Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Haematol ; 2023 Dec 16.
Article in English | MEDLINE | ID: mdl-38104534

ABSTRACT

BACKGROUND: Most patients with lower risk myelodysplastic neoplasms (MDS) become RBC transfusion-dependent, resulting in iron overload, which is associated with an increased oxidative stress state. Iron-chelation therapy is applied to attenuate the toxic effects of this state. Deferiprone (DFP) is an oral iron chelator, which is not commonly used in this patient population, due to safety concerns, mainly agranulocytosis. The purpose of this study was to assess the effect of DFP, on oxidative stress parameters in iron overloaded RBC transfusion-dependent patients with lower risk MDS. METHODS: Adult lower-risk MDS patients with a cumulative transfusion burden of >20 red blood cells units and evidence of iron overload (serum ferritin >1,000 ng/mL) were included in this study. DFP was administered (100 mg/kg/day) for 4 months. Blood samples for oxidative stress parameters and iron overload parameters were done at baseline and monthly: reactive oxygen species (ROS), phosphatidylserine, reduced glutathione, membrane lipid peroxidation, serum ferritin and cellular labile iron pool. The primary efficacy variable was ROS. Tolerability and side-effects were recorded as well. A paired t-test was applied for statistical analyses. RESULTS: Eighteen patients were treated with DFP. ROS significantly decreased in all cell lineages: median decrease of 58.6% in RBC, 33.3% in PMN, and 39.8% in platelets (p<0.01 for all). Other oxidative stress markers improved: phosphatidylserine decreased by 57.95%, lipid peroxidase decreased by 141.3%, and reduced gluthathione increased by 72.8% (p<0.01 for all). The iron-overload marker, cellular labile iron pool, decreased by 35% in RBCs, 44.3% in PMN, and 46.3% in platelets (p<0.01 for all). No significant changes were observed in SF levels. There were no events of agranulocytosis. All AEs were grade 1-2. CONCLUSIONS: Herein we showed preliminary evidence that DFP decreases iron-induced oxidative stress in MDS patients with a good tolerability profile (albeit a short follow-up period). No cases of severe neutropenia or agranulocytosis were reported. The future challenge is to prove that reduction in iron toxicity will eventually be translated into a clinically meaningful improvement.

2.
Mol Diagn Ther ; 23(2): 245-261, 2019 04.
Article in English | MEDLINE | ID: mdl-30484264

ABSTRACT

Cell oxidative status, which represents the balance between oxidants and antioxidants, is involved in normal functions. Under pathological conditions, there is a shift toward the oxidants, leading to oxidative stress, which is cytotoxic, causing oxidation of cellular components that result in cell death and organ damage. Thalassemia is a hereditary hemolytic anemia caused by mutations in globin genes that cause reduced or complete absence of specific globin chains (commonly, α or ß). Although oxidative stress is not the primary etiology of thalassemia, it mediates several of its pathologies. The main causes of oxidative stress in thalassemia are the degradation of the unstable hemoglobin and iron overload-both stimulate the production of excess free radicals. The symptoms aggravated by oxidative stress include increased hemolysis, ineffective erythropoiesis and functional failure of vital organs such as the heart and liver. The oxidative status of each patient is affected by multiple internal and external factors, including genetic makeup, health conditions, nutrition, physical activity, age, and the environment (e.g., air pollution, radiation). In addition, oxidative stress is influenced by the clinical manifestations of the disease (unpaired globin chains, iron overload, anemia, etc.). Application of personalized (theranostics) medicine principles, including diagnostic tests for selecting targeted therapy, is therefore important for optimal treatment of the oxidative stress of these patients. We summarize the role of oxidative stress and the current and potential antioxidative therapeutics in ß-thalassemia and describe some methodologies, mostly cellular, that might be helpful for application of a theranostics approach to therapy.


Subject(s)
Oxidative Stress , beta-Thalassemia/pathology , Erythropoiesis , Hemoglobins/metabolism , Hemolysis , Humans , Oxidation-Reduction , beta-Thalassemia/therapy
3.
Neonatology ; 114(4): 303-306, 2018.
Article in English | MEDLINE | ID: mdl-30011398

ABSTRACT

BACKGROUND: Fetal-maternal hemorrhage (FMH) occurs when fetal red blood cells (RBC) pass into the maternal circulation as a result of obstetric- or trauma-related complications to pregnancy. Their detection in the maternal blood is commonly used as a diagnostic test. There is, however, a serious and general limitation to this test that is sometimes ignored. Fetal RBC carrying the father's antigens (most crucially, the ABO blood antigens) may be incompatible with the mother's plasma. They are expected to be eliminated by the maternal natural antibodies, thus, negative results may be false. OBJECTIVES: By simulating fetal-maternal ABO incompatibility, we studied the fate of fetal RBC in vitro. METHODS: Adult blood samples (n = 6) of O-blood group (type) were mixed with 1-5% cord blood or neonatal blood of A- or O-type, representing incompatible and compatible fetal RBC, respectively. The survival of fetal RBC was quantified after an overnight incubation. The supernatant was assayed for fetal hemoglobin (HbF) using the spectrophotometric alkaline-resistance benzidine assay, while the pellet was assayed for HbF/carbonic anhydrase (CA) expression in RBC by flow cytometry. The HbFhigh/CAlow phenotype characterizes fetal RBC. RESULTS: Both assays demonstrated disappearance of the fetal RBC due to lysis upon incubation in incompatible blood. CONCLUSIONS: A similar situation may also occur in vivo. Thus, under these conditions, negative results in the FMH test may be false, and lead to misdiagnosis.


Subject(s)
ABO Blood-Group System/immunology , Blood Group Incompatibility/blood , Erythrocyte Indices , Fetal Blood/cytology , Fetomaternal Transfusion/diagnosis , Adult , Female , Fetomaternal Transfusion/blood , Hemoglobins/analysis , Humans , Pregnancy
4.
Hemoglobin ; 42(2): 138-140, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29745271

ABSTRACT

The major hemoglobin (Hb) during fetal life is fetal Hb (Hb F). It is mostly replaced by adult Hbs before birth and during the first year of life. In adults, where Hb F comprises <2.0% of the total Hb, it is not homogenously distributed among the red blood cells (RBCs) but is concentrated in a few RBCs, termed F-cells. Interestingly, for reasons that are unclear, Hb F increases in the maternal circulation during pregnancy. This increased Hb F could have two potential origins that are not mutually exclusive: A) maternal origin, due to inducing environment of Hb F in the maternal erythroid precursors; B) fetal origin, due to fetal cells crossing the placenta and entering the maternal circulation. The question we present herein is whether the observed increased Hb F in the maternal circulation during pregnancy is, at least partially, derived from the fetal origin. Peripheral blood was obtained from normal neonates (1-3 days old), adult men and pregnant and non pregnant women. The RBCs were stained for Hb F and carbonic anhydrase (CA) using a fetal cell count kit and analyzed by flow cytometry. Fetal and adult F-cells were distinguished by their expression of Hb F and CA. Fetal F-cells were Hb F++/CA-, while adult F-cells were Hb F+/CA+. Comparing pregnant and non pregnant women samples (n = 10), we found six samples of pregnant women with 0.2-1.7% fetal cells, but none in the non pregnant group. These results support the possibility that at least part of the increase in Hb F during pregnancy is due to fetal cells entering the maternal circulation.


Subject(s)
Fetal Hemoglobin/analysis , Fetomaternal Transfusion , Adult , Erythrocytes/chemistry , Female , Fetal Blood/cytology , Humans , Infant, Newborn , Male , Pregnancy
5.
Free Radic Biol Med ; 88(Pt A): 63-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25937178

ABSTRACT

The complement (C') system and redox status play important roles in the physiological functioning of the body, such as the defense system, but they are also involved in various pathological conditions, including hemolytic anemia. Herein, we review the interaction between the C' and the redox systems in C'-mediated hemolytic anemias, paroxysmal nocturnal hemoglobinuria (PNH) and autoimmune hemolytic anemia, including acute hemolytic transfusion reaction. Blood cells in these diseases have been shown to have increased oxidative status, which was further elevated by interaction with activated C'. The results suggest that oxidative stress, in conjunction with activated C', may cause the underlying symptoms of these diseases, such as intra- and extravascular hemolysis and thrombotic complications. Antioxidants ameliorate oxidative stress by preventing generation of free radicals, by scavenging and preventing their accumulation, and by correcting their cellular damage. Antioxidants have been shown to reduce the oxidative stress and inhibit hemolysis as well as platelet activation mediated by activated C'. This raises the possibility that treatment with antioxidants might be considered as a potential therapeutic modality for C'-mediated hemolytic anemias. Currently, eculizumab, a humanized monoclonal antibody that specifically targets the C' protein C5, is the main treatment modality for PNH. However, because antioxidants are well tolerated and relatively inexpensive, they might be considered as potential adjuvants or an alternative therapeutic modality for PNH and other C'-mediated hemolytic anemias.


Subject(s)
Anemia, Hemolytic, Autoimmune/physiopathology , Antioxidants/pharmacology , Complement Activation , Hemoglobinuria, Paroxysmal/physiopathology , Oxidative Stress/drug effects , Anemia, Hemolytic, Autoimmune/drug therapy , Complement System Proteins/metabolism , Hemoglobinuria, Paroxysmal/drug therapy , Humans
6.
Thromb Res ; 135(3): 521-5, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25613926

ABSTRACT

Patients with ß-thalassemia major and mainly intermedia have an increased risk for developing venous and arterial thrombosis which may be related to circulating pathological red blood cells (RBC) and continuous platelet activation. In the present study we used a modified thalassemic mice model in conjunction with a "real-time" carotid thrombus formation procedure to investigate thrombotic complications of thalassemia. Heterozygous Th3/+ mice, which lack one copy of their ß-major and ß-minor globin genes, exhibit anomalies in RBC size and shape, chronic anemia and splenomegaly which recapitulate the phenotype of human ß-thalassemia intermedia. Flow cytometry measurements showed higher reactive oxygen species generation, indicating oxidative stress, in platelets and RBC of the thalassemic mice compared with wild type mice concomitant with an increase in reduced glutathione content which may represent a compensatory response to oxidative stress, and exposed phosphatidylserine which indicates platelet activation. To elucidate the effect of thalassemia on the development of arterial thrombosis, we studied photochemical-induced real-time thrombus formation in the carotid artery of these mice. The results indicated a significantly shorter "time to occlusion" in the thalassemic mice compared to wild type mice, which was prolonged following in vivo aspirin treatment. We suggest that this mouse model may contribute to our understanding of platelet activation and the hypercoagulable state in thalassemia and lay foundations to screening of anti-platelet drugs as well as anti-oxidants as possible therapeutics for prevention of thrombosis in thalassemia patients.


Subject(s)
Oxidative Stress , Platelet Activation , Thalassemia/complications , Thrombophilia/etiology , Thrombosis/etiology , Animals , Blood Platelets/pathology , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reactive Oxygen Species , Thalassemia/blood , Thalassemia/metabolism , Thalassemia/pathology , Thrombophilia/blood , Thrombophilia/metabolism , Thrombophilia/pathology , Thrombosis/blood , Thrombosis/metabolism , Thrombosis/pathology
7.
Anemia ; 2012: 943974, 2012.
Article in English | MEDLINE | ID: mdl-22496973

ABSTRACT

We studied the nature of enucleated RBCs containing DNA remnants, Howell-Jolly (HJ) RBCs and reticulocytes (retics), that are characteristically present in the circulation of thalassemic patients, especially after splenectomy. Using flow cytometry methodology, we measured oxidative status parameters of these cells in patients with ß-thalassemia. In each patient studied, these cells had higher content of reactive oxygen species and exposed phosphatidylserine compared with their DNA-free counterparts. These results suggest that oxidative stress in thalassemic developing erythroid precursors might, through DNA-breakage, generate HJ-retics and HJ-RBCs and that oxidative stress-induced externalization of phosphatidylserine is involved in the removal of these cells from the circulation by the spleen, a mechanism similar to that of the removal of senescent RBCs.

8.
Ann Hematol ; 90(5): 509-13, 2011 May.
Article in English | MEDLINE | ID: mdl-21063708

ABSTRACT

In the present study, we questioned the role of oxidative stress in hereditary spherocytosis (HS), where red blood cells (RBC) have a shortened survival due to primary deficiency in membrane proteins. Using flow cytometry techniques, we showed that RBC derived from 17 HS patients of seven families generate more reactive oxygen species, membrane lipid peroxides, and less reduced glutathione than normal RBC. Following in vitro incubation of HS-RBC from seven patients with a fermentation bioproduct of Carica papaya (fermented papaya preparation (FPP)) with known antioxidative properties, oxidative stress markers were significantly reduced. Similar results were obtained following treatment with FPP for 3 months of 10 adult HS patients, as well as decreased tendency to undergo hemolysis. The hemoglobin levels increased by >1 g/dl, mean corpuscular hemoglobin concentration decreased by >1 g/dl, and the reticulocyte count decreased by 0.93%. Concomitantly, lactic dehydrogenase decreased by 17% and indirect bilirubin by 50%. A significant decrease in malonyldialdehyde was also detected. These data indicate that oxidative stress plays an important role in the pathophysiology of HS which can be ameliorated by an antioxidant such as FPP. Additional clinical trials with FPP and other antioxidants are warranted.


Subject(s)
Antioxidants/therapeutic use , Carica/chemistry , Hemolysis/drug effects , Oxidative Stress/drug effects , Phytotherapy , Plant Preparations/therapeutic use , Spherocytosis, Hereditary/drug therapy , Adolescent , Adult , Erythrocyte Indices/drug effects , Erythrocytes/drug effects , Female , Fermentation , Glutathione/blood , Humans , Lipid Peroxides/blood , Male , Oxidation-Reduction , Reactive Oxygen Species/blood , Reticulocyte Count , Spherocytosis, Hereditary/blood , Spherocytosis, Hereditary/physiopathology , Young Adult
9.
Anemia ; 2010: 978710, 2010.
Article in English | MEDLINE | ID: mdl-21490911

ABSTRACT

Because of its stimulating effect on RBC production, erythropoietin (Epo) is used to treat anemia, for example, in patients on dialysis or on chemotherapy. In ß-thalassemia, where Epo levels are low relative to the degree of anemia, Epo treatment improves the anemia state. Since RBC and platelets of these patients are under oxidative stress, which may be involved in anemia and thromboembolic complications, we investigated Epo as an antioxidant. Using flow-cytometry technology, we found that in vitro treatment with Epo of blood cells from these patients increased their glutathione content and reduced their reactive oxygen species, membrane lipid peroxides, and external phosphatidylserine. This resulted in reduced susceptibility of RBC to undergo hemolysis and phagocytosis. Injection of Epo into heterozygous (Hbb(th3/+)) ß-thalassemic mice reduced the oxidative markers within 3 hours. Our results suggest that, in addition to stimulating RBC and fetal hemoglobin production, Epo might alleviate symptoms of hemolytic anemias as an antioxidant.

SELECTION OF CITATIONS
SEARCH DETAIL
...