Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Proc Natl Acad Sci U S A ; 120(21): e2209639120, 2023 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-37186844

RESUMO

Renal medullary carcinoma (RMC) is an aggressive kidney cancer that almost exclusively develops in individuals with sickle cell trait (SCT) and is always characterized by loss of the tumor suppressor SMARCB1. Because renal ischemia induced by red blood cell sickling exacerbates chronic renal medullary hypoxia in vivo, we investigated whether the loss of SMARCB1 confers a survival advantage under the setting of SCT. Hypoxic stress, which naturally occurs within the renal medulla, is elevated under the setting of SCT. Our findings showed that hypoxia-induced SMARCB1 degradation protected renal cells from hypoxic stress. SMARCB1 wild-type renal tumors exhibited lower levels of SMARCB1 and more aggressive growth in mice harboring the SCT mutation in human hemoglobin A (HbA) than in control mice harboring wild-type human HbA. Consistent with established clinical observations, SMARCB1-null renal tumors were refractory to hypoxia-inducing therapeutic inhibition of angiogenesis. Further, reconstitution of SMARCB1 restored renal tumor sensitivity to hypoxic stress in vitro and in vivo. Together, our results demonstrate a physiological role for SMARCB1 degradation in response to hypoxic stress, connect the renal medullary hypoxia induced by SCT with an increased risk of SMARCB1-negative RMC, and shed light into the mechanisms mediating the resistance of SMARCB1-null renal tumors against angiogenesis inhibition therapies.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Traço Falciforme , Animais , Humanos , Camundongos , Carcinoma de Células Renais/patologia , Hipóxia/genética , Hipóxia/metabolismo , Rim/metabolismo , Neoplasias Renais/patologia , Traço Falciforme/genética , Traço Falciforme/metabolismo , Proteína SMARCB1/genética , Proteína SMARCB1/metabolismo
2.
Redox Biol ; 58: 102536, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36401887

RESUMO

The redox state of the host-parasite unit has been hypothesized to play a central role for the fitness of the intraerythrocytic blood stages of the human malaria parasite Plasmodium falciparum. In particular, hemoglobinopathies have been suggested to cause a more oxidizing environment, thereby protecting from severe malaria. Here we determined the redox potential of infected wild-type (hemoglobin AA) or sickle trait (hemoglobin AS) erythrocytes using parasite-encoded variants of the redox-sensitive green-fluorescent protein 2 (roGFP2). Our non-invasive roGFP2 single-cell measurements revealed a reducing steady-state redox potential of -304 ± 11 mV for the erythrocyte cytosol during ring-stage development and a rather sudden oxidation to -278 ± 12 mV during trophozoite-stage development around 28 h post invasion. There was no significant difference between wild-type or sickle trait erythrocytes regarding the stage dependence and the detected increase of the redox potential during the intraerythrocytic life cycle. The steady-state redox potential of the parasite cytosol, between -304 and -313 mV, was highly reducing throughout the life cycle. The redox potential in the parasitophorous vacuole at the interface between the secretory pathway and the erythrocyte was -284 ± 10 mV and remained stable during trophozoite-stage development with implications for the export of disulfide-containing proteins. In summary, P. falciparum blood stage development from the late ring to the early trophozoite stage causes a physiological jump in erythrocyte redox potential irrespective of the presence or absence of hemoglobin S.


Assuntos
Malária Falciparum , Traço Falciforme , Humanos , Plasmodium falciparum/metabolismo , Traço Falciforme/genética , Traço Falciforme/metabolismo , Eritrócitos/metabolismo , Malária Falciparum/parasitologia , Proteínas de Fluorescência Verde/metabolismo , Oxirredução
3.
PLoS Pathog ; 17(6): e1009659, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34115805

RESUMO

Sickle-trait hemoglobin protects against severe Plasmodium falciparum malaria. Severe malaria is governed in part by the expression of the Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) that are encoded by var genes, specifically those variants that bind Endothelial Protein C Receptor (EPCR). In this study, we investigate the effect of sickle-trait on parasite var gene expression and function in vitro and in field-collected parasites. We mapped var gene reads generated from RNA sequencing in parasite cultures in normal and sickle-cell trait blood throughout the asexual lifecycle. We investigated sickle-trait effect on PfEMP1 interactions with host receptors CD36 and EPCR using static adhesion assays and flow cytometry. Var expression in vivo was compared by assembling var domains sequenced from total RNA in parasites infecting Malian children with HbAA and HbAS. Sickle-trait did not alter the abundance or type of var gene transcripts in vitro, nor the abundance of overall transcripts or of var functional domains in vivo. In adhesion assays using recombinant host receptors, sickle-trait reduced adhesion by 73-86% to CD36 and 83% to EPCR. Similarly, sickle-trait reduced the surface expression of EPCR-binding PfEMP1. In conclusion, Sickle-cell trait does not directly affect var gene transcription but does reduce the surface expression and function of PfEMP1. This provides a direct mechanism for protection against severe malaria conferred by sickle-trait hemoglobin. Trial Registration: ClinicalTrials.gov Identifier: NCT02645604.


Assuntos
Hemoglobina Falciforme/metabolismo , Malária Falciparum/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Antígenos CD36/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Eritrócitos/parasitologia , Hemoglobina Falciforme/genética , Humanos , Malária Falciparum/metabolismo , Traço Falciforme/genética , Traço Falciforme/metabolismo
4.
PLoS One ; 14(5): e0216467, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31083675

RESUMO

Abnormal red blood cell (RBC) adhesion to endothelial αvß3 plays a crucial role in triggering vaso-occlusive episodes in sickle cell disease (SCD). It is known that epinephrine, a ß-adrenergic receptor (ß-AR) stimulator, increases the RBC surface density of active intercellular adhesion molecule-4 (ICAM-4) which binds to the endothelial αvß3. It has also been demonstrated that in human embryonic kidney 293 cells, mouse cardiomyocytes, and COS-7 cell lines, the ß-adrenergic and renin-angiotensin systems are interrelated and that there is a direct interaction and cross-regulation between ß-AR and angiotensin II type 1 receptor (AT1R). Selective blockade of AT1R reciprocally inhibits the downstream signaling of ß-ARs, similar to the inhibition observed in the presence of a ß-AR-blocker. However, it is not known if this mechanism is active in human RBCs. Here, we studied the effect of valsartan, an AT1R blocker, on the surface density of active ICAM-4 receptors in normal, sickle cell trait, and homozygous sickle RBCs. We applied single molecule force spectroscopy to detect active ICAM-4 receptors on the RBC plasma membrane with and without the presence of valsartan and epinephrine. We found that epinephrine significantly increased whereas valsartan decreased their surface density. Importantly, we found that pretreatment of RBCs with valsartan significantly impeded the activation of ICAM-4 receptors induced by epinephrine. The observed reduced expression of active ICAM-4 receptors on the RBC plasma membrane leads us to conjecture that valsartan may be used as a supporting remedy for the prevention and treatment of vaso-occlusive crisis in SCD.


Assuntos
Moléculas de Adesão Celular/metabolismo , Epinefrina/farmacologia , Membrana Eritrocítica/metabolismo , Eritrócitos Anormais/metabolismo , Traço Falciforme/metabolismo , Valsartana/farmacologia , Adolescente , Adulto , Animais , Células COS , Chlorocebus aethiops , Membrana Eritrocítica/ultraestrutura , Eritrócitos Anormais/ultraestrutura , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Microscopia de Força Atômica , Receptor Tipo 1 de Angiotensina/metabolismo , Traço Falciforme/patologia
5.
Mol Biochem Parasitol ; 227: 5-14, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472238

RESUMO

Deficiency of glucose-6-phosphate dehydrogenase (G6PD) and sickle cell trait (SCT) are described as the polymorphic disorders prevalent in erythrocytes. Both are considered the result of the selective pressure exerted by Plasmodium parasites over human genome, due to a certain degree of resistance to the clinical symptoms of severe malaria. There exist in both a prooxidant environment that favors the oxidative damage on membrane proteins, which probably is part of molecular protector mechanisms. Nevertheless, mechanisms are not completely understood at molecular level for each polymorphism yet, and even less if are commons for several of them. Here, synchronous cultures at high parasitemia levels of P. falciparum 3D7 were used to quantify oxidative damage in membrane proteins of erythrocytes with G6PD deficient and SCT. Carbonyl index by dot blot assay was used to calculate the variation of oxidative damage during the asexual phases. Besides, protein carbonylation profiles were obtained by Western blot and complemented with mass spectrometry using MALDI-TOF-TOF analysis. Erythrocytes with G6PD deficient and SCT showed higher carbonyl index values than control and similar profiles of carbonylated proteins; moreover, cytoskeletal and stress response proteins were identified as the main targets of oxidative damage. Therefore, both polymorphisms promote carbonylation on the same membrane proteins. Finally, these results allowed to reinforce the hypothesis of oxidative damage in erythrocyte membrane proteins as molecular mechanism of human adaptation to malaria infection.


Assuntos
Eritrócitos/parasitologia , Deficiência de Glucosefosfato Desidrogenase/sangue , Malária Falciparum/sangue , Proteínas de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Traço Falciforme/sangue , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia , Eritrócitos/enzimologia , Eritrócitos/metabolismo , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Deficiência de Glucosefosfato Desidrogenase/parasitologia , Humanos , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Oxirredução , Estresse Oxidativo , Plasmodium falciparum/genética , Carbonilação Proteica , Traço Falciforme/metabolismo , Traço Falciforme/parasitologia
6.
Proc Natl Acad Sci U S A ; 115(28): 7350-7355, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29946035

RESUMO

Sickle cell trait (AS) confers partial protection against lethal Plasmodium falciparum malaria. Multiple mechanisms for this have been proposed, with a recent focus on aberrant cytoadherence of parasite-infected red blood cells (RBCs). Here we investigate the mechanistic basis of AS protection through detailed temporal mapping. We find that parasites in AS RBCs maintained at low oxygen concentrations stall at a specific stage in the middle of intracellular growth before DNA replication. We demonstrate that polymerization of sickle hemoglobin (HbS) is responsible for this growth arrest of intraerythrocytic P. falciparum parasites, with normal hemoglobin digestion and growth restored in the presence of carbon monoxide, a gaseous antisickling agent. Modeling of growth inhibition and sequestration revealed that HbS polymerization-induced growth inhibition following cytoadherence is the critical driver of the reduced parasite densities observed in malaria infections of individuals with AS. We conclude that the protective effect of AS derives largely from effective sequestration of infected RBCs into the hypoxic microcirculation.


Assuntos
Replicação do DNA , DNA de Protozoário/biossíntese , Eritrócitos Anormais/metabolismo , Oxigênio/metabolismo , Plasmodium falciparum/metabolismo , Traço Falciforme/metabolismo , Antidrepanocíticos/farmacologia , Monóxido de Carbono/farmacologia , Eritrócitos Anormais/parasitologia , Humanos , Malária Falciparum/metabolismo , Traço Falciforme/parasitologia
7.
Nat Microbiol ; 2(12): 1600-1607, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28947801

RESUMO

The relevance of genetic factors in conferring protection to severe malaria has been demonstrated, as in the case of sickle cell trait and G6PD deficiency 1 . However, it remains unknown whether environmental components, such as dietary or metabolic variations, can contribute to the outcome of infection 2 . Here, we show that administration of a high-fat diet to mice for a period as short as 4 days impairs Plasmodium liver infection by over 90%. Plasmodium sporozoites can successfully invade and initiate replication but die inside hepatocytes, thereby are unable to cause severe disease. Transcriptional analyses combined with genetic and chemical approaches reveal that this impairment of infection is mediated by oxidative stress. We show that reactive oxygen species, probably spawned from fatty acid ß-oxidation, directly impact Plasmodium survival inside hepatocytes, and parasite load can be rescued by exogenous administration of the antioxidant N-acetylcysteine or the ß-oxidation inhibitor etomoxir. Together, these data reveal that acute and transient dietary alterations markedly impact the establishment of a Plasmodium infection and disease outcome.


Assuntos
Dieta Hiperlipídica/métodos , Interações Hospedeiro-Parasita/genética , Malária/dietoterapia , Acetilcisteína/metabolismo , Animais , Antioxidantes/metabolismo , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Células Hep G2 , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Humanos , Fígado/metabolismo , Fígado/parasitologia , Hepatopatias/metabolismo , Hepatopatias/parasitologia , Macrófagos/parasitologia , Macrófagos/patologia , Malária/sangue , Malária/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Carga Parasitária , Plasmodium berghei , Espécies Reativas de Oxigênio , Traço Falciforme/metabolismo , Esporozoítos/metabolismo
8.
EBioMedicine ; 14: 123-130, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27852523

RESUMO

BACKGROUND: Iron deficiency causes long-term adverse consequences for children and is the most common nutritional deficiency worldwide. Observational studies suggest that iron deficiency anemia protects against Plasmodium falciparum malaria and several intervention trials have indicated that iron supplementation increases malaria risk through unknown mechanism(s). This poses a major challenge for health policy. We investigated how anemia inhibits blood stage malaria infection and how iron supplementation abrogates this protection. METHODS: This observational cohort study occurred in a malaria-endemic region where sickle-cell trait is also common. We studied fresh RBCs from anemic children (135 children; age 6-24months; hemoglobin <11g/dl) participating in an iron supplementation trial (ISRCTN registry, number ISRCTN07210906) in which they received iron (12mg/day) as part of a micronutrient powder for 84days. Children donated RBCs at baseline, Day 49, and Day 84 for use in flow cytometry-based in vitro growth and invasion assays with P. falciparum laboratory and field strains. In vitro parasite growth in subject RBCs was the primary endpoint. FINDINGS: Anemia substantially reduced the invasion and growth of both laboratory and field strains of P. falciparum in vitro (~10% growth reduction per standard deviation shift in hemoglobin). The population level impact against erythrocytic stage malaria was 15.9% from anemia compared to 3.5% for sickle-cell trait. Parasite growth was 2.4 fold higher after 49days of iron supplementation relative to baseline (p<0.001), paralleling increases in erythropoiesis. INTERPRETATION: These results confirm and quantify a plausible mechanism by which anemia protects African children against falciparum malaria, an effect that is substantially greater than the protection offered by sickle-cell trait. Iron supplementation completely reversed the observed protection and hence should be accompanied by malaria prophylaxis. Lower hemoglobin levels typically seen in populations of African descent may reflect past genetic selection by malaria. FUNDING: National Institute of Child Health and Development, Bill and Melinda Gates Foundation, UK Medical Research Council (MRC) and Department for International Development (DFID) under the MRC/DFID Concordat.


Assuntos
Anemia/complicações , Anemia/tratamento farmacológico , Suplementos Nutricionais , Eritrócitos/parasitologia , Ferro/administração & dosagem , Malária Falciparum/etiologia , Malária Falciparum/prevenção & controle , Traço Falciforme/complicações , Anemia/etiologia , Anemia/metabolismo , Biomarcadores , Pré-Escolar , Suscetibilidade a Doenças , Feminino , Genótipo , Humanos , Lactente , Ferro/metabolismo , Malária Falciparum/epidemiologia , Malária Falciparum/metabolismo , Masculino , Plasmodium falciparum/crescimento & desenvolvimento , Vigilância da População , Traço Falciforme/genética , Traço Falciforme/metabolismo
9.
Endocrinology ; 157(8): 3036-46, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27171384

RESUMO

Characterization of the bone phenotype of 24-week-old female transgenic sickle cell disease (SCD), sickle cell trait (SCT) revealed significant reductions in bone mineral density and bone mineral content relative to control with a further significant decreased in SCD compared with SCT. By microcomputed tomography, femur middiaphyseal cortical area was significantly reduced in SCT and SCD. Cortical thickness was significantly decreased in SCD vs control. Diaphysis structural stiffness and strength were significantly reduced in SCT and SCD. Histomorphometry showed a significant increase in osteoclast perimeter in SCD and significantly decreased bone formation in SCD and SCT compared with control with a further significant decrease in SCD compared with SCT. Collagen-I mRNA was significantly decreased in tibiae from SCT and SCD and osterix, Runx2, osteoclacin, and Dmp-1 mRNA were significantly decreased in tibiae of SCD compared with control. Serum osteocalcin was significantly decreased and ferritin was significantly increased in SCD compared with control. Igf1 mRNA and serum IGF1 were significantly decreased in SCD and SCT. IGF1 protein was decreased in bone marrow stromal cells from SCT and SCD cultured in osteogenic media. Crystal violet staining revealed fewer cells and significantly reduced alkaline phosphatase positive mineralized nodules in SCT and SCD that was rescued by IGF1 treatment. We conclude that reduced bone mass in SCD and SCT mice carries architectural consequences that are detrimental to the mechanical integrity of femoral diaphysis. Furthermore reduced IGF1 and osteoblast terminal differentiation contributed to reduced bone formation in SCT and SCD mice.


Assuntos
Anemia Falciforme/complicações , Densidade Óssea , Doenças Ósseas Metabólicas , Osso e Ossos/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Traço Falciforme/complicações , Anemia Falciforme/sangue , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Animais , Doenças Ósseas Metabólicas/sangue , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/patologia , Reabsorção Óssea/sangue , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Traço Falciforme/sangue , Traço Falciforme/metabolismo , Traço Falciforme/patologia
10.
Am J Trop Med Hyg ; 94(5): 1002-7, 2016 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-27001763

RESUMO

Heterozygous hemoglobin S (HbAS), or sickle trait, protects children from life-threatening falciparum malaria, potentially by attenuating binding of Plasmodium-infected red blood cells (iRBCs) to extracellular ligands. Such binding is central to the pathogenesis of placental malaria (PM). We hypothesized that HbAS would be associated with reduced risks of PM and low birth weight (LBW). We tested this hypothesis in 850 delivering women in southern Malawi. Parasites were detected by polymerase chain reaction in placental and peripheral blood, and placentae were scored histologically for PM. The prevalence of HbAS was 3.7%, and 11.2% of infants were LBW (< 2,500 g). The prevalence of Plasmodium falciparum was 12.7% in placental and 8.5% in peripheral blood; 24.4% of placentae demonstrated histological evidence of P. falciparum HbAS was not associated with reduced prevalence of P. falciparum in placental (odds ratio [OR]: 1.27, 95% confidence interval [CI]: 0.50-3.23, P = 0.61) or peripheral blood (OR: 2.53, 95% CI: 1.08-2.54, P = 0.03), prevalence of histological PM (OR: 0.97, 95% CI: 0.40-2.34, P = 0.95), or prevalence of LBW (OR: 0.82, 95% CI: 0.24-2.73, P = 0.74). Mean (standard deviation) birth weights of infants born to HbAS (2,947 g [563]) and, homozygous hemoglobin A (2,991 g [465]) mothers were similar. Across a range of parasitologic, clinical, and histologic outcomes, HbAS did not confer protection from PM or its adverse effects.


Assuntos
Hemoglobina Falciforme/metabolismo , Malária Falciparum/parasitologia , Complicações Parasitárias na Gravidez/patologia , Traço Falciforme/metabolismo , Adulto , Antimaláricos/uso terapêutico , Estudos Transversais , Combinação de Medicamentos , Feminino , Genótipo , Humanos , Recém-Nascido de Baixo Peso , Recém-Nascido , Malaui/epidemiologia , Gravidez , Resultado da Gravidez , Prevalência , Pirimetamina/uso terapêutico , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Risco , Sulfadoxina/uso terapêutico , Adulto Jovem
11.
Sports Med ; 46(5): 629-39, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26666745

RESUMO

Sickle cell disease is a class of hemoglobinopathy in humans, which is the most common inherited disease in the world. Although complications of sickle cell disease start from polymerization of red blood cells during its deoxygenating phase, the oxidative stress resulting from the biological processes associated with this disease (ischaemic and hypoxic injuries, hemolysis and inflammation) has been shown to contribute to its pathophysiology. It is widely known that chronic exercise reduces oxidative stress in healthy people, mainly via improvement of antioxidant enzyme efficiency. In addition, recent studies in other diseases, as well as in sickle cell trait carriers and in a mouse model of sickle cell disease, have shown that regular physical activity could decrease oxidative stress. The purpose of this review is to summarize the role of oxidative stress in sickle cell disease and the effects of acute and chronic exercise on the pro-oxidant/antioxidant balance in sickle cell trait and sickle cell disease.


Assuntos
Anemia Falciforme/metabolismo , Exercício Físico/fisiologia , Estresse Oxidativo/fisiologia , Traço Falciforme/metabolismo , Animais , Modelos Animais de Doenças , Heme/metabolismo , Hemoglobina Falciforme/metabolismo , Hemólise , Humanos , Hipóxia/metabolismo , Ferro/sangue , Camundongos , NADPH Oxidases/metabolismo , Óxido Nítrico/metabolismo , Traumatismo por Reperfusão/metabolismo , Xantina Oxidase/metabolismo
12.
Hemoglobin ; 38(3): 165-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24601859

RESUMO

Studies dealing with rheological red blood cell (RBC) behavior in sickle cell trait carriers are scarce. Moreover, the association with α-thalassemia (α-thal), which also modifies erythrocyte behavior, has not always been taken into account. We analyzed erythrocyte deformability by means of a shear stress diffractometer, along with hematological and biochemical parameters (glucose and plasma lipids), given their possible influence on erythrocyte deformability, in 14 sickle cell trait carriers and 23 healthy controls. Nine patients were also α-thal carriers and five were not. Among the thalassemia carriers, eight were heterozygous and one was homozygous. When compared with controls, sickle cell trait carriers showed no differences for any of the biochemical parameters analyzed (p > 0.05), but significantly lower hemoglobin (Hb) (p = 0.003), mean corpuscular volume (MCV) and mean corpuscular Hb (MCH) (p < 0.001) levels, although no differences in erythrocyte deformability were observed at any of the shear stresses tested (p > 0.05). When comparing sickle cell trait carriers, with and without α-thal, no differences in erythrocyte deformability were observed (p > 0.05), in spite of the former showing lower MCV and MCH (p < 0.05) levels. Carriers of α-thal had lower Hb S [ß6(A3)Glu → Val; HBB: c.20A > T] levels (p = 0.013) than non carriers. The existence of a compensating mechanism seems reasonable because, despite presenting lower erythrocyte indices, which could worsen erythrocyte deformability, this rheological property improves when the percentage of Hb S is lower.


Assuntos
Deformação Eritrocítica , Eritrócitos/metabolismo , Traço Falciforme/metabolismo , Talassemia alfa/metabolismo , Adulto , Substituição de Aminoácidos , Índices de Eritrócitos , Eritrócitos/patologia , Feminino , Hemoglobina Falciforme/genética , Hemoglobina Falciforme/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Traço Falciforme/genética , Traço Falciforme/patologia , Talassemia alfa/genética , Talassemia alfa/patologia
13.
Blood Cells Mol Dis ; 52(4): 230-5, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24246527

RESUMO

Sickle cell disease (SCD) is associated with increase in oxidative stress and irreversible membrane changes that originates from the instability and polymerization of deoxygenated hemoglobin S (HbS). The relationship between erythrocyte membrane changes as assessed by a decrease in deformability and oxidative stress as assessed by an increase in heme degradation was investigated. The erythrocyte deformability and heme degradation for 27 subjects with SCD and 7 with sickle trait were compared with normal healthy adults. Changes in both deformability and heme degradation increased in the order of control to trait to non-crisis SCD to crisis SCD resulting in a very significantly negative correlation between deformability and heme degradation. However, a quantitative analysis of the changes in deformability and heme degradation for these different groups of subjects indicated that sickle trait had a much smaller effect on deformability than on heme degradation, while crisis affects deformability to a greater extent than heme degradation. These findings provide insights into the relative contributions of erythrocyte oxidative stress and membrane damage during the progression of SCD providing a better understanding of the pathophysiology of SCD.


Assuntos
Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Deformação Eritrocítica , Eritrócitos Anormais/metabolismo , Eritrócitos/metabolismo , Eritrócitos/patologia , Estresse Oxidativo , Adolescente , Adulto , Anemia Falciforme/diagnóstico , Criança , Feminino , Hemoglobina Fetal/metabolismo , Heme/metabolismo , Hemoglobina Falciforme/metabolismo , Hemoglobinas/metabolismo , Humanos , Masculino , Proteólise , Traço Falciforme/diagnóstico , Traço Falciforme/metabolismo , Traço Falciforme/patologia , Adulto Jovem
14.
Free Radic Biol Med ; 67: 377-86, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24316370

RESUMO

Red blood cells (RBCs) contain large amounts of iron and operate in highly oxygenated tissues. As a result, these cells encounter a continuous oxidative stress. Protective mechanisms against oxidation include prevention of formation of reactive oxygen species (ROS), scavenging of various forms of ROS, and repair of oxidized cellular contents. In general, a partial defect in any of these systems can harm RBCs and promote senescence, but is without chronic hemolytic complaints. In this review we summarize the often rare inborn defects that interfere with the various protective mechanisms present in RBCs. NADPH is the main source of reduction equivalents in RBCs, used by most of the protective systems. When NADPH becomes limiting, red cells are prone to being damaged. In many of the severe RBC enzyme deficiencies, a lack of protective enzyme activity is frustrating erythropoiesis or is not restricted to RBCs. Common hereditary RBC disorders, such as thalassemia, sickle-cell trait, and unstable hemoglobins, give rise to increased oxidative stress caused by free heme and iron generated from hemoglobin. The beneficial effect of thalassemia minor, sickle-cell trait, and glucose-6-phosphate dehydrogenase deficiency on survival of malaria infection may well be due to the shared feature of enhanced oxidative stress. This may inhibit parasite growth, enhance uptake of infected RBCs by spleen macrophages, and/or cause less cytoadherence of the infected cells to capillary endothelium.


Assuntos
Antioxidantes/metabolismo , Eritrócitos/metabolismo , Glutationa/metabolismo , NADP/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Anemia Hemolítica/metabolismo , Anemia Hemolítica/patologia , Eritrócitos/patologia , Eritropoese , Deficiência de Glucosefosfato Desidrogenase/metabolismo , Deficiência de Glucosefosfato Desidrogenase/patologia , Humanos , Malária/metabolismo , Malária/prevenção & controle , Oxirredução , Estresse Oxidativo , Traço Falciforme/metabolismo , Traço Falciforme/patologia , Talassemia/metabolismo , Talassemia/patologia
15.
Am J Hematol ; 88(11): E255-60, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23828223

RESUMO

2,2-Dimethylbutyrate (HQK-1001), an orally-bioavailable promoter-targeted fetal globin gene-inducing agent, was evaluated in an open-label, randomized dose-escalation study in 52 subjects with hemoglobin SS or S/ß(0) thalassemia. HQK-1001 was administered daily for 26 weeks at 30 mg/kg (n = 15), 40 mg/kg (n = 18) and 50 mg/kg (n = 19), either alone (n = 21) or with hydroxyurea (n = 31). The most common drug-related adverse events were usually mild or moderate and reversible. Gastritis was graded as severe in three subjects at 40 mg/kg and was considered the dose-limiting toxicity. Subsequently all subjects were switched to the maximum tolerated dose of 30 mg/kg. Due to early discontinuations for blood transfusions, adverse events or non-compliance, only 25 subjects (48%) completed the study. Drug plasma concentrations were sustained above targeted levels at 30 mg/kg. Increases in fetal hemoglobin (Hb F) were observed in 42 subjects (80%), and 12 (23%) had increases ≥4%. The mean increase in Hb F was 2% [95% confidence interval (CI), 0.8-3.2%] in 21 subjects receiving HQK-1001 alone and 2.7% (95% CI, 1.7-3.8%) in 31 subjects receiving HQK-1001 plus hydroxyurea. Total hemoglobin increased by a mean of 0.65 g/dL (95% CI, 0.5-1.0 g/dL), and 13 subjects (25%) had increases ≥1 g/dL. Future studies are warranted to evaluate the therapeutic potential of HQK-1001 in sickle cell disease. .


Assuntos
Anemia Falciforme/tratamento farmacológico , Butiratos/administração & dosagem , Hemoglobina Fetal/biossíntese , Hematínicos/administração & dosagem , Traço Falciforme/tratamento farmacológico , Regulação para Cima/efeitos dos fármacos , Administração Oral , Adolescente , Adulto , Anemia Falciforme/sangue , Anemia Falciforme/metabolismo , Antidrepanocíticos/uso terapêutico , Butiratos/efeitos adversos , Butiratos/farmacocinética , Butiratos/uso terapêutico , Criança , Estudos de Coortes , Relação Dose-Resposta a Droga , Feminino , Hemoglobina Fetal/análise , Hemoglobina Fetal/genética , Gastrite/induzido quimicamente , Gastrite/epidemiologia , Hematínicos/efeitos adversos , Hematínicos/farmacocinética , Hematínicos/uso terapêutico , Heterozigoto , Humanos , Hidroxiureia/uso terapêutico , Incidência , Masculino , Pessoa de Meia-Idade , Pacientes Desistentes do Tratamento , Regiões Promotoras Genéticas/efeitos dos fármacos , Traço Falciforme/sangue , Traço Falciforme/complicações , Traço Falciforme/metabolismo , Adulto Jovem , Talassemia beta/complicações
16.
J Appl Physiol (1985) ; 113(4): 549-56, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22700803

RESUMO

To assess the effects of regular physical activity on muscle functional characteristics of carriers of sickle cell trait (SCT), 39 untrained (U) and trained (T) hemoglobin (Hb)AA (CON) and SCT subjects (U-CON, n = 12; U-SCT, n = 8; T-CON, n = 10; and T-SCT, n = 9) performed a graded exercise and a time to exhaustion (T(ex)) test, and were subjected to a muscle biopsy. Maximal power, total work performed during T(ex), citrate synthase and cytochrome c oxidase (COX) activities, respiratory chain complexes I and IV content, and capillary density (CD), diameter (COD), and surface area (CSA) were upregulated by the same proportion in T-CON and T-SCT compared with their untrained counterparts. These proportionally similar differences imply that the observed discrepancies between U-SCT and U-CON remained in the trained subjects. Specifically, both CD and COX remained and tended to remain lower, and both COD and CSA remained and tended to remain higher in T-SCT than in T-CON. Besides, carriers of SCT displayed specific adaptations with regular physical activity: creatine kinase activity; complexes II, III, and V content; and type I fiber surface area and capillary tortuosity were lower or unchanged in T-SCT than in U-SCT. In summary, our results show that 1) carriers of SCT adapted almost similarly to CON to regular physical activity for most of the studied muscle characteristics, 2) oxidative potential remains altered in physically active carriers of SCT compared with HbAA counterparts, and 3) the specific remodeling of muscle microvascular network persists in the trained state.


Assuntos
Capilares/fisiopatologia , Metabolismo Energético , Exercício Físico , Microcirculação , Contração Muscular , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/fisiopatologia , Traço Falciforme/fisiopatologia , Adaptação Fisiológica , Adulto , Biópsia , Citrato (si)-Sintase/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Teste de Esforço , Hemoglobina A/genética , Hemoglobina A/metabolismo , Hemoglobina Falciforme/genética , Hemoglobina Falciforme/metabolismo , Heterozigoto , Humanos , Masculino , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Força Muscular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Resistência Física , Fluxo Sanguíneo Regional , Comportamento Sedentário , Traço Falciforme/sangue , Traço Falciforme/genética , Traço Falciforme/metabolismo , Traço Falciforme/patologia , Fatores de Tempo , Adulto Jovem
17.
Biophys J ; 102(5): 1137-43, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22404936

RESUMO

Collapse and sudden death in physical training are the most serious complications of sickle cell trait (SCT). There is evidence that erythrocytes in SCT patients aggregate during strenuous exercise, likely because of adhesive interactions with the extracellular matrix (ECM) and endothelial cells, and because of their irregular viscoelastic properties. This results in inflammation, blood flow impairment, and vaso-occlusive events. However, the exact role of stress conditions and how they lead to these complications is virtually unknown. Using single-molecule atomic force microscopy experiments, we found that epinephrine, a hormone that is secreted under stressful conditions, increases both the frequency and strength of adhesion events between basal cell adhesion molecule (BCAM/Lu) and ECM laminin, and between intercellular adhesion molecule-4 (ICAM-4) and endothelial α(v)ß(3), compared with nonstimulated SCT erythrocytes. Increases in adhesion frequency provide significant evidence of the role of epinephrine in BCAM/Lu-laminin and ICAM-4-α(v)ß(3) bonding, and suggest mechanisms of vaso-occlusion during physical exertion in SCT.


Assuntos
Moléculas de Adesão Celular/metabolismo , Epinefrina/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Sistema do Grupo Sanguíneo Lutheran/metabolismo , Traço Falciforme/sangue , Adulto , Adesão Celular/efeitos dos fármacos , Membrana Eritrocítica/metabolismo , Humanos , Integrina alfaVbeta3/metabolismo , Laminina/metabolismo , Traço Falciforme/metabolismo
18.
Acta Physiol (Oxf) ; 205(4): 541-50, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22409225

RESUMO

AIM: Alpha-thalassaemia is known to reduce intra-erythrocyte HbS (sickle haemoglobin) concentration in sickle cell trait (SCT) subjects. Because HbS was shown to increase oxidative stress, the purpose of this study was to assess the effects of the coexistence of α-thalassaemia and SCT on oxidative stress markers and nitric oxide (NO) metabolism after an acute physical exercise. METHODS: Forty subjects (age: 23.5 ± 2.21 years), SCT carriers (HbAS) or healthy subjects (HbAA), with (-αT) or without (-NαT) an associated α-thalassaemia took part in the study. Plasma markers of oxidative stress [advanced oxidation protein products (AOPP), protein carbonyl, malondialdehyde (MDA) and nitrotyrosine], anti-oxidant defences and NO metabolism (NOx) were measured at rest (T(rest)), immediately following an incremental maximal exercise test (T(ex)) and during recovery (T(1h), T(2h) and T(24h)). RESULTS: Malondialdehyde expressed as the percentage of changes from baseline was significantly higher in the HbAS-NαT compared with HbAS-αT during recovery (+36.3 ± 14.1% vs. -1.8 ± 13.2% at T(1h), P = 0.02; +36.6 ± 13.4% vs. -11.4 ± 12.5% at T(2h), P = 0.004 and +24.1 ± 12.3% vs. -14.4 ± 11.5% at T(24h), P = 0.02 in HbAS-NαT vs. HbAS-αT). Compared with HbAS-NαT, HbAS-αT had a higher NOx change from baseline at T(ex) (-23.4 ± 20.6% vs. +57.7 ± 19.3%, respectively; P = 0.005) and lower nitrotyrosine change from baseline at T(1h) (+7.2 ± 22.2% vs. +93.5%±29.3%, respectively; P = 0.04). CONCLUSION: All these data suggest that the presence of α-thalassaemia may blunt the higher level of oxidative stress and the impaired bioavailability of NO observed in the SCT carriers.


Assuntos
Exercício Físico/fisiologia , Estresse Oxidativo/fisiologia , Traço Falciforme/metabolismo , Talassemia alfa/metabolismo , Adulto , Antioxidantes/metabolismo , Biomarcadores , Recuperação de Fluorescência Após Fotodegradação , Humanos , Masculino , Óxido Nítrico/sangue , Óxido Nítrico/metabolismo , Traço Falciforme/complicações , Tirosina/análogos & derivados , Tirosina/sangue , Tirosina/metabolismo , Adulto Jovem , Talassemia alfa/complicações
19.
Diabet Med ; 29(9): e334-7, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22356301

RESUMO

AIMS: To determine the prevalence and effects of sickle cell trait on metabolic control in a Cameroonian diabetic population in a tertiary care setup. METHODS: This was a cross-sectional study involving 73 consecutive outpatients with Type 2 diabetes recruited from the Yaounde National Diabetes and Obesity Centre. Sickle cell trait status was based on haemoglobin electrophoresis. Metabolic control was assessed by plasma glucose and HbA(1c), and comparisons made between participants with and without sickle cell trait, with adjustment for confounders through linear regressions models. RESULTS: The prevalence of sickle cell trait was 19%, without sex difference, and comparable with figures in individuals without diabetes in this setting. Participants with diabetes and sickle cell trait were older than the non-trait participants (66 vs. 58 years, P = 0.02). Otherwise, clinical and biological profile including indicators of metabolic control were similarly distributed between trait and non-trait participants (all P >0.08). After adjustment for confounders, sickle cell trait was unrelated to fasting glucose (ß = 0.02; 95% confidence interval -37.68-43.30) and HbA(1c) (ß = -0.03, 95% confidence interval -1.18-0.93), and did not affect the relationship between the two markers of diabetes control (ß = -0.03, 95% confidence interval -1.18-0.89). CONCLUSIONS: Sickle cell trait was as frequent in this subgroup of patients with Type 2 diabetes as in the general population, suggesting no specific association with diabetes. It does not affect the metabolic control of diabetes. However, how this translates into long-term outcome needs to be fully elucidated in this setting, with an increasing population with both sickle cell trait and diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/metabolismo , Traço Falciforme/epidemiologia , Traço Falciforme/metabolismo , África Subsaariana/epidemiologia , Idoso , Glicemia/metabolismo , Camarões/epidemiologia , Comorbidade , Estudos Transversais , Diabetes Mellitus Tipo 2/etnologia , Feminino , Hemoglobinas Glicadas/metabolismo , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Prevalência , Traço Falciforme/etnologia
20.
Science ; 334(6060): 1283-6, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-22075726

RESUMO

The hemoglobins S and C protect carriers from severe Plasmodium falciparum malaria. Here, we found that these hemoglobinopathies affected the trafficking system that directs parasite-encoded proteins to the surface of infected erythrocytes. Cryoelectron tomography revealed that the parasite generated a host-derived actin cytoskeleton within the cytoplasm of wild-type red blood cells that connected the Maurer's clefts with the host cell membrane and to which transport vesicles were attached. The actin cytoskeleton and the Maurer's clefts were aberrant in erythrocytes containing hemoglobin S or C. Hemoglobin oxidation products, enriched in hemoglobin S and C erythrocytes, inhibited actin polymerization in vitro and may account for the protective role in malaria.


Assuntos
Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Eritrócitos/parasitologia , Eritrócitos/ultraestrutura , Hemoglobina C/análise , Hemoglobina Falciforme/análise , Plasmodium falciparum/fisiologia , Citoesqueleto de Actina/metabolismo , Citoplasma/ultraestrutura , Tomografia com Microscopia Eletrônica , Membrana Eritrocítica/ultraestrutura , Eritrócitos/metabolismo , Hemoglobina A/análise , Hemoglobina C/genética , Doença da Hemoglobina C/complicações , Doença da Hemoglobina C/metabolismo , Hemoglobina Falciforme/genética , Humanos , Malária Falciparum/complicações , Malária Falciparum/metabolismo , Malária Falciparum/patologia , Oxirredução , Plasmodium falciparum/crescimento & desenvolvimento , Transporte Proteico , Proteínas de Protozoários/metabolismo , Traço Falciforme/complicações , Traço Falciforme/metabolismo , Vesículas Transportadoras/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...