Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Curr Opin Hematol ; 28(3): 171-176, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33631786

ABSTRACT

PURPOSE OF REVIEW: Sickle cell disease (SCD) is a hemolytic anemia caused by a point mutation in the ß globin gene leading to the expression of an abnormal hemoglobin (HbS) that polymerizes under hypoxic conditions driving red cell sickling. Circulating red cells have been extensively characterized in SCD, as their destruction and removal from peripheral blood are the major contributors to anemia. However, few reports showed cellular abnormalities during erythropoiesis in SCD, suggesting that anemia could also be influenced by defects of central origin. RECENT FINDINGS: El Hoss et al. demonstrated ineffective erythropoiesis (IE) in SCD and deciphered the molecular mechanism underlying cell death during the hemoglobin synthesis phase of terminal differentiation. They showed that HbS polymerization induces apoptosis of differentiating erythroblasts and that fetal hemoglobin rescues these cells through its antipolymerization function. SUMMARY: IE is the major cause of anemia in ß-thalassemia patients, and it is generally surmised that it contributes little to anemia of SCD. Recent reports demonstrate the occurrence of IE in SCD patients and show important alterations in the hematopoietic and erythroid niches, both in SCD patients and in the humanized Townes SCD mouse model. This implies that therapeutic strategies initially designed to improve red cell survival in the circulation of SCD patients would also positively impact erythropoiesis and bone marrow cellularity.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/etiology , Erythropoiesis , Anemia, Sickle Cell/diagnosis , Anemia, Sickle Cell/therapy , Animals , Apoptosis , Cellular Microenvironment , Disease Management , Disease Models, Animal , Disease Susceptibility , Erythrocyte Indices , Erythrocytes/metabolism , Erythropoiesis/genetics , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Expression Regulation , Hemoglobins/genetics , Humans , Mutation , Protein Multimerization , beta-Globins/genetics
2.
Proc Natl Acad Sci U S A ; 117(26): 15018-15027, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32527859

ABSTRACT

The pathology of sickle cell disease is caused by polymerization of the abnormal hemoglobin S upon deoxygenation in the tissues to form fibers in red cells, causing them to deform and occlude the circulation. Drugs that allosterically shift the quaternary equilibrium from the polymerizing T quaternary structure to the nonpolymerizing R quaternary structure are now being developed. Here we update our understanding on the allosteric control of fiber formation at equilibrium by showing how the simplest extension of the classic quaternary two-state allosteric model of Monod, Wyman, and Changeux to include tertiary conformational changes provides a better quantitative description. We also show that if fiber formation is at equilibrium in vivo, the vast majority of cells in most tissues would contain fibers, indicating that it is unlikely that the disease would be survivable once the nonpolymerizing fetal hemoglobin has been replaced by adult hemoglobin S at about 1 y after birth. Calculations of sickling times, based on a recently discovered universal relation between the delay time prior to fiber formation and supersaturation, show that in vivo fiber formation is very far from equilibrium. Our analysis indicates that patients survive because the delay period allows the majority of cells to escape the small vessels of the tissues before fibers form. The enormous sensitivity of the duration of the delay period to intracellular hemoglobin composition also explains why sickle trait, the heterozygous condition, and the compound heterozygous condition of hemoglobin S with pancellular hereditary persistence of fetal hemoglobin are both relatively benign conditions.


Subject(s)
Anemia, Sickle Cell/metabolism , Hemoglobin, Sickle/chemistry , Oxygen/metabolism , Allosteric Regulation , Erythrocytes/chemistry , Erythrocytes/metabolism , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/metabolism , Hemoglobin, Sickle/metabolism , Humans , Kinetics , Oxygen/chemistry
3.
Subcell Biochem ; 94: 275-296, 2020.
Article in English | MEDLINE | ID: mdl-32189304

ABSTRACT

During the past two decades, significant advances have been made in our understanding of the human fetal and embryonic hemoglobins made possible by the availability of pure, highly characterized materials and novel methods, e.g., nano gel filtration, to study their properties and to correct some misconceptions. For example, whereas the structures of the human adult, fetal, and embryonic hemoglobins are very similar, it has generally been assumed that functional differences between them are due to primary sequence effects. However, more recent studies indicate that the strengths of the interactions between their subunits are very different leading to changes in their oxygen binding properties compared to adult hemoglobin. Fetal hemoglobin in the oxy conformation is a much stronger tetramer than adult hemoglobin and dissociates to dimers 70-times less than adult hemoglobin. This property may form the basis for its protective effect against malaria. A major source of the increased strength of fetal hemoglobin resides within the A-helix of its gamma subunit as demonstrated in studies with the hybrid hemoglobin Felix and related hybrids. Re-activating fetal hemoglobin synthesis in vivo is currently a major focus of clinical efforts designed to treat sickle cell anemia since it inhibits the aggregation of sickle hemoglobin. The mechanisms for both the increased oxygen affinity of fetal hemoglobin and its decreased response to DPG have been clarified. Acetylated fetal hemoglobin, which makes up 10-20% of total fetal hemoglobin, has a significantly weakened tetramer structure suggesting a similar role for other kinds of protein acetylation. Embryonic hemoglobins have the weakest tetramer and dimer structures. In general, the progressively increasing strength of the subunit interfaces of the hemoglobin family during development from the embryonic to the fetal and ultimately to the adult types correlates with their temporal appearance and disappearance in vivo, i.e., ontogeny.


Subject(s)
Embryo, Mammalian/blood supply , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/metabolism , Oxygen/metabolism , Fetus/blood supply , Humans
4.
Haematologica ; 104(9): 1720-1730, 2019 09.
Article in English | MEDLINE | ID: mdl-31399526

ABSTRACT

The complex, frequently devastating, multi-organ pathophysiology of sickle cell disease has a single root cause: polymerization of deoxygenated sickle hemoglobin. A logical approach to disease modification is, therefore, to interdict this root cause. Ideally, such interdiction would utilize small molecules that are practical and accessible for worldwide application. Two types of such small molecule strategies are actively being evaluated in the clinic. The first strategy intends to shift red blood cell precursor hemoglobin manufacturing away from sickle hemoglobin and towards fetal hemoglobin, which inhibits sickle hemoglobin polymerization by a number of mechanisms. The second strategy intends to chemically modify sickle hemoglobin directly in order to inhibit its polymerization. Important lessons have been learnt from the pre-clinical and clinical evaluations to date. Open questions remain, but this review summarizes the valuable experience and knowledge already gained, which can guide ongoing and future efforts for molecular mechanism-based, practical and accessible disease modification of sickle cell disease.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/physiopathology , Fetal Hemoglobin/drug effects , Hemoglobin, Sickle/drug effects , Antisickling Agents/therapeutic use , DNA Methylation , Drug Design , Epigenesis, Genetic , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/metabolism , Hemoglobin, Sickle/chemistry , Hemoglobin, Sickle/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxyurea/therapeutic use , Polymerization , gamma-Globins/metabolism
5.
Blood Adv ; 3(9): 1493-1498, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31072835

ABSTRACT

Fetal hemoglobin (HbF) induction therapy has become the most promising strategy for treating ß-hemoglobinopathies, including sickle-cell diseases and ß-thalassemia. However, subtle but critical structural difference exists between HbF and normal adult hemoglobin (HbA), which inevitably leads to reduced binding of the endogenous modulator 2,3-bisphosphoglycerate (2,3-BPG) to HbF and thus increased oxygen affinity and decreased oxygen transport efficiency of HbF. We combined the oxygen equilibrium experiments, resonance Raman (RR) spectroscopy, and molecular docking modeling, and we discuss 2 phthalides, z-butylidenephthalide and z-ligustilide, that can effectively lower the oxygen affinity of HbF. They adjust it to a level closer to that of HbA and make it a more satisfactory oxygen carrier for adults. From the oxygen equilibrium curve measurements, we show that the 2 phthalides are more effective than 2,3-BPG for modulating HbF. The RR spectra show that phthalides allosterically stabilize the oxygenated HbF in the low oxygen affinity conformation, and the molecular docking modeling reveals that the 2 chosen phthalides interact with HbF via the cleft around the γ1/γ2 interface with a binding strength ∼1.6 times stronger than that of 2,3-BPG. We discuss the implications of z-butylidenephthalide and z-ligustilide in boosting the efficacy of HbF induction therapy to mitigate the clinical severities of ß-hemoglobinopathies.


Subject(s)
Benzofurans/therapeutic use , Fetal Hemoglobin/metabolism , Hemoglobinopathies/drug therapy , 2,3-Diphosphoglycerate/metabolism , Benzofurans/chemistry , Benzofurans/metabolism , Binding Sites , Catalytic Domain , Fetal Hemoglobin/chemistry , Humans , Molecular Docking Simulation , Oxygen/chemistry , Oxygen/metabolism , Protein Binding , Spectrum Analysis, Raman
6.
J Biomol Struct Dyn ; 37(14): 3848-3857, 2019 09.
Article in English | MEDLINE | ID: mdl-30295130

ABSTRACT

Quasi-palindromic sequences (AT)XN12(AT)Y present in HS2 (hypersensitive site 2) of the human ß-globin locus are known to be significantly associated with increased fetal hemoglobin (HbF) levels. High HbF levels in some adults arise due to pathological conditions such as sickle cell disease and ß-thalassemia. However, elevated levels of HbF are also associated with a reducing morbidity and mortality in patients with ß-thalassemia and thus ameliorate the severity of the disease. Using gel-electrophoresis, ultraviolet (UV)-thermal denaturation, and circular dichroism (CD) techniques, we demonstrated that it exhibits a hairpin-duplex equilibrium. Intramolecular species (hairpin) were observed in both low and high salt concentrations in gel assay studies displaying the unusual stability of intramolecular species even at the high counter-ion concentration. The unusual stability of hairpin secondary structures was also demonstrated by the monophasic nature of the melting profiles for the oligonucleotides which persisted at low as well as high salt and oligomer concentrations. Change in CD spectra as a function of oligomer concentration indicates that the bimolecular duplex formation is selectively favored over monomolecular hairpin formation at and above 9 µM oligomer concentration. Thus, we hypothesize that imperfect inverted repeat sequence (AT)XN12(AT)Y of HS2 of ß-globin gene LCR forms the unusually stable hairpins which may result in the formation of a cruciform structure that may be recruited for binding by various nuclear proteins that could result in elevated HbF levels. Communicated by Ramaswamy H. Sarma.


Subject(s)
Fetal Hemoglobin/genetics , Nucleotide Motifs/genetics , Circular Dichroism , Fetal Hemoglobin/chemistry , Humans , Models, Molecular , Nucleic Acid Conformation , Nucleic Acid Denaturation , Temperature
7.
Redox Biol ; 19: 218-225, 2018 10.
Article in English | MEDLINE | ID: mdl-30193183

ABSTRACT

Redox active cysteine residues including ßCys93 are part of hemoglobin's "oxidation hotspot". Irreversible oxidation of ßCys93 ultimately leads to the collapse of the hemoglobin structure and release of heme. Human fetal hemoglobin (HbF), similarly to the adult hemoglobin (HbA), carries redox active γCys93 in the vicinity of the heme pocket. Site-directed mutagenesis has been used in this study to examine the impact of removal and/or addition of cysteine residues in HbF. The redox activities of the recombinant mutants were examined by determining the spontaneous autoxidation rate, the hydrogen peroxide induced ferric to ferryl oxidation rate, and irreversible oxidation of cysteine by quantitative mass spectrometry. We found that substitution of γCys93Ala resulted in oxidative instability characterized by increased oxidation rates. Moreover, the addition of a cysteine residue at α19 on the exposed surface of the α-chain altered the regular electron transfer pathway within the protein by forming an alternative oxidative site. This may also create an accessible site for di-sulfide bonding between Hb subunits. Engineering of cysteine residues at suitable locations may be useful as a tool for managing oxidation in a protein, and for Hb, a way to stave off oxidation reactions resulting in a protein structural collapse.


Subject(s)
Cysteine/genetics , Fetal Hemoglobin/genetics , Cysteine/chemistry , Fetal Hemoglobin/chemistry , Humans , Models, Molecular , Mutagenesis, Site-Directed , Oxidation-Reduction , Oxidative Stress , Protein Stability , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
8.
Br J Haematol ; 180(2): 189-200, 2018 01.
Article in English | MEDLINE | ID: mdl-29143315

ABSTRACT

Fetal haemoglobin (HbF, α2γ2) induction has long been an area of investigation, as it is known to ameliorate the clinical complications of sickle cell disease (SCD). Progress in identifying novel HbF-inducing strategies has been stymied by limited understanding of gamma (γ)-globin regulation. Genome-wide association studies (GWAS) have identified variants in BCL11A and HBS1L-MYB that are associated with HbF levels. Functional studies have established the roles of BCL11A, MYB, and KLF1 in γ-globin regulation, but this information has not yielded new pharmacological agents. Several drugs are under investigation in clinical trials as HbF-inducing agents, but hydroxycarbamide remains the only widely used pharmacologic therapy for SCD. Autologous transplant of edited haematopoietic stem cells holds promise as a cure for SCD, either through HbF induction or correction of the causative mutation, but several technical and safety hurdles must be overcome before this therapy can be offered widely, and pharmacological therapies are still needed.


Subject(s)
Anemia, Sickle Cell/therapy , Fetal Hemoglobin/biosynthesis , Anemia, Sickle Cell/etiology , Anemia, Sickle Cell/metabolism , Animals , Antisickling Agents/pharmacology , Antisickling Agents/therapeutic use , Clinical Trials as Topic , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/genetics , Gene Expression Regulation/drug effects , Genetic Therapy , Humans , Treatment Outcome
9.
Adv Exp Med Biol ; 977: 367-373, 2017.
Article in English | MEDLINE | ID: mdl-28685467

ABSTRACT

The worldwide blood shortage has generated demands for alternatives to transfusible human blood. One such important option is based on recombinant hemoglobin-based oxygen carriers (rHBOCs). Most efforts have been focused on various E. coli based production systems. One of the key challenges in these systems is to devise an efficient and economical protein production strategy involving selection of suitable host cell and Hb variant, growth conditions and media engineering. Hb also influences the heterologous host cell metabolism and therefore the identification of modified protein-protein interactions is critical for optimizing Hb production. In this study, molecularly imprinted polymers (MIPs) directed against Hb were used to identify the human Hb protein interaction network in E. coli. One E. coli host protein, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), interacted strongly with Hb, especially fetal Hb (HbF).


Subject(s)
Hemoglobins/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Recombinant Proteins/metabolism , Adult , Escherichia coli/metabolism , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/metabolism , Hemoglobins/chemistry , Humans , Mass Spectrometry , Models, Molecular , Polymers/chemistry , Polymers/metabolism , Protein Binding , Recombinant Proteins/chemistry
10.
Redox Biol ; 12: 114-120, 2017 08.
Article in English | MEDLINE | ID: mdl-28222378

ABSTRACT

Hemoglobin (Hb) is well protected inside the red blood cells (RBCs). Upon hemolysis and when free in circulation, Hb can be involved in a range of radical generating reactions and may thereby attack several different biomolecules. In this study, we have examined the potential damaging effects of cell-free Hb on plasmid DNA (pDNA). Hb induced cleavage of supercoiled pDNA (sc pDNA) which was proportional to the concentration of Hb applied. Almost 70% of sc pDNA was converted to open circular or linear DNA using 10µM of Hb in 12h. Hb can be present in several different forms. The oxy (HbO2) and met forms are most reactive, while the carboxy-protein shows only low hydrolytic activity. Hemoglobin A (HbA) could easily induce complete pDNA cleavage while fetal hemoglobin (HbF) was three-fold less reactive. By inserting, a redox active cysteine residue on the surface of the alpha chain of HbF by site-directed mutagenesis, the DNA cleavage reaction was enhanced by 82%. Reactive oxygen species were not directly involved in the reaction since addition of superoxide dismutase and catalase did not prevent pDNA cleavage. The reactivity of Hb with pDNA can rather be associated with the formation of protein based radicals.


Subject(s)
DNA Cleavage , DNA/chemistry , Fetal Hemoglobin/metabolism , Hemoglobin A/metabolism , Adult , Cell-Free System , Cysteine/genetics , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/genetics , Hemoglobin A/chemistry , Hemoglobin A/genetics , Humans , Mutagenesis, Site-Directed , Plasmids/genetics , Protein Structure, Secondary , Reactive Oxygen Species/metabolism
11.
Biochemistry ; 56(10): 1444-1459, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28218841

ABSTRACT

Removal of heme from human hemoglobin (Hb) results in formation of an apoglobin heterodimer. Titration of this apodimer with guanidine hydrochloride (GdnHCl) leads to biphasic unfolding curves indicating two distinct steps. Initially, the heme pocket unfolds and generates a dimeric intermediate in which ∼50% of the original helicity is lost, but the α1ß1 interface is still intact. At higher GdnHCl concentrations, this intermediate dissociates into unfolded monomers. This structural interpretation was verified by comparing GdnHCl titrations for adult human hemoglobin A (HbA), recombinant fetal human hemoglobin (HbF), recombinant Hb cross-linked with a single glycine linker between the α chains, and recombinant Hbs with apolar heme pocket mutations that markedly stabilize native conformations in both subunits. The first phase of apoHb unfolding is independent of protein concentration, little affected by genetic cross-linking, but significantly shifted toward higher GdnHCl concentrations by the stabilizing distal pocket mutations. The second phase depends on protein concentration and is shifted to higher GdnHCl concentrations by genetic cross-linking. This model for apoHb unfolding allowed us to quantitate subtle differences in stability between apoHbA and apoHbF, which suggest that the ß and γ heme pockets have similar stabilities, whereas the α1γ1 interface is more resistant to dissociation than the α1ß1 interface.


Subject(s)
Apoproteins/chemistry , Fetal Hemoglobin/chemistry , Guanidine/chemistry , Hemoglobin A/chemistry , Hemoglobins/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Gene Expression , Glycine/chemistry , Glycine/metabolism , Heme/chemistry , Heme/isolation & purification , Heme/metabolism , Hemoglobin A/genetics , Hemoglobin A/metabolism , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Kinetics , Protein Denaturation , Protein Domains , Protein Folding , Protein Multimerization , Protein Stability , Protein Structure, Secondary , Protein Unfolding , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
12.
Expert Rev Hematol ; 9(12): 1129-1137, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27801605

ABSTRACT

INTRODUCTION: Stress erythropoiesis induces fetal hemoglobin (HbF) expression in ß-thalassemias, however the level of expression is highly variable. The last decade has seen dramatic advances in our understanding of the molecular regulators of HbF production and the genetic factors associated with HbF levels, leading to the promise of new methods of the clinical induction of HbF. Areas covered: This article will review the heterogeneity and genetic modifiers of HbF and HbF induction therapy in ß-thalassemia. Expert commentary: One promising curative ß-thalassemia therapy is to induce HbF synthesis in ß-thalassemic erythrocytes to therapeutic levels before clinical symptom occurs. Further understanding of HbF level variation and regulation is needed in order to predict the response from HbF-inducing approaches.


Subject(s)
Fetal Hemoglobin/genetics , Gene Expression Regulation , beta-Thalassemia/genetics , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Epistasis, Genetic , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/metabolism , Gene Editing , Gene Expression Regulation/drug effects , Genetic Heterogeneity , Genetic Therapy , Hemoglobin Subunits/chemistry , Hemoglobin Subunits/genetics , Hemoglobin Subunits/metabolism , Humans , Hydroxyurea/pharmacology , Hydroxyurea/therapeutic use , Multigene Family , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Multimerization , Repressor Proteins , beta-Thalassemia/diagnosis , beta-Thalassemia/metabolism , beta-Thalassemia/therapy
13.
Int J Lab Hematol ; 37(5): 668-72, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25989219

ABSTRACT

INTRODUCTION: Among hemoglobin (Hb) H disease cases diagnosed by DNA testing in our hemoglobinopathy laboratory, we have noted instances of unreported Hb H from high-performance liquid chromatography (HPLC) results of referring laboratories. METHODS: To characterize these issues, we identified all cases of genotypic Hb H disease diagnosed in our laboratory. HPLC chromatograms were reviewed to determine the presence and retention time of the Hb H peak. RESULTS: Hemoglobin H was not reported in 24.2% of patients (23 of 95) with genotypic Hb H disease. The characteristic prerun peak of Hb H was present on review of all eight Variant or Variant II ß-thalassemia short-program chromatograms. Elevated Hb F (≥3%) was reported in 14 cases. The Hb H peak was found in the Hb F window in 11 dual program cases. The incorrect identification of Hb H as elevated Hb F resulted in two testing referrals for 'δß-thalassemia'. CONCLUSIONS: Hemoglobin H may go unreported due to failure to examine for or recognize its peak on Variant or Variant II ß-thalassemia short-program chromatograms. Elution of Hb H in the Hb F window resulted in misidentification of Hb H for Hb F and may indicate a Variant II HbA2 /HbA1C program software error. Our findings highlight the need for careful chromatogram inspection and clinical correlation in the diagnosis of Hb H disease.


Subject(s)
Chromatography, High Pressure Liquid , Hemoglobin H/chemistry , alpha-Thalassemia/diagnosis , Adult , Chromatography, High Pressure Liquid/methods , Erythrocyte Indices , Female , Fetal Hemoglobin/chemistry , Genotype , Hemoglobin H/genetics , Humans , Male , Middle Aged , Young Adult , alpha-Globins/chemistry , alpha-Globins/genetics , alpha-Thalassemia/blood , alpha-Thalassemia/genetics
14.
Int J Lab Hematol ; 37(3): 420-5, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25324031

ABSTRACT

INTRODUCTION: Capillary zone electrophoresis (CZE) at alkaline pH is one of the techniques used for hemoglobinopathy screening. In this study, an evaluation of the performance of a lower throughput CZE instrument, the Sebia Minicap Flex Piercing system, for this purpose is reported for the first time. METHODS: The analytical performance of the Sebia Minicap Flex Piercing system was evaluated. Furthermore, a method comparison between the Sebia Minicap Flex Piercing and two HPLC methods, that is, the Bio-Rad Variant Classic(™) and the Bio-Rad D-10(™) systems was performed by measuring samples with and without clinically relevant hemoglobin disorders. RESULTS: The analytical performance was acceptable for the determination of HbA, HbA2, HbS, and HbF, with an imprecision ≤2.0%. Method comparison showed a linear correlation for HbA2, HbF, and HbS measurements. Clinical concordance was acceptable when comparing CZE and HPLC. CONCLUSIONS: Lower throughput CZE using the Sebia Minicap Flex Piercing can be used for precise and accurate first line screening and follow-up of hemoglobinopathies.


Subject(s)
Electrophoresis, Capillary/methods , Hemoglobinopathies/diagnosis , Chromatography, High Pressure Liquid , Fetal Hemoglobin/chemistry , Hemoglobin A2/chemistry , Hemoglobin, Sickle/chemistry , Hemoglobinopathies/blood , Hemoglobins/chemistry , Humans , Reproducibility of Results
15.
Hemoglobin ; 38(5): 376-80, 2014.
Article in English | MEDLINE | ID: mdl-25271994

ABSTRACT

The total number of hemoglobin (Hb) variants so far reported to the HbVar database is 1598 (April 9 2014) and 130 of them are fetal Hb variants. Fetal Hb are categorized as two different subunits, (G)γ- and (A)γ-globin chains, and γ chain variants can be observed in both subunits. There are 72 (G)γ- and 58 (A)γ-globin chain variants. Most of them are clinically silent and detected during newborn screening programs in the USA and outside the USA. In this report, we discuss the molecular characteristics and diagnostic difficulties of two new γ-globin chain variants found in an African American baby with no clinical symptoms. One is a new (G)γ-globin chain variant, Hb F-Augusta GA [(G)γ59(E3)Lys → Arg; HBG2: c.179A > G] and the other one is Hb F-Port Royal-II [(A)γ125(H3)Glu → Ala; HBG1: c.377A > C].


Subject(s)
Fetal Hemoglobin/genetics , Hemoglobinopathies/genetics , Mutation , gamma-Globins/genetics , Black or African American , Amino Acid Substitution , Fathers , Female , Fetal Hemoglobin/chemistry , Georgia , Hemoglobinopathies/blood , Hemoglobinopathies/diagnosis , Hemoglobinopathies/physiopathology , Heterozygote , Homozygote , Humans , Infant, Newborn , Mothers , Neonatal Screening , Point Mutation , Severity of Illness Index , gamma-Globins/chemistry
16.
J Biol Chem ; 289(32): 22342-57, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24939847

ABSTRACT

A pathogenic V67M mutation occurs at the E11 helical position within the heme pockets of variant human fetal and adult hemoglobins (Hb). Subsequent post-translational modification of Met to Asp was reported in γ subunits of human fetal Hb Toms River (γ67(E11)Val → Met) and ß subunits of adult Hb (HbA) Bristol-Alesha (ß67(E11)Val → Met) that were associated with hemolytic anemia. Using kinetic, proteomic, and crystal structural analysis, we were able to show that the Met → Asp transformation involves heme cycling through its oxoferryl state in the recombinant versions of both proteins. The conversion to Met and Asp enhanced the spontaneous autoxidation of the mutants relative to wild-type HbA and human fetal Hb, and the levels of Asp were elevated with increasing levels of hydrogen peroxide (H2O2). Using H2(18)O2, we verified incorporation of (18)O into the Asp carboxyl side chain confirming the role of H2O2 in the oxidation of the Met side chain. Under similar experimental conditions, there was no conversion to Asp at the αMet(E11) position in the corresponding HbA Evans (α62(E11)Val → Met). The crystal structures of the three recombinant Met(E11) mutants revealed similar thioether side chain orientations. However, as in the solution experiments, autoxidation of the Hb mutant crystals leads to electron density maps indicative of Asp(E11) formation in ß subunits but not in α subunits. This novel post-translational modification highlights the nonequivalence of human Hb α, ß, and γ subunits with respect to redox reactivity and may have direct implications to α/ß hemoglobinopathies and design of oxidatively stable Hb-based oxygen therapeutics.


Subject(s)
Heme/metabolism , Hemoglobins/chemistry , Hemoglobins/metabolism , Iron/metabolism , Adult , Amino Acid Sequence , Amino Acid Substitution , Aspartic Acid/chemistry , Crystallography, X-Ray , Fetal Hemoglobin/chemistry , Fetal Hemoglobin/genetics , Fetal Hemoglobin/metabolism , Heme/chemistry , Hemoglobin A/chemistry , Hemoglobin A/genetics , Hemoglobin A/metabolism , Hemoglobins/genetics , Hemoglobins, Abnormal/chemistry , Hemoglobins, Abnormal/genetics , Hemoglobins, Abnormal/metabolism , Humans , Hydrogen Peroxide/metabolism , Iron/chemistry , Methionine/chemistry , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Oxidation-Reduction , Protein Processing, Post-Translational , Protein Subunits , Proteomics , Static Electricity
17.
J Med Assoc Thai ; 97 Suppl 4: S49-55, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24851565

ABSTRACT

Fetal blood sampling is a procedure that involves the drawing of a blood sample from the umbilical vein of the umbilical cord, which can be performed after 18 weeks gestation. Fetal blood sampling is a preferable method for prenatal diagnosis of thalassemia in second trimester or late pregnancy. Additionally, it is suggested to be performed in cases in which mosaicisms are identified by amniocentesis or chorionic villus sampling (CVS), areas where DNA analysis is not available, and when mutations of the parents are not known. Laboratory steps regarding prenatal diagnosis by fetal blood sampling were summarized, including the ensuring of fetal origin, determination of red blood cell parameters, fetal hemoglobin analysis, and finally fetal DNA analysis. The objective of this review is to present an overview of procedures in terms of benefits, laboratory interpretations, and some limitations.


Subject(s)
Fetal Blood/chemistry , Fetal Blood/cytology , Fetal Diseases/blood , Fetal Diseases/diagnosis , Prenatal Diagnosis/methods , Thalassemia/blood , Thalassemia/diagnosis , Female , Fetal Hemoglobin/analysis , Fetal Hemoglobin/chemistry , Humans , Pregnancy
18.
J Mass Spectrom ; 49(2): 178-83, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24677308

ABSTRACT

Oxidative stress plays an important role in the development of various disease processes and is a putative mechanism in the development of bronchopulmonary dysplasia, the most common complication of extreme preterm birth. Glutathione, a major endogenous antioxidant and redox buffer, also mediates cellular functions through protein thiolation. We sought to determine if post-translational thiol modification of hemoglobin F occurs in neonates by examining erythrocyte samples obtained during the first month of life from premature infants, born at 23 0/7 - 28 6/7 weeks gestational age, who were enrolled at our center in the Prematurity and Respiratory Outcomes Program (PROP). Using liquid chromatography-mass spectrometry (LC-MS), we report the novel finding of in vivo and in vitro glutathionylation of γG and γA subunits of Hgb F. Through tandem mass spectrometry (nanoLC-MS/MS), we confirmed the adduction site as the Cys-γ94 residue and through high-resolution mass spectrometry determined that the modification occurs in both γ subunits. We also identified glutathionylation of the ß subunit of Hgb A in our patient samples; we did not find modified α subunits of Hgb A or F. In conclusion, we are the first to report that glutathionylation of γG and γA of Hgb F occurs in premature infants. Additional studies of this post-translational modification are needed to determine its physiologic impact on Hgb F function and if sG-Hgb is a biomarker for clinical morbidities associated with oxidative stress in premature infants.


Subject(s)
Chromatography, Liquid/methods , Fetal Hemoglobin/analysis , Fetal Hemoglobin/chemistry , Glutathione/chemistry , Infant, Extremely Premature/blood , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Humans , Infant, Newborn , Molecular Sequence Data , Spectrometry, Mass, Electrospray Ionization
19.
Proteomics ; 14(10): 1232-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24482221

ABSTRACT

We have shown previously that liquid extraction surface analysis of dried blood spots coupled to high resolution top-down MS may be applied for the diagnosis of hemoglobin (Hb) variants FS, FAS, FC, FAC, FAD in newborn samples. The objective of the current work was to determine whether the structural variant HbE, compound heterozygote variants FSC and FSD, and ß-thalassemia were amenable to diagnosis by this approach. Anonymized residual neonatal dried blood spot samples, taken as part of the routine newborn screening program, were analyzed by liquid extraction surface analysis coupled to high resolution MS/MS. The samples had been previously screened and were known to be FAE, FSC, FSD, or ß-thalassemia. Manual analysis of the mass spectra revealed that, in all cases, the variants may be confirmed. Direct surface sampling MS should be considered as an alternative to current screening techniques for the diagnosis of Hb variants.


Subject(s)
Hemoglobins, Abnormal/chemistry , Tandem Mass Spectrometry/methods , beta-Thalassemia/diagnosis , Amino Acid Sequence , Dried Blood Spot Testing , Fetal Hemoglobin/analysis , Fetal Hemoglobin/chemistry , Hemoglobins, Abnormal/analysis , Heterozygote , Humans , Infant, Newborn , Molecular Sequence Data
20.
Blood ; 123(10): 1586-95, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24443441

ABSTRACT

In this study, we report on 8 compound heterozygotes for mutations in the key erythroid transcription factor Krüppel-like factor 1 in patients who presented with severe, transfusion-dependent hemolytic anemia. In most cases, the red cells were hypochromic and microcytic, consistent with abnormalities in hemoglobin synthesis. In addition, in many cases, the red cells resembled those seen in patients with membrane defects or enzymopathies, known as chronic nonspherocytic hemolytic anemia (CNSHA). Analysis of RNA and protein in primary erythroid cells from these individuals provided evidence of abnormal globin synthesis, with persistent expression of fetal hemoglobin and, most remarkably, expression of large quantities of embryonic globins in postnatal life. The red cell membranes were abnormal, most notably expressing reduced amounts of CD44 and, consequently, manifesting the rare In(Lu) blood group. Finally, all tested patients showed abnormally low levels of the red cell enzyme pyruvate kinase, a known cause of CNSHA. These patients define a new type of severe, transfusion-dependent CNSHA caused by mutations in a trans-acting factor (Krüppel-like factor 1) and reveal an important pathway regulating embryonic globin gene expression in adult humans.


Subject(s)
Anemia, Hemolytic/etiology , Fetal Hemoglobin/genetics , Gene Expression Regulation , Kruppel-Like Transcription Factors/genetics , Mutation , Transfusion Reaction , Adolescent , Adult , Amino Acid Sequence , Anemia, Hemolytic/blood , Anemia, Hemolytic/genetics , Child , Child, Preschool , Conserved Sequence , Erythrocyte Indices , Erythrocytes/metabolism , Female , Fetal Hemoglobin/chemistry , Gene Order , Humans , Infant , Male , Molecular Sequence Data , Protein Binding , Protein Interaction Domains and Motifs , Sequence Alignment , Young Adult , alpha-Globins/metabolism , beta-Globins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...