of mHb to the mouse membrane compared to normal hHbA, supporting a species-specific Hb-mRBC membrane interaction. Enhanced hmutant Hb (HbE, HbS and HbC)-mRBC membrane affinities correlates with enhanced membrane lipid peroxidation and parallel those reported in hRBC, lending support to transgenic mRBC as models of hemoglobinopathies. Species-specific Hb-membrane interaction may be overridden by Hb charge and conformational alterations.


Subject(s)
Erythrocyte Membrane/metabolism , Hemoglobins/metabolism , Animals , Hemoglobin A/metabolism , Hemoglobins/genetics , Humans , Lipid Peroxidation , Mice , Mice, Transgenic , Mutation , Species Specificity
14.
Free Radic Res ; 40(3): 285-94, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16484045

ABSTRACT

Fluorescence emission of free protoporphyrin IX (PPIX, em. approximately 626 nm), zinc protoporphyrin IX (ZPP, em. approximately 594 nm) and fluorescent heme degradation product (FHDP, em. approximately 466 nm) are identified and simultaneously detected in mouse and human red cell hemolysates, when excited at 365 nm. A novel method is established for comparing relative FHDP, PPIX and ZPP levels in hemolysates without performing red cell porphyrin extractions. The ZPP fluorescence directly measured in hemolysates (F(365/594)) correlates with the ZPP fluorescence obtained from acetone/water extraction (R(2) = 0.9515, P < 0.0001). The relative total porphyrin (ZPP and PPIX) fluorescence obtained from direct hemolysate fluorescence measurements also correlates with red blood cell total porphyrins determined by ethyl acetate extraction (Piomelli extraction, R(2) = 0.88, P < 0.0001). These fluorescent species serves as biomarkers for alterations in Hb synthesis and Hb stability.


Subject(s)
Erythrocytes/metabolism , Heme/metabolism , Protoporphyrins/blood , Animals , Erythrocytes/drug effects , Fluorescence , Hemoglobin E/metabolism , Hydrogen Peroxide/pharmacology , Mice , Mice, Transgenic , Oxidants/pharmacology
15.
Acta Crystallogr D Biol Crystallogr ; 61(Pt 5): 566-73, 2005 May.
Article in English | MEDLINE | ID: mdl-15858266

ABSTRACT

Human hemoglobin binds oxygen cooperatively and functions as a tetramer composed of two identical alphabeta heterodimers. While human hemoglobin is the best characterized allosteric protein, the quaternary R (oxygenated or liganded) to T (deoxygenated) structural transition remains controversial. The R2 state has been postulated to represent either an intermediate or final quaternary state elicited by ligand binding. However, the biological relevance of the R2 state has been questioned as it has not been observed crystallographically under physiological conditions. The high-resolution R2 quaternary structures of human COHbC (betaE6K) and COHbS (betaE6V) are reported at neutral pH and low ionic strength using PEG 4000 as a precipitant. Crystals of COHbC, COHbS and their mixtures are isomorphous, indicating that they share the same tertiary and quaternary structures. In contrast, oxyHbA or COHbA did not yield crystals at neutral pH under similar conditions. Solubility studies and modeling suggest that at neutral pH and low ionic strength the beta6 mutant hemoglobins crystallize (betaK6 > betaV6) as a result of more favorable lattice contacts.


Subject(s)
Carboxyhemoglobin/chemistry , Hemoglobin, Sickle/chemistry , Carboxyhemoglobin/genetics , Chemical Phenomena , Chemistry, Physical , Crystallization , Crystallography, X-Ray , Hemoglobin, Sickle/genetics , Humans , Hydrogen-Ion Concentration , Indicators and Reagents , Models, Molecular , Mutation , Polyethylene Glycols , Solubility
16.
Biophys J ; 88(3): 2057-67, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15626716

ABSTRACT

Effector binding to liganded hemoglobin (Hb) provides a new understanding of structural determinants of Hb function. L35, a bezafibrate-related compound, is one of the more potent synthetic regulators of Hb oxygen (O(2)) affinity. In the presence of inositol hexaphosphate and bezafibrate (or derivatives), liganded Hb at low pH (pH approximately 6.5) exhibits extremely low O(2) affinity and very low cooperativity. In this study, the nature of L35 binding to COHbA at pH 6.35, an altered R-state, is presented. Solution-active site-specific spectroscopic probings by front-face fluorescence and circular dichroism reveal that L35 induces a global heterogeneous conformation in COHbA at pH 6.35 that includes: a T-like structural feature at the alpha1beta2 interface; an R-like structural feature within the heme environment; and an intermediate-like state at the central cavity. These long-range structural perturbations appear to stem from L35 binding to two classes of binding sites: the central cavity (primarily at the alphaalpha cleft) and the surface. These results indicate that L35 induces an allosteric transition species, characterized by domain-specific tertiary and quaternary-like conformation within a global R-quaternary structure.


Subject(s)
Hemoglobins/chemistry , Phenylurea Compounds/chemistry , Binding Sites , Hemoglobins/analysis , Ligands , Protein Binding , Protein Conformation , Spectrometry, Fluorescence , Stereoisomerism , Structure-Activity Relationship , Surface Plasmon Resonance
17.
Blood Cells Mol Dis ; 33(3): 303-7, 2004.
Article in English | MEDLINE | ID: mdl-15528149

ABSTRACT

Hemoglobin E (HbE, beta26 Glu-->Lys) is the most common abnormal Hb variant in the world, and found in greatest frequency in Southeast (SE) Asia. In the United States, HbE is the third most prevalent variant (after HbS and HbC); and its now increasing frequency is due to immigration from SE Asia. HbE homozygotes present a benign clinical picture, but when HbE is coupled with beta0-thalassemia or HbS, variably severe hemoglobinopathies arise. To date, there are no transgenic animal models of HbE-related diseases. We report here the creation of transgenic mice expressing human HbE as a step toward creating animal models for HbE-related diseases. The betaE mice exhibit red blood cell hypochromia and target cells consistent with those observed in human patients exhibiting HbE trait. Furthermore, the transgenic HbE hemolysates contain increased amounts of Hb oxidation products.


Subject(s)
Gene Expression Regulation/genetics , Hemoglobin E/genetics , Homozygote , beta-Thalassemia/genetics , Animals , Disease Models, Animal , Mice , Mice, Transgenic , beta-Thalassemia/pathology
18.
Biophys J ; 87(4): 2621-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15454456

ABSTRACT

Crystallization of the mutated hemoglobin, HbC, which occurs inside red blood cells of patients expressing betaC-globin and exhibiting the homozygous CC and the heterozygous SC (in which two mutant beta-globins, S and C, are expressed) diseases, is a convenient model for processes underlying numerous condensation diseases. As a first step, we investigated the molecular-level mechanisms of crystallization of this protein from high-concentration phosphate buffer in its stable carbomonoxy form using high-resolution atomic force microscopy. We found that in conditions of equilibrium with the solution, the crystals' surface reconstructs into four-molecule-wide strands along the crystallographic a (or b) axis. However, the crystals do not grow by the alignment of such preformed strands. We found that the crystals grow by the attachment of single molecules to suitable sites on the surface. These sites are located along the edges of new layers generated by two-dimensional nucleation or by screw dislocations. During growth, the steps propagate with random velocities, with the mean being an increasing function of the crystallization driving force. These results show that the crystallization mechanisms of HbC are similar to those found for other proteins. Therefore, strategies developed to control protein crystallization in vitro may be applicable to pathology-related crystallization systems.


Subject(s)
Crystallization/methods , Hemoglobin C/chemistry , Hemoglobin C/ultrastructure , Microscopy, Atomic Force , Hemoglobin C/analysis , Multiprotein Complexes/chemistry , Protein Conformation
19.
Biophys J ; 86(3): 1702-12, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14990498

ABSTRACT

Reversible liquid-liquid (L-L) phase separation in the form of high concentration hemoglobin (Hb) solution droplets is favored in an equilibrium with a low-concentration Hb solution when induced by inositol-hexaphosphate in the presence of polyethylene glycol 4000 at pH 6.35 HEPES (50 mM). The L-L phase separation of Hb serves as a model to elucidate intermolecular interactions that may give rise to accelerated nucleation kinetics of liganded HbC (beta6 Lys) compared to HbS (beta6 Val) and HbA (beta6 Glu). Under conditions of low pH (pH 6.35) in the presence of inositol-hexaphosphate, COHb assumes an altered R-state. The phase lines for the three Hb variants in concentration and temperature coordinates indicate that liganded HbC exhibits a stronger net intermolecular attraction with a longer range than liganded HbS and HbA. Over time, L-L phase separation gives rise to amorphous aggregation and subsequent formation of crystals of different kinetics and habits, unique to the individual Hb. The composite of R- and T-like solution aggregation behavior indicates that this is a conformationally driven event. These results indicate that specific contact sites, thermodynamics, and kinetics all play a role in L-L phase separation and differ for the beta6 mutant hemoglobins compared to HbA. In addition, the dense liquid droplet interface or aggregate interface noticeably participates in crystal nucleation.


Subject(s)
Crystallization/methods , Hemoglobins/chemistry , Hemoglobins/ultrastructure , Phytic Acid/chemistry , Polyethylene Glycols/chemistry , Amino Acid Substitution , Binding Sites , Macromolecular Substances , Mutation , Phase Transition , Protein Binding , Protein Conformation , Solubility , Solutions , Structure-Activity Relationship , Temperature
20.
Br J Haematol ; 122(2): 317-25, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12846903

ABSTRACT

The liganded (R-state) form of sickle cell haemoglobin (HbS) is of particular relevance at non-polymerizing concentrations as oxy HbS exhibits unusual properties compared with oxy HbA: mechanical precipitability (resulting from surface denaturation), greater unfolding at an air-water interface and a tendency to oxidize more readily. In human haemoglobins, the beta7 (A4) Glu residue forms an intrachain salt bridge with beta132 (H10) Lys in both liganded and deoxy structures. In the present study, recombinant haemoglobins with substitutions in the beta7 and beta132 sites were studied in order to determine the role of the beta7-beta132 salt bridge on Hb conformational integrity and stability. The elimination of this interhelix bridge correlates with enhanced surface denaturation and conformational alterations in the central cavity 2,3-diphosphoglycerate (DPG) cleft and alpha1beta2 interface. The A-helix beta7 Ala substitution generates a class of conformational change at the DPG pocket and alpha1beta2 interface that is distinct from that dictated by the H-helix beta132 Ala substitution. These results are significant with regard to the communication pathway between the alpha1beta1 and alpha1beta2 interfaces, and the new understanding of Hb allostery dependent upon tertiary structural constraints caused by effector binding to the R-state.


Subject(s)
Hemoglobins, Abnormal/genetics , Fluorometry , Hemoglobin A , Hemoglobin, Sickle , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL