Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
Add more filters

Publication year range
1.
Arterioscler Thromb Vasc Biol ; 41(2): 769-782, 2021 02.
Article in English | MEDLINE | ID: mdl-33267657

ABSTRACT

OBJECTIVE: Chronic hemolysis is a hallmark of sickle cell disease (SCD) and a driver of vasculopathy; however, the mechanisms contributing to hemolysis remain incompletely understood. Although XO (xanthine oxidase) activity has been shown to be elevated in SCD, its role remains unknown. XO binds endothelium and generates oxidants as a byproduct of hypoxanthine and xanthine catabolism. We hypothesized that XO inhibition decreases oxidant production leading to less hemolysis. Approach and Results: Wild-type mice were bone marrow transplanted with control (AA) or sickle (SS) Townes bone marrow. After 12 weeks, mice were treated with 10 mg/kg per day of febuxostat (Uloric), Food and Drug Administration-approved XO inhibitor, for 10 weeks. Hematologic analysis demonstrated increased hematocrit, cellular hemoglobin, and red blood cells, with no change in reticulocyte percentage. Significant decreases in cell-free hemoglobin and increases in haptoglobin suggest XO inhibition decreased hemolysis. Myographic studies demonstrated improved pulmonary vascular dilation and blunted constriction, indicating improved pulmonary vasoreactivity, whereas pulmonary pressure and cardiac function were unaffected. The role of hepatic XO in SCD was evaluated by bone marrow transplanting hepatocyte-specific XO knockout mice with SS Townes bone marrow. However, hepatocyte-specific XO knockout, which results in >50% diminution in circulating XO, did not affect hemolysis levels or vascular function, suggesting hepatocyte-derived elevation of circulating XO is not the driver of hemolysis in SCD. CONCLUSIONS: Ten weeks of febuxostat treatment significantly decreased hemolysis and improved pulmonary vasoreactivity in a mouse model of SCD. Although hepatic XO accounts for >50% of circulating XO, it is not the source of XO driving hemolysis in SCD.


Subject(s)
Anemia, Sickle Cell/drug therapy , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Febuxostat/pharmacology , Hemodynamics/drug effects , Hemolysis/drug effects , Pulmonary Artery/drug effects , Xanthine Oxidase/antagonists & inhibitors , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/physiopathology , Animals , Disease Models, Animal , Erythrocytes/enzymology , Liver/enzymology , Male , Mice, Inbred C57BL , Mice, Knockout , Pulmonary Artery/enzymology , Pulmonary Artery/physiopathology , Ventricular Function/drug effects , Xanthine Oxidase/genetics , Xanthine Oxidase/metabolism
2.
Ann Hematol ; 100(4): 903-911, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33523291

ABSTRACT

Hyperbilirubinemia in patients with sickle cell anemia (SCA) as a result of enhanced erythrocyte destruction, lead to cholelithiasis development in a subset of patients. Evidence suggests that hyperbilirubinemia may be related to genetic variations, such as the UGT1A1 gene promoter polymorphism, which causes Gilbert syndrome (GS). Here, we aimed to determine the frequencies of UGT1A1 promoter alleles, alpha thalassemia, and ßS haplotypes and analyze their association with cholelithiasis and bilirubin levels. The UGT1A1 alleles, -3.7 kb alpha thalassemia deletion and ßS haplotypes were determined using DNA sequencing and PCR-based assays in 913 patients with SCA. The mean of total and unconjugated bilirubin and the frequency of cholelithiasis in GS patients were higher when compared to those without this condition, regardless of age (P < 0.05). Cumulative analysis demonstrated an early age-at-onset for cholelithiasis in GS genotypes (P < 0.05). Low fetal hemoglobin (HbF) levels and normal alpha thalassemia genotype were related to cholelithiasis development (P > 0.05). However, not cholelithiasis but total and unconjugated bilirubin levels were associated with ßS haplotype. These findings confirm in a large cohort that the UGT1A1 polymorphism influences cholelithiasis and hyperbilirubinemia in SCA. HbF and alpha thalassemia also appear as modulators for cholelithiasis risk.


Subject(s)
Anemia, Sickle Cell/blood , Bilirubin/blood , Cholelithiasis/etiology , Gilbert Disease/blood , Glucuronosyltransferase/physiology , Promoter Regions, Genetic/genetics , alpha-Thalassemia/blood , Adolescent , Adult , Aged , Alleles , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/genetics , Child , Child, Preschool , Cholelithiasis/blood , Cholelithiasis/genetics , Female , Fetal Hemoglobin/analysis , Genotype , Gilbert Disease/enzymology , Gilbert Disease/genetics , Glucuronosyltransferase/genetics , Haplotypes/genetics , Hemolysis , Humans , Hyperbilirubinemia/enzymology , Hyperbilirubinemia/etiology , Hyperbilirubinemia/genetics , Male , Middle Aged , Young Adult , alpha-Thalassemia/complications , alpha-Thalassemia/enzymology , alpha-Thalassemia/genetics
3.
Arterioscler Thromb Vasc Biol ; 40(5): 1220-1230, 2020 05.
Article in English | MEDLINE | ID: mdl-32160775

ABSTRACT

OBJECTIVE: Sickle cell anemia (SCA) causes chronic inflammation and multiorgan damage. Less understood are the arterial complications, most evident by increased strokes among children. Proteolytic mechanisms, biomechanical consequences, and pharmaceutical inhibitory strategies were studied in a mouse model to provide a platform for mechanistic and intervention studies of large artery damage due to sickle cell disease. Approach and Results: Townes humanized transgenic mouse model of SCA was used to test the hypothesis that elastic lamina and structural damage in carotid arteries increased with age and was accelerated in mice homozygous for SCA (sickle cell anemia homozygous genotype [SS]) due to inflammatory signaling pathways activating proteolytic enzymes. Elastic lamina fragmentation observed by 1 month in SS mice compared with heterozygous littermate controls (sickle cell trait heterozygous genotype [AS]). Positive immunostaining for cathepsin K, a powerful collagenase and elastase, confirmed accelerated proteolytic activity in SS carotids. Larger cross-sectional areas were quantified by magnetic resonance angiography and increased arterial compliance in SS carotids were also measured. Inhibiting JNK (c-jun N-terminal kinase) signaling with SP600125 significantly reduced cathepsin K expression, elastin fragmentation, and carotid artery perimeters in SS mice. By 5 months of age, continued medial thinning and collagen degradation was mitigated by treatment of SS mice with JNK inhibitor. CONCLUSIONS: Arterial remodeling due to SCA is mediated by JNK signaling, cathepsin proteolytic upregulation, and degradation of elastin and collagen. Demonstration in Townes mice establishes their utility for mechanistic studies of arterial vasculopathy, related complications, and therapeutic interventions for large artery damage due to SCA.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anthracenes/pharmacology , Carotid Arteries/drug effects , Carotid Artery Diseases/prevention & control , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Vascular Remodeling/drug effects , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/genetics , Anemia, Sickle Cell/physiopathology , Animals , Carotid Arteries/enzymology , Carotid Arteries/physiopathology , Carotid Artery Diseases/enzymology , Carotid Artery Diseases/genetics , Carotid Artery Diseases/physiopathology , Cathepsin K/metabolism , Collagen/metabolism , Disease Models, Animal , Elastin/metabolism , Hemoglobins/genetics , Homozygote , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Transgenic , Mutation , Proteolysis , Signal Transduction , Time Factors
4.
Circulation ; 139(10): 1300-1319, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30586735

ABSTRACT

BACKGROUND: Platelet-neutrophil interactions contribute to vascular occlusion and tissue damage in thromboinflammatory disease. Platelet glycoprotein Ibα (GPIbα), a key receptor for the cell-cell interaction, is believed to be constitutively active for ligand binding. Here, we established the role of platelet-derived protein disulfide isomerase (PDI) in reducing the allosteric disulfide bonds in GPIbα and enhancing the ligand-binding activity under thromboinflammatory conditions. METHODS: Bioinformatic analysis identified 2 potential allosteric disulfide bonds in GPIbα. Agglutination assays, flow cytometry, surface plasmon resonance analysis, a protein-protein docking model, proximity ligation assays, and mass spectrometry were used to demonstrate a direct interaction between PDI and GPIbα and to determine a role for PDI in regulating GPIbα function and platelet-neutrophil interactions. Also, real-time microscopy and animal disease models were used to study the pathophysiological role of PDI-GPIbα signaling under thromboinflammatory conditions. RESULTS: Deletion or inhibition of platelet PDI significantly reduced GPIbα-mediated platelet agglutination. Studies using PDI-null platelets and recombinant PDI or Anfibatide, a clinical-stage GPIbα inhibitor, revealed that the oxidoreductase activity of platelet surface-bound PDI was required for the ligand-binding function of GPIbα. PDI directly bound to the extracellular domain of GPIbα on the platelet surface and reduced the Cys4-Cys17 and Cys209-Cys248 disulfide bonds. Real-time microscopy with platelet-specific PDI conditional knockout and sickle cell disease mice demonstrated that PDI-regulated GPIbα function was essential for platelet-neutrophil interactions and vascular occlusion under thromboinflammatory conditions. Studies using a mouse model of ischemia/reperfusion-induced stroke indicated that PDI-GPIbα signaling played a crucial role in tissue damage. CONCLUSIONS: Our results demonstrate that PDI-facilitated cleavage of the allosteric disulfide bonds tightly regulates GPIbα function, promoting platelet-neutrophil interactions, vascular occlusion, and tissue damage under thromboinflammatory conditions.


Subject(s)
Anemia, Sickle Cell/enzymology , Blood Platelets/enzymology , Inflammation/enzymology , Neutrophils/metabolism , Platelet Adhesiveness , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Disulfide-Isomerases/metabolism , Thrombosis/enzymology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Animals , Disease Models, Animal , Hemoglobins/genetics , Hemoglobins/metabolism , Humans , Inflammation/blood , Inflammation/genetics , Ligands , Mice, Inbred C57BL , Mice, Knockout , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Binding , Protein Disulfide-Isomerases/deficiency , Protein Disulfide-Isomerases/genetics , Signal Transduction , Thrombosis/blood , Thrombosis/genetics
5.
Blood ; 131(14): 1600-1610, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29437594

ABSTRACT

Patients with sickle cell disease (SCD) suffer from intravascular hemolysis associated with vascular injury and dysfunction in mouse models, and painful vaso-occlusive crisis (VOC) involving increased attachment of sickle erythrocytes and activated leukocytes to damaged vascular endothelium. Patrolling monocytes, which normally scavenge damaged cells and debris from the vasculature, express higher levels of anti-inflammatory heme oxygenase 1 (HO-1), a heme degrading enzyme. Here, we show that HO-1-expressing patrolling monocytes protect SCD vasculature from ongoing hemolytic insult and vaso-occlusion. We found that a mean 37% of patrolling monocytes from SCD patients express very high levels of HO-1 (HO-1hi) vs 6% in healthy controls and demonstrated that HO-1hi expression was dependent on uptake of heme-exposed endothelium. SCD patients with a recent VOC episode had lower numbers of HO-1hi patrolling monocytes. Heme-mediated vaso-occlusion by mouse SCD red blood cells was exacerbated in mice lacking patrolling monocytes, and reversed following transfer of patrolling monocytes. Altogether, these data indicate that SCD patrolling monocytes remove hemolysis-damaged endothelial cells, resulting in HO-1 upregulation and dampening of VOC, and that perturbation in patrolling monocyte numbers resulting in lower numbers of HO-1hi patrolling monocyte may predispose SCD patients to VOC. These data suggest that HO-1hi patrolling monocytes are key players in VOC pathophysiology and have potential as therapeutic targets for VOC.


Subject(s)
Anemia, Sickle Cell/enzymology , Heme Oxygenase-1/metabolism , Hemolysis , Monocytes/enzymology , Vascular Diseases/prevention & control , Adolescent , Adult , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/pathology , Child , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Humans , Male , Middle Aged , Monocytes/pathology , Vascular Diseases/enzymology , Vascular Diseases/genetics , Vascular Diseases/pathology
6.
Biochem Genet ; 58(4): 580-594, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32277314

ABSTRACT

The impaired bioavailability of endogenous nitric oxide (NO) in sickle cell anemia (SCA) may be influenced by polymorphisms in the endothelial nitric oxide synthase gene (eNOS). We compared allelic/genotypic frequencies of the eNOS polymorphisms T-786C, VNTR4a/b and G894T between 89 adult SCA patients and 100 healthy controls, and investigated the relationship between these SNPs and markers of hemolysis [lactate dehydrogenase (LDH), indirect bilirubin (IB) and reticulocyte counts], inflammation [interleukins IL-1ß, IL-6, IL-8, Tumor Necrosis Factor (TNF-α) and C-reactive protein (CRP)] and endothelial dysfunction (ED) [soluble vascular cell adhesion molecule-1 (sVCAM-1), soluble intercellular adhesion molecule-1 (sICAM-1), soluble L-selectin (sL-selectin), von Willebrand Factor (vWF) antigen and D-dimers] in the patients. The frequencies of the mutant -786C allele and -786C/C genotype were significantly higher in patients (p = 0.02 and p = 0.04, respectively) but not significantly correlated with the markers. For VNTR4a/b and G894T, the allelic/genotypic frequencies did not statistically differ between patient and control groups. Patients carrying the 4a allele and those with the 894G/G genotype showed a significant decrease in IB (p = 0.02 and p = 0.04, respectively), and only patients with the 4a allele exhibited reduced IL-1ß (p = 0.01). The correlation profiles between markers of inflammation and ED significantly differed between patients carrying the mutant alleles and those with wild-type genotypes. This appears to be the first report on the relationship between eNOS gene polymorphisms and markers of hemolysis, inflammation and ED in Brazilian SCA patients. Our results indicate that the SNPs analyzed may influence the phenotypic variability of these patients.


Subject(s)
Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/genetics , Fibrin Fibrinogen Degradation Products/analysis , Hemolysis , Intercellular Adhesion Molecule-1/blood , Nitric Oxide Synthase Type III/genetics , Polymorphism, Single Nucleotide , Vascular Cell Adhesion Molecule-1/blood , von Willebrand Factor/analysis , Adult , Alleles , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/epidemiology , Bilirubin/blood , Biomarkers/blood , Brazil/epidemiology , Case-Control Studies , Cytokines/blood , Female , Gene Frequency , Haplotypes , Humans , Inflammation/blood , L-Lactate Dehydrogenase/blood , Male , Reticulocyte Count , Young Adult
7.
Br J Haematol ; 187(3): 386-395, 2019 11.
Article in English | MEDLINE | ID: mdl-31273765

ABSTRACT

Hereditary spherocytosis (HS) is characterised by increased osmotic fragility and enhanced membrane loss of red blood cells (RBC) due to defective membrane protein complexes. In our diagnostic laboratory, we observed that pyruvate kinase (PK) activity in HS was merely slightly elevated with respect to the amount of reticulocytosis. In order to evaluate whether impaired PK activity is a feature of HS, we retrospectively analysed laboratory data sets from 172 unrelated patients with HS, hereditary elliptocytosis (HE), glucose-6-phosphate dehydrogenase (G6PD) or PK deficiency, sickle cell or haemoglobin C disease, or ß-thalassaemia minor. Results from linear regression analysis provided proof that PK activity decreases with rising reticulocyte counts in HS (R2  = 0·15; slope = 9·09) and, less significantly, in HE (R2  = 0·021; slope = 8·92) when compared with other haemolytic disorders (R2  ≥ 0·65; slopes ≥ 78·6). Reticulocyte-adjusted erythrocyte PK activity levels were significantly lower in HS and even declined with increasing reticulocytes (R2  = 0·48; slope = -9·74). In this report, we describe a novel association between HS and decreased PK activity that is apparently caused by loss of membrane-bound PK due to impaired structural integrity of the RBC membrane and may aggravate severity of haemolysis in HS.


Subject(s)
Erythrocyte Membrane/enzymology , Erythrocytes, Abnormal/enzymology , Pyruvate Kinase/metabolism , Spherocytosis, Hereditary/enzymology , Adolescent , Adult , Aged , Anemia, Hemolytic, Congenital Nonspherocytic/enzymology , Anemia, Hemolytic, Congenital Nonspherocytic/pathology , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/pathology , Child , Child, Preschool , Erythrocyte Membrane/pathology , Erythrocytes, Abnormal/pathology , Female , Hemoglobin C Disease/enzymology , Hemoglobin C Disease/pathology , Humans , Infant , Male , Middle Aged , Pyruvate Kinase/deficiency , Pyruvate Metabolism, Inborn Errors/enzymology , Pyruvate Metabolism, Inborn Errors/pathology , Reticulocytes/enzymology , Reticulocytes/pathology , Spherocytosis, Hereditary/pathology , beta-Thalassemia/enzymology , beta-Thalassemia/pathology
8.
Ann Hematol ; 98(7): 1561-1572, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31098737

ABSTRACT

Sickle cell disease (SCD), a hereditary form of chronic hemolytic anemia, is characterized by acute vascular occlusion and chronic complications as pulmonary hypertension (PH), a hallmark of higher mortality. This study aimed to determine peripheral blood expression of superoxide dismutase 2 (SOD2), a major mitochondrial antioxidant enzyme in SCD patients on the mRNA level and compared it with SOD2 expression in healthy individuals. It also aimed to detect possible differences in SOD2 expression among patients with/without specific SCD complications and to detect possible correlations with patient laboratory parameters. SOD2 mRNA levels were significantly lower in SCD patients in comparison with controls and correlated with red blood cell count, reticulocyte count, platelet count, C-reactive protein, ferritin, and brain natriuretic peptide values. SCD patients with echocardiographic indications of PH featured significantly reduced SOD2 expression in comparison with patients without such indications. Consequently, SOD2 expression emerges as a potential biomarker of PH in SCD being a link among hemolysis, inflammation, iron overload, oxidative stress, and SCD cardiopathy.


Subject(s)
Anemia, Sickle Cell/enzymology , Gene Expression Regulation, Enzymologic , Superoxide Dismutase/blood , Adult , Anemia, Sickle Cell/pathology , Biomarkers/blood , C-Reactive Protein/metabolism , Female , Ferritins/blood , Humans , Male , Natriuretic Peptide, Brain/blood , Platelet Count , Reticulocyte Count
9.
Curr Opin Hematol ; 25(6): 494-499, 2018 11.
Article in English | MEDLINE | ID: mdl-30239377

ABSTRACT

PURPOSE OF REVIEW: Glucose-6-phosphate dehydrogenase (G6PD) deficiency and sickle cell disease (SCD) cause hemolysis, often occurring in individuals of African descent. These disorders co-occur frequently, and possibly interact, altering clinical outcomes in SCD. However, epidemiological investigations of SCD with G6PD deficiency have produced variable results. This contribution reviews the available data about the interaction of G6PD deficiency and SCD. RECENT FINDINGS: Overall, G6PD deficiency contributes few, if any, effects to laboratory values and clinical outcomes in SCD patients, but may impact transfusion efficacy. This observation is most likely because of the relatively increased G6PD activity in the young red blood cell (RBC) population seen in SCD patients with or without G6PD deficiency. In addition, G6PD deficiency possibly interacts with other genetic modifiers, such as α thalassemia, hemoglobin F levels and SCD haplotype. SUMMARY: Although G6PD deficiency is relatively common, it does not appear to clinically impact patients with SCD. Nonetheless, it is important to evaluate G6PD status in patients with SCD to avoid the use of medications that may cause hemolysis. Future studies evaluating the clinical impact of transfusions from G6PD-deficient RBC donors would be of the greatest benefit to the current literature.


Subject(s)
Anemia, Sickle Cell/metabolism , Glucosephosphate Dehydrogenase Deficiency/metabolism , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/epidemiology , Erythrocytes/metabolism , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase Deficiency/genetics , Humans
10.
Br J Haematol ; 181(1): 111-121, 2018 04.
Article in English | MEDLINE | ID: mdl-29527679

ABSTRACT

Recent studies have demonstrated pleiotropic effects of statins in various mouse models of kidney disease. In this study, Townes humanized sickle cell mice were treated for 8 weeks with atorvastatin at a dose of 10 mg/kg/day starting at 10 weeks of age. Treatment with atorvastatin significantly reduced albuminuria, and improved both urine concentrating ability and glomerular filtration rate. Atorvastatin also decreased markers of kidney injury and endothelial activation, and ameliorated oxidant stress in renal tissues and peripheral macrophages. Atorvastatin downregulated the expression of mRNA levels of the NADPH oxidases, Cybb (also termed Nox2) and Nox4, which are major sources of oxidant stress in the kidney. These findings highlight the pleiotropic effects of atorvastatin and suggest that it may provide beneficial effects in sickle cell nephropathy.


Subject(s)
Anemia, Sickle Cell/drug therapy , Atorvastatin/pharmacology , Kidney Diseases/prevention & control , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/enzymology , Anemia, Sickle Cell/pathology , Animals , Disease Models, Animal , Female , Kidney/enzymology , Kidney/pathology , Kidney Diseases/enzymology , Kidney Diseases/etiology , Kidney Diseases/pathology , Male , Mice , Mice, Mutant Strains , NADPH Oxidase 2/metabolism , NADPH Oxidase 4/metabolism
11.
J Cell Sci ; 129(18): 3511-7, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27505892

ABSTRACT

Sickle cell disease is a destructive genetic disorder characterized by the formation of fibrils of deoxygenated hemoglobin, leading to the red blood cell (RBC) morphology changes that underlie the clinical manifestations of this disease. Using cryogenic soft X-ray tomography (SXT), we characterized the morphology of sickled RBCs in terms of volume and the number of protrusions per cell. We were able to identify statistically a relationship between the number of protrusions and the volume of the cell, which is known to correlate to the severity of sickling. This structural polymorphism allows for the classification of the stages of the sickling process. Recent studies have shown that elevated sphingosine kinase 1 (Sphk1)-mediated sphingosine 1-phosphate production contributes to sickling. Here, we further demonstrate that compound 5C, an inhibitor of Sphk1, has anti-sickling properties. Additionally, the variation in cellular morphology upon treatment suggests that this drug acts to delay the sickling process. SXT is an effective tool that can be used to identify the morphology of the sickling process and assess the effectiveness of potential therapeutics.


Subject(s)
Anemia, Sickle Cell/enzymology , Erythrocytes/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Tomography, X-Ray/methods , Animals , Cell Surface Extensions/drug effects , Cell Surface Extensions/metabolism , Erythrocytes/drug effects , Humans , Mice , Protein Kinase Inhibitors/pharmacology
12.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R840-R847, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30067082

ABSTRACT

Sickle cell disease (SCD) is caused by a mutation of the ß-globin gene (Ingram VM. Nature 180: 326-328, 1957), which triggers the polymerization of deoxygenated sickle hemoglobin (HbS). Approximately 100,000 SCD patients in the United States and millions worldwide (Piel FB, et al. PLoS Med 10: e1001484, 2013) suffer from chronic hemolytic anemia, painful crises, multisystem organ damage, and reduced life expectancy (Rees DC, et al. Lancet 376: 2018-2031, 2010; Serjeant GR. Cold Spring Harb Perspect Med 3: a011783, 2013). Hematopoietic stem cell transplantation can be curative, but the majority of patients do not have a suitable donor (Talano JA, Cairo MS. Eur J Haematol 94: 391-399, 2015). Advanced gene-editing technologies also offer the possibility of a cure (Goodman MA, Malik P. Ther Adv Hematol 7: 302-315, 2016; Lettre G, Bauer DE. Lancet 387: 2554-2564, 2016), but the likelihood that these strategies can be mobilized to treat the large numbers of patients residing in developing countries is remote. A pharmacological treatment to increase fetal hemoglobin (HbF) as a therapy for SCD has been a long-sought goal, because increased levels of HbF (α2γ2) inhibit the polymerization of HbS (Poillin WN, et al. Proc Natl Acad Sci USA 90: 5039-5043, 1993; Sunshine HR, et al. J Mol Biol 133: 435-467, 1979) and are associated with reduced symptoms and increased lifespan of SCD patients (Platt OS, et al. N Engl J Med 330: 1639-1644, 1994; Platt OS, et al. N Engl J Med 325: 11-16, 1991). Only two drugs, hydroxyurea and l-glutamine, are approved by the US Food and Drug Administration for treatment of SCD. Hydroxyurea is ineffective at HbF induction in ~50% of patients (Charache S, et al. N Engl J Med 332: 1317-1322, 1995). While polymerization of HbS has been traditionally considered the driving force in the hemolysis of SCD, the excessive reactive oxygen species generated from red blood cells, with further amplification by intravascular hemolysis, also are a major contributor to SCD pathology. This review highlights a new class of drugs, lysine-specific demethylase (LSD1) inhibitors, that induce HbF and reduce reactive oxygen species.


Subject(s)
Anemia, Sickle Cell/drug therapy , Enzyme Inhibitors/pharmacology , Erythrocytes/drug effects , Histone Demethylases/antagonists & inhibitors , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Animals , Biomarkers/blood , Disease Models, Animal , Drug Design , Erythrocytes/enzymology , Fetal Hemoglobin/metabolism , Histone Demethylases/metabolism , Humans , Mice , Molecular Targeted Therapy , Papio , Reactive Oxygen Species/blood , Risk Assessment
14.
Blood ; 125(10): 1643-52, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25587035

ABSTRACT

Erythrocyte possesses high sphingosine kinase 1 (SphK1) activity and is the major cell type supplying plasma sphingosine-1-phosphate, a signaling lipid regulating multiple physiological and pathological functions. Recent studies revealed that erythrocyte SphK1 activity is upregulated in sickle cell disease (SCD) and contributes to sickling and disease progression. However, how erythrocyte SphK1 activity is regulated remains unknown. Here we report that adenosine induces SphK1 activity in human and mouse sickle and normal erythrocytes in vitro. Next, using 4 adenosine receptor-deficient mice and pharmacological approaches, we determined that the A2B adenosine receptor (ADORA2B) is essential for adenosine-induced SphK1 activity in human and mouse normal and sickle erythrocytes in vitro. Subsequently, we provide in vivo genetic evidence that adenosine deaminase (ADA) deficiency leads to excess plasma adenosine and elevated erythrocyte SphK1 activity. Lowering adenosine by ADA enzyme therapy or genetic deletion of ADORA2B significantly reduced excess adenosine-induced erythrocyte SphK1 activity in ADA-deficient mice. Finally, we revealed that protein kinase A-mediated extracellular signal-regulated kinase 1/2 activation functioning downstream of ADORA2B underlies adenosine-induced erythrocyte SphK1 activity. Overall, our findings reveal a novel signaling network regulating erythrocyte SphK1 and highlight innovative mechanisms regulating SphK1 activity in normal and SCD.


Subject(s)
Adenosine/blood , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Erythrocytes, Abnormal/metabolism , Phosphotransferases (Alcohol Group Acceptor)/blood , Receptor, Adenosine A2B/blood , Adenosine Deaminase/blood , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Adenosine-5'-(N-ethylcarboxamide)/pharmacology , Agammaglobulinemia/blood , Agammaglobulinemia/enzymology , Agammaglobulinemia/genetics , Anemia, Sickle Cell/genetics , Animals , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/blood , Erythrocytes/drug effects , Erythrocytes/enzymology , Erythrocytes/metabolism , Erythrocytes, Abnormal/drug effects , Erythrocytes, Abnormal/enzymology , Hemoglobin, Sickle/genetics , Hemoglobin, Sickle/metabolism , Humans , MAP Kinase Signaling System , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Receptor, Adenosine A2B/deficiency , Receptor, Adenosine A2B/genetics , Severe Combined Immunodeficiency/blood , Severe Combined Immunodeficiency/enzymology , Severe Combined Immunodeficiency/genetics , Signal Transduction
15.
FASEB J ; 30(3): 1171-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26631480

ABSTRACT

In sickle cell disease (SCD), treatment of recurrent vasoocclusive episodes, leading to pain crises and organ damage, is still a therapeutic challenge. Vasoocclusion is caused primarily by adherence of homozygous for hemoglobin S (SS) red blood cells (SSRBCs) and leukocytes to the endothelium. We tested the therapeutic benefits of MEK1/2 inhibitors in reversing vasoocclusion in nude and humanized SCD mouse models of acute vasoocclusive episodes using intravital microscopy. Administration of 0.2, 0.3, 1, or 2 mg/kg MEK1/2 inhibitor to TNF-α-pretreated nude mice before human SSRBC infusion inhibited SSRBC adhesion in inflamed vessels, prevented the progression of vasoocclusion, and reduced SSRBC organ sequestration. By use of a more clinically relevant protocol, 0.3 or 1 mg/kg MEK1/2 inhibitor given to TNF-α-pretreated nude mice after human SSRBC infusion and onset of vasoocclusion reversed SSRBC adhesion and vasoocclusion and restored blood flow. In SCD mice, 0.025, 0.05, or 0.1 mg/kg MEK1/2 inhibitor also reversed leukocyte and erythrocyte adhesion after the inflammatory trigger of vasoocclusion and improved microcirculatory blood flow. Cell adhesion was reversed by shedding of endothelial E-selectin, P-selectin, and αvß3 integrin, and leukocyte CD44 and ß2 integrin. Thus, MEK1/2 inhibitors, by targeting the adhesive function of SSRBCs and leukocytes, could represent a valuable therapeutic intervention for acute sickle cell vasoocclusive crises.


Subject(s)
Anemia, Sickle Cell/drug therapy , Anemia, Sickle Cell/enzymology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/metabolism , Animals , Cell Adhesion/drug effects , Cell Line , Disease Models, Animal , E-Selectin/metabolism , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Hemoglobin, Sickle/metabolism , Humans , Hyaluronan Receptors/metabolism , Integrin alphaVbeta3/metabolism , Leukocytes/drug effects , Leukocytes/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Male , Mice , Mice, Nude , Mice, Transgenic , P-Selectin/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Diseases/drug therapy , Vascular Diseases/metabolism
16.
Niger J Clin Pract ; 20(11): 1428-1433, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29303127

ABSTRACT

BACKGROUND AND OBJECTIVE: Multiorgan failure including liver dysfunction is a common finding in sickle cell anemia (SCA) patients, the cause of which is multifactorial with advancing age said to be a major determinant. There is a paucity of data on liver function among SCA patients in relation to age in northern Nigerian hospitals, including Ahmadu Bello University Teaching Hospital (ABUTH), Zaria. This study was to assess the biochemical liver function tests (LFTs) as they relate to age among SCA patients in steady state, with a view to improving the overall monitoring of these patients. SUBJECTS AND METHODS: This study was carried out in ABUTH, Zaria, Northern Nigeria. LFTs were carried out in 100 SCA and 100 apparently healthy participants (controls). The SCA group was made up of fifty adults and fifty children diagnosed of SCA, whereas the control group was made up of fifty adults and fifty children who were apparently healthy and had hemoglobin AA. Paired two-tailed Student's t-test for matched samples and Pearson's linear correlation statistical methods were employed for the data analysis using Microsoft Office Excel 2007. A P ≤ 0.05 was considered as statistically significant. RESULTS: The serum concentrations of total bilirubin (TB), alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), and AST/ALT ratio were significantly higher in SCA patients compared to the controls (P = 0.001, P = 0.001, P = 0.05, P = 0.05 and P = 0.001, respectively). Serum total protein (TP) and ALB were significantly lower (P = 0.01 and P < 0.05, respectively) in SCA patients compared with the controls. The levels of TB, ALT, AST, ALP, and AST/ALT were significantly lower in SCA adults compared to SCA children, whereas TP and ALB were higher in SCA adults compared to the SCA children. There were significant negative correlations between age and each of TB, ALT, AST, ALP, and AST/ALT, and significant positive correlations between age and each of TP and ALB in SCA patients. CONCLUSION: There are mild LFTs derangements in SCA patients even in steady state with the extent of the abnormalities decreasing with advancing age of the patients.


Subject(s)
Alanine Transaminase/blood , Alkaline Phosphatase/blood , Anemia, Sickle Cell/enzymology , Aspartate Aminotransferases/blood , Liver Diseases/etiology , Liver/enzymology , Adolescent , Adult , Age Factors , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/complications , Bilirubin/blood , Case-Control Studies , Child , Female , Hemoglobin A , Hospitals, Teaching , Humans , Liver/pathology , Liver Diseases/blood , Liver Function Tests/methods , Male , Middle Aged , Nigeria
17.
J Biol Chem ; 290(46): 27939-58, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26396189

ABSTRACT

Polymerization of intraerythrocytic deoxyhemoglobin S (HbS) is the primary molecular event that leads to hemolytic anemia in sickle cell disease (SCD). We reasoned that HbS may contribute to the complex pathophysiology of SCD in part due to its pseudoperoxidase activity. We compared oxidation reactions and the turnover of oxidation intermediates of purified human HbS and HbA. Hydrogen peroxide (H2O2) drives a catalytic cycle that includes the following three distinct steps: 1) initial oxidation of ferrous (oxy) to ferryl Hb; 2) autoreduction of the ferryl intermediate to ferric (metHb); and 3) reaction of metHb with an additional H2O2 molecule to regenerate the ferryl intermediate. Ferrous and ferric forms of both proteins underwent initial oxidation to the ferryl heme in the presence of H2O2 at equal rates. However, the rate of autoreduction of ferryl to the ferric form was slower in the HbS solutions. Using quantitative mass spectrometry and the spin trap, 5,5-dimethyl-1-pyrroline-N-oxide, we found more irreversibly oxidized ßCys-93in HbS than in HbA. Incubation of the ferric or ferryl HbS with cultured lung epithelial cells (E10) induced a drop in mitochondrial oxygen consumption rate and impairment of cellular bioenergetics that was related to the redox state of the iron. Ferryl HbS induced a substantial drop in the mitochondrial transmembrane potential and increases in cytosolic heme oxygenase (HO-1) expression and mitochondrial colocalization in E10 cells. Thus, highly oxidizing ferryl Hb and heme, the product of oxidation, may be central to the evolution of vasculopathy in SCD and may suggest therapeutic modalities that interrupt heme-mediated inflammation.


Subject(s)
Cysteine/chemistry , Hemoglobin, Sickle/chemistry , Iron/chemistry , Mitochondria/metabolism , Respiratory Mucosa/enzymology , Anemia, Hemolytic/enzymology , Anemia, Sickle Cell/enzymology , Catalysis , Cyclic N-Oxides/chemistry , Energy Metabolism , Heme/chemistry , Heme Oxygenase (Decyclizing)/chemistry , Humans , Hydrogen Peroxide/chemistry , Lung/enzymology , Methemoglobin/chemistry , Oxidation-Reduction , Oxygen Consumption , Respiratory Mucosa/ultrastructure
18.
Blood ; 124(12): 1941-50, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25075126

ABSTRACT

Sphingolipids are a class of lipids containing a backbone of sphingoid bases that can be produced de novo through the reaction of palmitate and serine and further metabolized through the activity of various enzymes to produce intermediates with diverse roles in cellular processes and signal transduction. One of these intermediates, sphingosine 1-phosphate (S1P), is stored at high concentrations (1 µM) in red blood cells (RBCs) and directs a wide array of cellular processes mediated by 5 known G-protein coupled receptors (S1P1-S1P5). In this study, we show that RBC membrane alterations in sickle cell disease enhance the activation acid sphingomyelinase by 13%, resulting in increased production and storage of sphingosine (2.6-fold) and S1P (3.5-fold). We also show that acid sphingomyelinase enhances RBC-derived microparticle (MP) generation. These MPs are internalized by myeloid cells and promote proinflammatory cytokine secretion and endothelial cell adhesion, suggesting that potential crosstalk between circulating inflammatory cells and MPs may contribute to the inflammation-rooted pathogenesis of the disease. Treatment with amitriptyline reduces MP generation in vitro and in vivo and might be used to mitigate inflammatory processes in sickle cell disease.


Subject(s)
Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Cell-Derived Microparticles/enzymology , Erythrocytes, Abnormal/enzymology , Sphingomyelin Phosphodiesterase/blood , Anemia, Sickle Cell/etiology , Animals , Cell Adhesion , Cell Adhesion Molecules/blood , Disease Models, Animal , Endothelial Cells/pathology , Hemoglobin, Sickle/genetics , Humans , Inflammation/blood , Inflammation/enzymology , Lysophospholipids/blood , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Sphingolipids/blood , Sphingosine/analogs & derivatives , Sphingosine/blood
19.
Microvasc Res ; 106: 128-36, 2016 07.
Article in English | MEDLINE | ID: mdl-27089822

ABSTRACT

The sickle cell disease (SCD) is a hemolytic genetic anemia characterized by free heme and hemoglobin release into intravascular spaces, with endothelial activation. Heme is a proinflammatory molecule able to directly activate vascular endothelium, thus, endothelial dysfunction and vascular disease are major chronic events described in SCD. The aim of this study was to evaluate the production of endothelial nitric oxide synthase (eNOS), nitrite and hypoxia inducible factor alpha (HIF-α) in HUVECs (human umbilical vein endothelial cells) activated by heme in response to simvastatin, hydroxyurea (HU), and ascorbic acid therapies. eNOS and HIF-α production were evaluated by ELISA and nitrite was measured by the Griess technique. The production of HIF-α increased when the cells were stimulated by heme (p<0.01), while treatment with HU and simvastatin reduced the production (p<0.01), and treatment with ascorbic acid increased HIF-1a production by the cells (p<0.01). Heme increased eNOS production, (p<0.01) but showed a heterogeneous pattern, and the lowest concentrations of all the treatments reduced the enzyme production (p<0.01). The nitrite production by HUVECs was enhanced by stimulation with heme (p<0.001) and was reduced by treatment with HU (p<0.001), ascorbic acid (p<0.001) and simvastatin (p<0.01). In summary, our results suggest that the hemolytic vascular microenvironment in SCD requires different therapeutic approaches to promote clinical improvement, and that a combination of therapies may be a viable strategy for treating patients.


Subject(s)
Anemia, Sickle Cell/drug therapy , Ascorbic Acid/pharmacology , Heme/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Hydroxyurea/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitrites/metabolism , Simvastatin/pharmacology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/enzymology , Cells, Cultured , Dose-Response Relationship, Drug , Human Umbilical Vein Endothelial Cells/enzymology , Humans
20.
Pediatr Blood Cancer ; 63(6): 1046-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26840990

ABSTRACT

BACKGROUND: Stroke is a severe complication of sickle cell anemia (SCA). The role of glucose-6-phosphate dehydrogenase (G6PD) deficiency in the development of stroke in children with SCA is controversial. PROCEDURE: The aim of this study was to investigate the association of clinical ischemic stroke, high-risk transcranial Doppler measurements (TCD), and hematological features with molecular variants usually linked to G6PD deficiency or with the biochemical activity of G6PD in a cohort of 395 Brazilian children with SCA. G6PD activity was quantitatively determined using an enzymatic-colorimetric assay. G6PD mutations were determined by PCR-RFLP and sequencing. Clinical and hematological data were retrieved from the children's records. RESULTS: The prevalence of molecularly defined deficiency (hereafter, molecular deficiency) was 4.3% (95% confidence interval: 2.3-6.3%). The mean G6PD activity was 16.88 U/g hemoglobin (Hb) (standard error of the mean [SEM] 0.28) in the group without G6PD molecular deficiency and 8.43 (SEM 1.01) U/g Hb in the group with G6PD A(-) molecular deficiency. G6PD molecular deficiency was not associated with any hematological features. No effects of G6PD molecular deficiency on clinical ischemic stroke or high-risk TCD were detected. The mean G6PD activity was similar in children who had clinical ischemic stroke and in those without stroke. Similar results were obtained in analyses comparing children who had high-risk TCD and those without high-risk TCD. CONCLUSIONS: Our study demonstrated that G6PD molecular deficiency was not associated either with clinical ischemic stroke or high-risk TCD. Similarly, we found no associations between G6PD enzyme activity and stroke or high-risk TCD. Small sample size precludes definitive conclusions.


Subject(s)
Anemia, Sickle Cell/complications , Glucosephosphate Dehydrogenase Deficiency/complications , Stroke/epidemiology , Anemia, Sickle Cell/enzymology , Brazil/epidemiology , Child , Cohort Studies , Female , Glucosephosphate Dehydrogenase/genetics , Glucosephosphate Dehydrogenase Deficiency/epidemiology , Glucosephosphate Dehydrogenase Deficiency/genetics , Humans , Male , Polymorphism, Restriction Fragment Length , Prevalence , Retrospective Studies , Stroke/diagnostic imaging , Stroke/etiology , Ultrasonography, Doppler, Transcranial
SELECTION OF CITATIONS
SEARCH DETAIL