Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
1.
Acta Haematol ; 2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38104534

RESUMO

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.
Elife ; 122023 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-38153418

RESUMO

Myelodysplastic syndrome (MDS) is a heterogeneous group of bone marrow stem cell disorders characterized by ineffective hematopoiesis and cytopenias, most commonly anemia. Red cell transfusion therapy for anemia in MDS results in iron overload, correlating with reduced overall survival. Whether the treatment of iron overload benefits MDS patients remains controversial. We evaluate underlying iron-related pathophysiology and the effect of iron chelation using deferiprone on erythropoiesis in NUP98-HOXD13 transgenic mice, a highly penetrant well-established MDS mouse model. Our results characterize an iron overload phenotype with aberrant erythropoiesis in these mice which was reversed by deferiprone-treatment. Serum erythropoietin levels decreased while erythroblast erythropoietin receptor expression increased in deferiprone-treated MDS mice. We demonstrate, for the first time, normalized expression of the iron chaperones Pcbp1 and Ncoa4 and increased ferritin stores in late-stage erythroblasts from deferiprone-treated MDS mice, evidence of aberrant iron trafficking in MDS erythroblasts. Importantly, erythroblast ferritin is increased in response to deferiprone, correlating with decreased erythroblast ROS. Finally, we confirmed increased expression of genes involved in iron uptake, sensing, and trafficking in stem and progenitor cells from MDS patients. Taken together, our findings provide evidence that erythroblast-specific iron metabolism is a novel potential therapeutic target to reverse ineffective erythropoiesis in MDS.


Assuntos
Anemia , Sobrecarga de Ferro , Humanos , Camundongos , Animais , Eritropoese , Deferiprona , Sobrecarga de Ferro/complicações , Ferro , Camundongos Transgênicos , Ferritinas , Quelantes de Ferro/farmacologia
4.
Front Physiol ; 12: 604738, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33664673

RESUMO

Membrane shedding in the form of extracellular vesicles plays a key role in normal physiology and pathology. Partial disturbance of the membrane-cytoskeleton linkage and increased in the intracellular Ca content are considered to be mechanisms underlying the process, but it is questionable whether they constitute the primary initiating steps. Homeostasis of the redox system, which depends on the equilibrium between oxidants and antioxidants, is crucial for many cellular processes. Excess oxidative power results in oxidative stress, which affects many cellular components, including the membrane. Accumulating evidence suggests that oxidative stress indirectly affects membrane shedding most probably by affecting the membrane-cytoskeleton and the Ca content. In red blood cells (RBCs), changes in both the redox system and membrane shedding occur throughout their life-from birth-their production in the bone marrow, to death-aging in the peripheral blood and removal by macrophages in sites of the reticuloendothelial system. Both oxidative stress and membrane shedding are disturbed in diseases affecting the RBC, such as the hereditary and acquired hemolytic anemias (i.e., thalassemia, sickle cell anemia, and autoimmune hemolytic anemia). Herein, I review some data-based and hypothetical possibilities that await experimental confirmation regarding some aspects of the interaction between the redox system and membrane shedding and its role in the normal physiology and pathology of RBCs.

5.
Br J Haematol ; 193(3): 637-658, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33723861

RESUMO

Increasing evidence suggests that free haem and iron exert vasculo-toxic and pro-inflammatory effects by activating endothelial and immune cells. In the present retrospective study, we compared serum samples from transfusion-dependent patients with ß-thalassaemia major and intermedia, hereditary spherocytosis and sickle cell disease (SCD). Haemolysis, transfusions and ineffective erythropoiesis contribute to haem and iron overload in haemolytic patients. In all cohorts we observed increased systemic haem and iron levels associated with scavenger depletion and toxic 'free' species formation. Endothelial dysfunction, oxidative stress and inflammation markers were significantly increased compared to healthy donors. In multivariable logistic regression analysis, oxidative stress markers remained significantly associated with both haem- and iron-related parameters, while soluble vascular cell adhesion molecule 1 (sVCAM-1), soluble endothelial selectin (sE-selectin) and tumour necrosis factor α (TNFα) showed the strongest association with haem-related parameters and soluble intercellular adhesion molecule 1 (sICAM-1), sVCAM-1, interleukin 6 (IL-6) and vascular endothelial growth factor (VEGF) with iron-related parameters. While hereditary spherocytosis was associated with the highest IL-6 and TNFα levels, ß-thalassaemia major showed limited inflammation compared to SCD. The sVCAM1 increase was significantly lower in patients with SCD receiving exchange compared to simple transfusions. The present results support the involvement of free haem/iron species in the pathogenesis of vascular dysfunction and sterile inflammation in haemolytic diseases, irrespective of the underlying haemolytic mechanism, and highlight the potential therapeutic benefit of iron/haem scavenging therapies in these conditions.


Assuntos
Anemia Falciforme/sangue , Heme/metabolismo , Hemoglobinas/metabolismo , Ferro/sangue , Esferocitose Hereditária/sangue , Talassemia beta/sangue , Adolescente , Adulto , Anemia Falciforme/terapia , Transfusão de Sangue , Criança , Pré-Escolar , Endotélio Vascular/metabolismo , Feminino , Humanos , Inflamação/sangue , Molécula 1 de Adesão Intercelular/sangue , Interleucina-6/sangue , Masculino , Esferocitose Hereditária/terapia , Fator de Necrose Tumoral alfa/sangue , Molécula 1 de Adesão de Célula Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/sangue , Talassemia beta/terapia
6.
J Clin Med ; 8(12)2019 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-31810354

RESUMO

Thalassemia (thal) is a hereditary chronic hemolytic anemia due to a partial or complete deficiency in the production of globin chains, in most cases, α or ß, which compose, together with the iron-containing porphyrins (hemes), the hemoglobin molecules in red blood cells (RBC). The major clinical symptom of ß-thal is severe chronic anemia-a decrease in RBC number and their hemoglobin content. In spite of the improvement in therapy, thal still severely affects the quality of life of the patients and their families and imposes a substantial financial burden on the community. These considerations position ß-thal, among other hemoglobinopathies, as a major health and social problem that deserves increased efforts in research and its clinical application. These efforts are based on clinical studies, experiments in animal models and the use of erythroid cells grown in culture. The latter include immortal cell lines and cultures initiated by erythroid progenitor and stem cells derived from the blood and RBC producing (erythropoietic) sites of normal and thal donors, embryonic stem cells, and recently, "induced pluripotent stem cells" generated by manipulation of differentiated somatic cells. The present review summarizes the use of erythroid cultures, their technological aspects and their contribution to the research and its clinical application in thal. The former includes deciphering of the normal and pathological biology of the erythroid cell development, and the latter-their role in developing innovative therapeutics-drugs and methods of gene therapy, as well as providing an alternative source of RBC that may complement or substitute blood transfusions.

7.
Mol Diagn Ther ; 23(2): 245-261, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30484264

RESUMO

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.


Assuntos
Estresse Oxidativo , Talassemia beta/patologia , Eritropoese , Hemoglobinas/metabolismo , Hemólise , Humanos , Oxirredução , Talassemia beta/terapia
8.
Neonatology ; 114(4): 303-306, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30011398

RESUMO

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.


Assuntos
Sistema ABO de Grupos Sanguíneos/imunologia , Incompatibilidade de Grupos Sanguíneos/sangue , Índices de Eritrócitos , Sangue Fetal/citologia , Transfusão Feto-Materna/diagnóstico , Adulto , Feminino , Transfusão Feto-Materna/sangue , Hemoglobinas/análise , Humanos , Gravidez
9.
Hemoglobin ; 42(2): 138-140, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29745271

RESUMO

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.


Assuntos
Hemoglobina Fetal/análise , Transfusão Feto-Materna , Adulto , Eritrócitos/química , Feminino , Sangue Fetal/citologia , Humanos , Recém-Nascido , Masculino , Gravidez
11.
Presse Med ; 46(12 Pt 2): e296-e305, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29174474

RESUMO

While most common symptom of impairment of iron homeostasis is iron deficiency anemia, some hematological disorders are associated with iron overload (IO). These disorders are related mainly to chronic severe hemolytic anemia, where red blood cells (RBC) or their precursors are destroyed prematurely (hemolyzed), leading to anemia that cannot be compensated by increased production of new RBC. In such cases, IO is mainly due to repeated RBC transfusions and/or increased uptake of iron in the gastrointestinal tract. Normally, iron is present in the plasma and in the cells bound to compounds that render it redox inactive. Iron overload leaves a fraction of the iron free (labile iron pool) and redox active, leading to the generation of excess free radicals such as the reactive oxygen species. This condition upsets the cellular redox balance between oxidants and antioxidants, leading to oxidative stress. The free radicals bind to various cellular components, thereby becoming toxic to vital organs. Oxidative stress may also affect blood cells, such as RBC, platelets and neutrophils, exacerbating the anemia, and causing recurrent infections and thrombotic events, respectively. The toxic effect of IO can be decreased by treating the patients with iron chelators that enter cells, bind free iron and remove it from the body through the urine and feces. Iron toxicity may be also ameliorated by treatment with anti-oxidants that scavenge free radicals and/or correct their damage. The use of iron chelators is widely accepted when started in young patients with severe chronic anemia, but is still debatable as a therapeutic modality for older patients suffering from IO due to myelodysplastic syndromes. It should be noted that in addition to preventing iron toxicity, some compounds with iron chelator activity may also benefit other aspects of hematological disorders. These aspects include stimulation of platelet production, inhibition of leukemic cell proliferation and induction of their differentiation. Compounds with such multiple activities may prove beneficial for at least some patients with leukemia and myelodysplastic syndromes.


Assuntos
Doenças Hematológicas/complicações , Sobrecarga de Ferro/etiologia , Anemia Hemolítica/complicações , Doenças Hematológicas/metabolismo , Humanos , Sobrecarga de Ferro/terapia , Estresse Oxidativo
12.
Hemoglobin ; 41(2): 144-146, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28670942

RESUMO

ß-Thalassemia (ß-thal) is a very common disease in the Palestinian population of the Gaza Strip. We studied their mutation frequency and clinical features. Thirteen different mutations were identified. The most common mutation was IVS-I-1 (G>A) (HBB: c.92+1G>A), which was prevalent in 31.5% of the thalassemia alleles studied. The IVS-I-110 (G>A) (HBB: c.93-21G>A) mutation was found in 25.0% of the alleles. Homozygotes for the IVS-I-1 mutation had higher mean hemoglobin (Hb) levels, required less blood transfusions, and lower transferrin saturation than the homozygotes for the IVS-I-110 mutation. This milder phenotype was, most likely, the result of the persistent production of Hb F; it was 9-fold higher in absolute terms (g/dL) and 7.7-fold higher in relative terms (percentage of total Hb). About half of our IVS-I-1 patients carried the XmnI polymorphism, which is known to be associated with elevated Hb F levels.


Assuntos
Hemoglobina Fetal/metabolismo , Frequência do Gene , Hemoglobinas , Mutação , Talassemia beta , Árabes , Feminino , Hemoglobina Fetal/genética , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Masculino , Oriente Médio , Talassemia beta/sangue , Talassemia beta/genética
13.
Crit Rev Oncol Hematol ; 113: 156-170, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28427505

RESUMO

Iron is an essential element for key cellular metabolic processes. However, transfusional iron overload (IOL) may result in significant cellular toxicity. IOL occurs in transfusion dependent hematologic malignancies (HM), may lead to pathological clinical outcomes, and IOL reduction may improve outcomes. In hematopoietic stem cell transplantation (SCT) for HM, IOL may have clinical importance; endpoints examined regarding an impact of IOL and IOL reduction include transplant-related mortality, organ function, infection, relapse risk, and survival. Here we review the clinical consequences of IOL and effects of IOL reduction before, during and following SCT for HM. IOL pathophysiology is discussed as well as available tests for IOL quantification including transfusion history, serum ferritin level, transferrin saturation, hepcidin, labile plasma iron and other parameters of iron-catalyzed oxygen free radicals, and organ IOL by imaging. Data-based recommendations for IOL measurement, monitoring and reduction before, during and following SCT for HM are made.


Assuntos
Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/fisiopatologia , Transfusão de Sangue , Neoplasias Hematológicas/complicações , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/complicações , Sobrecarga de Ferro/diagnóstico , Sobrecarga de Ferro/tratamento farmacológico
14.
Biomed Pharmacother ; 89: 578-590, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28258040

RESUMO

The formation of new blood vessels plays a crucial for the development and progression of pathophysiological changes associated with a variety of disorders, including carcinogenesis. Angiogenesis inhibitors (anti-angiogenics) are an important part of treatment for some types of cancer. Some natural products isolated from marine invertebrates have revealed antiangiogenic activities, which are diverse in structure and mechanisms of action. Many preclinical studies have generated new models for further modification and optimization of anti-angiogenic substances, and new information for mechanistic studies and new anti-cancer drug candidates for clinical practice. Moreover, in the last decade it has become apparent that galectins are important regulators of tumor angiogenesis, as well as microRNA. MicroRNAs have been validated to modulate endothelial cell migration or endothelial tube organization. In the present review we summarize the current knowledge regarding the role of marine-derived natural products, galectins and microRNAs in tumor angiogenesis.


Assuntos
Moduladores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Inibidores da Angiogênese/farmacologia , Animais , Produtos Biológicos/farmacologia , Galectinas/efeitos dos fármacos , Humanos , Toxinas Marinhas/farmacologia , MicroRNAs/efeitos dos fármacos
15.
Blood Cells Mol Dis ; 63: 45-51, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28126623

RESUMO

The JAK2V617F mutation that results in a hyper-activation of the JAK2 kinase in the erythropoietin pathway is a molecular marker for myeloproliferative neoplasms. Using allele-specific Real-Time PCR, we detected the mutation in the blood of 17.3% (17/98) of normal donors; the mutant allele burden was, however, very low (<0.01% compared to >1% in polycythemia vera). It was much higher in differentiated blood cells in the peripheral blood than in undifferentiated CD34+ cells. Erythropoietin-stimulated differentiation of normal CD34+ cells in liquid culture increased the mutation frequency by 3.34-fold. When progenitors from 9 normal donors were grown in erythropoietin-stimulated semi-solid cultures, the mutation was found in 8.69% of the colonies, but only in <3% of the JAK2 alleles in each positive colony, suggesting that the mutation occurred only in a few cells per colony. In mouse erythroleukemia cells carrying human JAK2 DNA, wild-type or JAK2V617F, the frequencies of mutations from JAK2 wild-type to JAK2V617F and vice versa increased following erythroid differentiation. These results suggest that the mutation occurs and accumulates during differentiation. We hypothesize that genetic stability, which relies on DNA repair, is efficient in normal hematopoietic stem cells but is downgraded in differentiating cells, rendering them susceptible to mutations, including JAK2V617F.


Assuntos
Diferenciação Celular/genética , Janus Quinase 2/genética , Taxa de Mutação , Animais , Doadores de Sangue , Células Precursoras Eritroides/citologia , Eritropoetina/farmacologia , Humanos , Camundongos , Mutação , Transtornos Mieloproliferativos
16.
F1000Res ; 6: 2156, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29333256

RESUMO

Thalassemia (thal) is an autosomal recessive, hereditary, chronic hemolytic anemia due to a partial or complete deficiency in the synthesis of α-globin chains (α-thal) or ß-globin chains (ß-thal) that compose the major adult hemoglobin (α 2ß 2). It is caused by one or more mutations in the corresponding genes. The unpaired globin chains are unstable; they precipitate intracellularly, resulting in hemolysis, premature destruction of red blood cell [RBC] precursors in the bone marrow, and a short life-span of mature RBCs in the circulation. The state of anemia is treated by frequent RBC transfusions. This therapy results in the accumulation of iron (iron overload), a condition that is exacerbated by the breakdown products of hemoglobin (heme and iron) and the increased iron uptake for the chronic accelerated, but ineffective, RBC production. Iron catalyzes the generation of reactive oxygen species, which in excess are toxic, causing damage to vital organs such as the heart and liver and the endocrine system. Herein, we review recent findings regarding the pathophysiology underlying the major symptoms of ß-thal and potential therapeutic modalities for the amelioration of its complications, as well as new modalities that may provide a cure for the disease.

17.
Eur J Pharm Sci ; 91: 91-7, 2016 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-27283485

RESUMO

Anemia is a major cause of morbidity and mortality worldwide resulting from a wide variety of pathological conditions. In severe cases it is treated by blood transfusions or injection of erythroid stimulating agents, e.g., erythropoietin (Epo), which can be associated with serious adverse effects. Therefore, there is a need to develop new treatment modalities. We recently reported that treatment of erythroleukemic cells with the novel the bi-functional prodrugs of 5-aminolevulinic acid (ALA) and butyric acid (BA), AN233 and AN908, enhanced hemoglobin (Hb) synthesis to a substantially higher level than did ALA and BA individually or their mixture. Herein, we describe that these prodrugs when given orally to mice induced histone deacetylase inhibition in the kidneys, bone marrow and spleen, thus, indicating good penetrability to the tissues. In mice where anemia was chemically induced, treatment with the prodrugs increased the Hb, the number of red blood cells (RBCs) and the percentage of reticulocytes to normal levels. The prodrugs had no adverse effects even after repeated treatment at 100-200mg/kg for 50days. The lack of increased levels of Epo in the blood of mice that were treated with the prodrugs suggests that AN233 and AN908 affected the Hb and RBC levels in an Epo-independent manner. Taken together with our previous studies, we propose that the prodrugs increase globin expression by BA inhibition of histone deacetylase and elevation heme synthesis by ALA. These results support an Epo-independent approach for treating anemia with these prodrugs.


Assuntos
Anemia/tratamento farmacológico , Inibidores de Histona Desacetilases/uso terapêutico , Ácidos Levulínicos/uso terapêutico , Pró-Fármacos/uso terapêutico , Acetilação/efeitos dos fármacos , Ácido Aminolevulínico/metabolismo , Anemia/sangue , Anemia/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Ácido Butírico/metabolismo , Contagem de Eritrócitos , Eritropoese/efeitos dos fármacos , Eritropoetina/sangue , Eritropoetina/farmacologia , Hemoglobinas/análise , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases , Histonas/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Ácidos Levulínicos/farmacologia , Masculino , Camundongos Endogâmicos BALB C , Pró-Fármacos/farmacologia , Baço/efeitos dos fármacos , Baço/metabolismo
18.
Radiat Res ; 184(3): 304-13, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26291738

RESUMO

Exposure to ionizing radiation causes cellular damage, which can lead to premature cell death or accumulation of somatic mutations, resulting in malignancy. The damage is mediated in part by free radicals, particularly reactive oxygen species. Fermented papaya preparation (FPP), a product of yeast fermentation of Carica papaya Linn, has been shown to act as an antioxidant. In this study, we investigated the potential of FPP to prevent radiation-induced damage. FPP (0-100 µg/ml) was added to cultured human foreskin fibroblasts and myeloid leukemia (HL-60) cells either before or after irradiation (0-18 Gy). After 1-3 days, the cells were assayed for: intracellular labile iron, measured by staining with calcein; reactive oxygen species generation, measured with dichlorofluorescein diacetate; apoptosis, determined by phosphatidylserine exposure; membrane damage, determined by propidium iodide uptake; and cell survival, determined by a cell proliferation assay. DNA damage was estimated by measuring 8-oxoguanine, a parameter of DNA oxidation, using a fluorescent-specific probe and by the comet assay. These parameters were also assayed in bone marrow cells of mice treated with FPP (by adding it to the drinking water) either before or after irradiation. Somatic mutation accumulation was determined in their peripheral red blood cells, and their survival was monitored. FPP significantly reduced the measured radiation-induced cytotoxic parameters. These findings suggest that FPP might serve as a radioprotector, and its effect on DNA damage and mutagenicity might reduce the long-term effects of radiation, such as primary and secondary malignancy.


Assuntos
Carica , Fermentação , Protetores contra Radiação/farmacologia , Animais , Dano ao DNA , Feminino , Células HL-60 , Humanos , Camundongos , Camundongos Endogâmicos C3H , Mutação
19.
Free Radic Biol Med ; 88(Pt A): 63-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25937178

RESUMO

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.


Assuntos
Anemia Hemolítica Autoimune/fisiopatologia , Antioxidantes/farmacologia , Ativação do Complemento , Hemoglobinúria Paroxística/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , Anemia Hemolítica Autoimune/tratamento farmacológico , Proteínas do Sistema Complemento/metabolismo , Hemoglobinúria Paroxística/tratamento farmacológico , Humanos
20.
Eur J Pharmacol ; 752: 84-91, 2015 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-25701725

RESUMO

Several investigations have demonstrated a mild clinical status in patients with ß-globin disorders and congenital high persistence of foetal haemoglobin. This can be mimicked by a pharmacological increase of foetal γ-globin genes expression and foetal haemoglobin production. Our goal was to apply a multistep assay including few screening methods (benzidine staining, RT-PCR and HPLC analyses) and erythroid cellular model systems (the K562 cell line and erythroid precursors collected from peripheral blood) to select erythroid differentiation agents with foetal haemoglobin inducing potential. With this methodology, we have identified a butyric acid derivative, namely the 4174 cyclopropanecarboxylic acid compound, able to induce erythroid differentiation without antiproliferative effect in K562 cells and increase of γ-globin gene expression in erythroid precursor cells. The results are relevant for pharmacological treatments of haemoglobinopathies, including ß-thalassaemia and sickle cell anaemia.


Assuntos
Ácido Butírico/química , Ácido Butírico/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Hemoglobina Fetal/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Patentes como Assunto , Talassemia beta/genética , Talassemia beta/patologia , gama-Globinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA