Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Br J Haematol ; 160(3): 399-403, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23216540

RESUMEN

This exploratory study assessed apoptosis in peripheral blood leucocytes (PBL) from ß-thalassaemia patients receiving chronic transfusions and chelation therapy (deferasirox or deferoxamine) at baseline, 1, 6, and 12 months. At baseline, thalassaemic PBLs presented 50% greater levels of Bax (BAX), 75% higher caspase-3/7, 48% higher caspase-8 and 88% higher caspase-9 activities and 428% more nucleosomal DNA fragmentation than control subjects. Only neutrophils correlated significantly with apoptotic markers. Previously, we showed that over the treatment year, hepatic iron declined; we now show that the ratio of Bax/Bcl-2 (BCL2), (-27·3%/year), and caspase-9 activity (-13·3%/year) declined in both treatment groups, suggesting that chelation decreases body iron and indicators of PBL apoptosis.


Asunto(s)
Apoptosis , Leucocitos/metabolismo , Talasemia beta/metabolismo , Adolescente , Adulto , Transfusión Sanguínea , Caspasas/metabolismo , Terapia por Quelación , Niño , Preescolar , Fragmentación del ADN , Femenino , Humanos , Masculino , Adulto Joven , Proteína X Asociada a bcl-2/metabolismo , Talasemia beta/terapia
2.
Blood ; 117(4): 1379-89, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21059897

RESUMEN

In hereditary hemochromatosis, mutations in HFE lead to iron overload through abnormally low levels of hepcidin. In addition, HFE potentially modulates cellular iron uptake by interacting with transferrin receptor, a crucial protein during erythropoiesis. However, the role of HFE in this process was never explored. We hypothesize that HFE modulates erythropoiesis by affecting dietary iron absorption and erythroid iron intake. To investigate this, we used Hfe-KO mice in conditions of altered dietary iron and erythropoiesis. We show that Hfe-KO mice can overcome phlebotomy-induced anemia more rapidly than wild-type mice (even when iron loaded). Second, we evaluated mice combining the hemochromatosis and ß-thalassemia phenotypes. Our results suggest that lack of Hfe is advantageous in conditions of increased erythropoietic activity because of augmented iron mobilization driven by deficient hepcidin response. Lastly, we demonstrate that Hfe is expressed in erythroid cells and impairs iron uptake, whereas its absence exclusively from the hematopoietic compartment is sufficient to accelerate recovery from phlebotomy. In summary, we demonstrate that Hfe influences erythropoiesis by 2 distinct mechanisms: limiting hepcidin expression under conditions of simultaneous iron overload and stress erythropoiesis, and impairing transferrin-bound iron uptake by erythroid cells. Moreover, our results provide novel suggestions to improve the treatment of hemochromatosis.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyesis/genética , Antígenos de Histocompatibilidad Clase I/genética , Hierro/metabolismo , Proteínas de la Membrana/genética , Envejecimiento/sangre , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteína de la Hemocromatosis , Hepcidinas , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase I/fisiología , Homeostasis/genética , Homeostasis/fisiología , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Transferrina/metabolismo , Regulación hacia Arriba/genética , Regulación hacia Arriba/fisiología
3.
Haematologica ; 98(1): 129-35, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22875626

RESUMEN

Patients with ß-thalassemia require iron chelation therapy to protect against progressive iron overload and non-transferrin-bound iron. Some patients fail to respond adequately to deferoxamine and deferasirox monotherapy while others have side effects which limit their use of these drugs. Since combining deferiprone and deferoxamine has an additive effect, placing all patients into net negative iron balance, we investigated the possibility that combining deferasirox and deferoxamine would lead to similar results. We conducted 34-day metabolic iron balance studies in six patients in whom the relative effectiveness of deferasirox (30 mg/kg/day) and deferoxamine (40 mg/kg/day) was compared, alone and in combination. Patients consumed fixed low-iron diets; daily urinary and stool iron excretion were determined by atomic absorption. Red blood cell transfusions were given prior to each drug treatment to minimize the effects of ineffective erythropoiesis. Serial safety measures, hematologic parameters, serum chemistries, ferritin levels and urinalyses were determined. All patients were in negative iron balance when treated with deferoxamine alone while four of six patients remained in positive balance when deferasirox monotherapy was evaluated. Daily use of both drugs had a synergistic effect in two patients and an additive effect in three others. Five of six patients would be in negative iron balance if they used the combination of drugs just 3 days a week. No significant or drug-related changes were observed in the blood work-ups or urinalyses performed. We conclude that supplementing the daily use of deferasirox with 2 - 3 days of deferoxamine therapy would place all patients into net negative iron balance thereby providing a convenient way to tailor chelation therapy to the individual needs of each patient.


Asunto(s)
Benzoatos/administración & dosificación , Deferoxamina/administración & dosificación , Triazoles/administración & dosificación , Talasemia beta/tratamiento farmacológico , Talasemia beta/metabolismo , Adulto , Deferasirox , Quimioterapia Combinada , Femenino , Humanos , Hierro/sangre , Hierro/orina , Quelantes del Hierro/administración & dosificación , Masculino
4.
Blood ; 116(14): 2582-9, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20554970

RESUMEN

Osteoporosis is a frequent problem in disorders characterized by iron overload, such as the thalassemias and hereditary hemochromatosis. The exact role of iron in the development of osteoporosis in these disorders is not established. To define the effect of iron excess in bone, we generated an iron-overloaded mouse by injecting iron dextran at 2 doses into C57/BL6 mice for 2 months. Compared with the placebo group, iron-overloaded mice exhibited dose-dependent increased tissue iron content, changes in bone composition, and trabecular and cortical thinning of bone accompanied by increased bone resorption. Iron-overloaded mice had increased reactive oxygen species and elevated serum tumor necrosis factor-α and interleukin-6 concentrations that correlated with severity of iron overload. Treatment of iron-overloaded mice with the antioxidant N-acetyl-L-cysteine prevented the development of trabecular but not cortical bone abnormalities. This is the first study to demonstrate that iron overload in mice results in increased bone resorption and oxidative stress, leading to changes in bone microarchitecture and material properties and thus bone loss.


Asunto(s)
Sobrecarga de Hierro/complicaciones , Osteoporosis/etiología , Estrés Oxidativo , Acetilcisteína/uso terapéutico , Animales , Antioxidantes/uso terapéutico , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Sobrecarga de Hierro/inducido químicamente , Sobrecarga de Hierro/metabolismo , Complejo Hierro-Dextran , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis/patología
5.
Blood Cells Mol Dis ; 47(3): 166-75, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21843958

RESUMEN

The effectiveness of deferoxamine (DFO), deferiprone (DFP), or deferasirox (DFX) in thalassemia major was assessed. Outcomes were reported as means±SD, mean differences with 95% CI, or standardized mean differences. Statistical heterogeneity was tested using χ2 (Q) and I2. Sources of bias and Grading of Recommendations Assessment, Development and Evaluation system (GRADE) were considered. Overall, 1520 patients were included. Only 7.4% of trials were free of bias. Overall measurements suggest low trial quality (GRADE). The meta-analysis suggests lower final liver iron concentrations during associated versus monotherapy treatment (p<0.0001), increases in serum ferritin levels during DFX 5, 10, and 20 mg/kg versus DFO-treated groups (p<0.00001, p<0.00001, and p=0.002, respectively), but no statistically significant difference during DFX 30 mg/kg versus DFO (p=0.70), no statistically significant variations in heart T2* signal during associated or sequential versus mono-therapy treatment (p=0.46 and p=0.14, respectively), increases in urinary iron excretion during associated or sequential versus monotherapy treatment (p=0.008 and p=0.02, respectively), and improved ejection fraction during associated or sequential versus monotherapy treatment (p=0.01 and p<0.00001, respectively). These findings do not support any specific chelation treatment. The literature shows risks of bias, and additional larger and longer trials are needed.


Asunto(s)
Benzoatos/administración & dosificación , Deferoxamina/administración & dosificación , Quelantes del Hierro/administración & dosificación , Piridonas/administración & dosificación , Sideróforos/administración & dosificación , Triazoles/administración & dosificación , Talasemia beta , Terapia por Quelación/estadística & datos numéricos , Deferasirox , Deferiprona , Quimioterapia Combinada , Ferritinas/sangre , Humanos , Hierro , Hígado/metabolismo , MEDLINE , Miocardio/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento , Función Ventricular/fisiología , Talasemia beta/sangre , Talasemia beta/tratamiento farmacológico
6.
Blood Cells Mol Dis ; 45(2): 136-9, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20678715

RESUMEN

Myocardial iron overload is the leading cause of death in patients with beta-thalassemia major. An intensification monotherapy with deferoxamine (DFO) as well as a combination therapy with DFO and deferiprone (DFP) reduces myocardial iron and improves cardiac function. However, the prognosis for thalassemia major patients with established cardiac disease switched from DFO monotherapy to combined DFP/DFO chelation is unknown. Twenty-eight thalassemia major patients with cardiac disease were enrolled in a prospective study lasting 42+/-6 months. Fifteen (9 high-ferritin and 6 low-ferritin) were placed on DFP/DFO (DFP, 75 mg/kg t.i.d.; DFO, 40-50mg/kg over 8-12h at night 5-7 days/week), while 13 (5 high- and 8 low-ferritin) received DFO alone. No cardiac events were observed among high-ferritin patients on combination therapy, whereas 4 cardiac events (p=0.0049), including three deaths, occurred in high-ferritin patients on DFO monotherapy. These findings demonstrate that in thalassemia major patients with well-established cardiac disease combined iron-chelation therapy with DFP/DFO is superior to DFO monotherapy.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Deferoxamina/uso terapéutico , Quimioterapia Combinada , Quelantes del Hierro/uso terapéutico , Piridonas/uso terapéutico , Talasemia beta/tratamiento farmacológico , Adulto , Cardiomiopatías/inducido químicamente , Terapia por Quelación , Deferiprona , Femenino , Humanos , Hierro/sangre , Quelantes del Hierro/administración & dosificación , Sobrecarga de Hierro/tratamiento farmacológico , Masculino , Estudios Prospectivos , Tasa de Supervivencia , Talasemia beta/mortalidad
7.
Blood ; 112(3): 875-85, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18480424

RESUMEN

In beta-thalassemia, the mechanism driving ineffective erythropoiesis (IE) is insufficiently understood. We analyzed mice affected by beta-thalassemia and observed, unexpectedly, a relatively small increase in apoptosis of their erythroid cells compared with healthy mice. Therefore, we sought to determine whether IE could also be characterized by limited erythroid cell differentiation. In thalassemic mice, we observed that a greater than normal percentage of erythroid cells was in S-phase, exhibiting an erythroblast-like morphology. Thalassemic cells were associated with expression of cell cycle-promoting genes such as EpoR, Jak2, Cyclin-A, Cdk2, and Ki-67 and the antiapoptotic protein Bcl-X(L). The cells also differentiated less than normal erythroid ones in vitro. To investigate whether Jak2 could be responsible for the limited cell differentiation, we administered a Jak2 inhibitor, TG101209, to healthy and thalassemic mice. Exposure to TG101209 dramatically decreased the spleen size but also affected anemia. Although our data do not exclude a role for apoptosis in IE, we propose that expansion of the erythroid pool followed by limited cell differentiation exacerbates IE in thalassemia. In addition, these results suggest that use of Jak2 inhibitors has the potential to profoundly change the management of this disorder.


Asunto(s)
Diferenciación Celular , Células Eritroides/patología , Eritropoyesis , Janus Quinasa 2/genética , Talasemia beta/sangre , Animales , Apoptosis , Quinasas Ciclina-Dependientes/genética , Janus Quinasa 2/antagonistas & inhibidores , Ratones , Bazo/patología
8.
Calcif Tissue Int ; 86(6): 484-94, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20449578

RESUMEN

Osteoporosis and fractures occur frequently in patients with beta-thalassemias, a group of congenital hemolytic anemias characterized by decreased synthesis of the beta chain of hemoglobin. In this study, we determined the bone abnormalities of the th3 thalassemia mouse, generated by deletion of the mouse beta-chain genes. The heterozygous th3/+ mouse has moderate anemia and serves as a model of beta-thalassemia intermedia, which represents the mild thalassemia phenotype. The th3/th3 mouse has lethal anemia and is a model of beta-thalassemia major, which is characterized by life-threatening anemia requiring regular transfusions to sustain life. Compared to controls, (1) microCT of trabecular bone showed decreased bone volume fraction, number of trabeculae, and trabecular thickness in both th3/+ and th3/th3 (P < 0.05); (2) cortical bone analysis showed thinner cortices and increased marrow area in th3/+ (P < 0.05); (3) microCT abnormalities in th3/+ mice were present by 2 months and did not worsen with age; (4) histomorphometry was significant for decreased bone formation and resorption in both th3/+ and th3/th3, and expression of cathepsin K and osteocalcin from bone of both th3/+ and th3/th3 animals was reduced (P < 0.05); (5) biomechanics showed reduced maximum load, maximum moment, and structural stiffness in both th3/+ and th3/th3 (P < 0.01). In conclusion, the th3 mouse model of thalassemia manifests bone changes reminiscent of those in humans and can be used for further bone studies in thalassemia. Bone changes are associated with decreased bone turnover and develop early during the period of bone accrual.


Asunto(s)
Remodelación Ósea/fisiología , Osteoporosis/patología , Talasemia/complicaciones , Animales , Fenómenos Biomecánicos , Huesos , Eritropoyesis , Ratones , Ratones Endogámicos C57BL , Osteoporosis/etiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectroscopía Infrarroja por Transformada de Fourier , Talasemia/genética , Tomografía Computarizada por Rayos X
9.
Br J Haematol ; 146(5): 546-56, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19604241

RESUMEN

This study aimed to determine differences in the rates of growth, endocrine- and calcium-related abnormalities in the various thalassemia syndromes in North America treated with current therapies. Medical history, physical examinations and blood and urine collections were obtained from patients with all thalassemia syndromes age 6 years and older in the Thalassemia Clinical Research Network. 361 subjects, 49% male, mean age 23.2 years (range 6.1-75 years) were studied. Approximately 25% of children and adults, regardless of the thalassemia syndrome, had short stature. Overall growth in children was mildly affected. Final height was close to midparental height (z = -0.73 +/- 1.24). Patients with beta thalassemia major (TM) had higher rates of hypogonadism, multiple endocrinopathies, worse hyperglycaemia, subclinical hypoparathyroidism and hypercalciuria. Hypogonadism remained the most frequent endocrinopathy and was frequently under-treated. 12.8% of the subjects had 25 vitamin D concentrations less than 27 nmol/l and 82% less than 75 nmol/l, regardless of the thalassemia syndrome. Adolescents had lower 25 vitamin D levels than children and adults. Compared to patients with other thalassemia syndromes, those with beta TM suffered from higher rates of multiple endocrinopathies, abnormal calcium metabolism and hypercalciuria. Vitamin D abnormalities were high among adolescents.


Asunto(s)
Trastornos del Crecimiento/complicaciones , Talasemia/complicaciones , Talasemia/epidemiología , Deficiencia de Vitamina D/complicaciones , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Calcio/sangre , Niño , Estudios Transversales , Femenino , Ferritinas/sangre , Trastornos del Crecimiento/sangre , Trastornos del Crecimiento/fisiopatología , Hormona del Crecimiento/deficiencia , Humanos , Hipogonadismo/sangre , Hipogonadismo/complicaciones , Hipogonadismo/fisiopatología , Modelos Lineales , Modelos Logísticos , Masculino , Persona de Mediana Edad , Hormona Paratiroidea/sangre , Prevalencia , Somatomedinas/análisis , Talasemia/metabolismo , Talasemia/fisiopatología , Estados Unidos/epidemiología , Deficiencia de Vitamina D/sangre , Deficiencia de Vitamina D/fisiopatología , Adulto Joven
11.
Ann N Y Acad Sci ; 1054: 417-22, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16339690

RESUMEN

To develop new treatments for beta-thalassemia, it is essential to identify the genes involved in the relevant pathophysiological processes. Iron metabolism in thalassemia mice being investigated, focusing on the expression of a gene called hepcidin (Hamp), which is expressed in the liver and whose product (Hamp) is secreted into the bloodstream. In mice, iron overload leads to overexpression of Hamp, while Hamp-knockout mice suffer from hemochromatosis. The aim of this study is to investigate Hamp in the mouse model of beta-thalassemia and to address the potential gene transfer of Hamp to prevent abnormal iron absorption.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/fisiología , Hemocromatosis/genética , Absorción Intestinal/fisiología , Sobrecarga de Hierro/etiología , Hierro/farmacocinética , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/genética , Carcinoma Hepatocelular/patología , Línea Celular/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Vectores Genéticos/uso terapéutico , Hemocromatosis/metabolismo , Hepatocitos/metabolismo , Hepcidinas , Humanos , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Lentivirus/genética , Hígado/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Células 3T3 NIH , Transducción Genética , Talasemia beta/metabolismo , Talasemia beta/terapia
12.
Nat Med ; 19(4): 437-45, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23502961

RESUMEN

Regulation of erythropoiesis is achieved by the integration of distinct signals. Among them, macrophages are emerging as erythropoietin-complementary regulators of erythroid development, particularly under stress conditions. We investigated the contribution of macrophages to physiological and pathological conditions of enhanced erythropoiesis. We used mouse models of induced anemia, polycythemia vera and ß-thalassemia in which macrophages were chemically depleted. Our data indicate that macrophages contribute decisively to recovery from induced anemia, as well as the pathological progression of polycythemia vera and ß-thalassemia, by modulating erythroid proliferation and differentiation. We validated these observations in primary human cultures, showing a direct impact of macrophages on the proliferation and enucleation of erythroblasts from healthy individuals and patients with polycythemia vera or ß-thalassemia. The contribution of macrophages to stress and pathological erythropoiesis, which we have termed stress erythropoiesis macrophage-supporting activity, may have therapeutic implications.


Asunto(s)
Eritropoyesis/fisiología , Macrófagos/fisiología , Policitemia Vera/fisiopatología , Talasemia beta/fisiopatología , Animales , Ácido Clodrónico/farmacología , Modelos Animales de Enfermedad , Recuento de Eritrocitos , Eritropoyesis/efectos de los fármacos , Femenino , Hematócrito , Hemoglobinas/análisis , Humanos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Reticulocitos/fisiología , Estrés Fisiológico/fisiología
13.
PLoS One ; 7(3): e32345, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22479321

RESUMEN

Preclinical and clinical studies demonstrate the feasibility of treating ß-thalassemia and Sickle Cell Disease (SCD) by lentiviral-mediated transfer of the human ß-globin gene. However, previous studies have not addressed whether the ability of lentiviral vectors to increase hemoglobin synthesis might vary in different patients.We generated lentiviral vectors carrying the human ß-globin gene with and without an ankyrin insulator and compared their ability to induce hemoglobin synthesis in vitro and in thalassemic mice. We found that insertion of an ankyrin insulator leads to higher, potentially therapeutic levels of human ß-globin through a novel mechanism that links the rate of transcription of the transgenic ß-globin mRNA during erythroid differentiation with polysomal binding and efficient translation, as reported here for the first time. We also established a preclinical assay to test the ability of this novel vector to synthesize adult hemoglobin in erythroid precursors and in CD34(+) cells isolated from patients affected by ß-thalassemia and SCD. Among the thalassemic patients, we identified a subset of specimens in which hemoglobin production can be achieved using fewer copies of the vector integrated than in others. In SCD specimens the treatment with AnkT9W ameliorates erythropoiesis by increasing adult hemoglobin (Hb A) and concurrently reducing the sickling tetramer (Hb S).Our results suggest two major findings. First, we discovered that for the purpose of expressing the ß-globin gene the ankyrin element is particularly suitable. Second, our analysis of a large group of specimens from ß-thalassemic and SCD patients indicates that clinical trials could benefit from a simple test to predict the relationship between the number of vector copies integrated and the total amount of hemoglobin produced in the erythroid cells of prospective patients. This approach would provide vital information to select the best candidates for these clinical trials, before patients undergo myeloablation and bone marrow transplant.


Asunto(s)
Anemia de Células Falciformes/terapia , Terapia Genética/métodos , Hemoglobinas/metabolismo , Talasemia beta/terapia , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/genética , Animales , Ancirinas/genética , Antígenos CD34/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Células Precursoras Eritroides/metabolismo , Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Hemoglobinas/genética , Humanos , Elementos Aisladores/genética , Lentivirus/genética , Ratones , Datos de Secuencia Molecular , Mutación , Células 3T3 NIH , Globinas beta/genética , Talasemia beta/sangre , Talasemia beta/genética
14.
Hematol Oncol Clin North Am ; 24(6): 1089-107, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21075282

RESUMEN

ß-Thalassemia is a genetic disorder caused by mutations in the ß-globin gene and characterized by chronic anemia caused by ineffective erythropoiesis, and accompanied by a variety of serious secondary complications such as extramedullary hematopoiesis, splenomegaly, and iron overload. In the past few years, numerous studies have shown that such secondary disease conditions have a genetic basis caused by the abnormal expression of genes with a role in controlling erythropoiesis and iron metabolism. In this article, the most recent discoveries related to the mechanism(s) responsible for anemia/ineffective erythropoiesis and iron overload are discussed in detail. Particular attention is paid to the pathway(s) controlling the expression of hepcidin, which is the main regulator of iron metabolism, and the Epo/EpoR/Jak2/Stat5 signaling pathway, which regulates erythropoiesis. Better understanding of how these pathways function and are altered in ß-thalassemia has revealed several possibilities for development of new therapeutic approaches to treat of the complications of this disease.


Asunto(s)
Anemia/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Eritropoyesis , Talasemia beta/metabolismo , Animales , Eritropoyetina/metabolismo , Hepcidinas , Humanos , Hierro/metabolismo , Sobrecarga de Hierro , Receptores de Eritropoyetina/metabolismo
15.
Ann N Y Acad Sci ; 1202: 24-30, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20712768

RESUMEN

beta-thalassemia is a disease associated with decreased beta-globin production leading to anemia, ineffective erythropoiesis, and iron overload. New mechanisms associated with modulation of erythropoiesis and iron metabolism have recently been discovered in thalassemic mice, improving our understanding of the pathophysiology of this disease. These discoveries have the potential to be translated into clinically-relevant therapeutic options to reduce ineffective erythropoiesis and iron overload. A new generation of therapies based on limiting ineffective erythropoiesis, iron absorption, and the correction of iron maldistribution could be on the way, possibly complementing and improving the current standard of patient care.


Asunto(s)
Modelos Animales de Enfermedad , Eritropoyesis/fisiología , Hierro/metabolismo , Talasemia beta/metabolismo , Talasemia beta/fisiopatología , Animales , Antibacterianos/metabolismo , Péptidos Catiónicos Antimicrobianos/metabolismo , Hepcidinas , Humanos , Sobrecarga de Hierro/metabolismo , Ratones
16.
Ann N Y Acad Sci ; 1202: 79-86, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20712777

RESUMEN

Patients with thalassemia major accumulate body iron over time as a consequence of continuous red blood cell transfusions which cause hepatic, endocrine, and cardiac complications. Despite the availability of three iron chelators, some patients fail to respond adequately to monotherapy with any of them. Combination therapy, consisting in the use of two chelators on the same day, has been introduced to increase the efficacy and to induce negative iron balance in patients with severe iron overload. Extensive long-term experience has shown that combined chelation with deferiprone and deferoxamine (DFO) rapidly reduces liver iron, serum ferritin, and myocardial siderosis, improves cardiac function, reverses and prevents endocrine complications, reduces cardiac mortality, and improves survival. Side effects, though significant, are manageable if properly monitored. Preliminary promising results have been obtained using combined chelation with deferasirox and DFO. As more drug combination regimes are evaluated, it should be possible to better tailor iron chelation to the needs of the patients, minimizing toxicity and maximizing efficacy throughout life.


Asunto(s)
Terapia por Quelación/métodos , Quelantes del Hierro/uso terapéutico , Talasemia beta/terapia , Deferiprona , Deferoxamina/efectos adversos , Deferoxamina/uso terapéutico , Combinación de Medicamentos , Ferritinas/sangre , Humanos , Hierro/metabolismo , Quelantes del Hierro/efectos adversos , Sobrecarga de Hierro/etiología , Piridonas/efectos adversos , Piridonas/uso terapéutico , Reacción a la Transfusión , Talasemia beta/complicaciones
17.
Ann N Y Acad Sci ; 1202: 221-5, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20712796

RESUMEN

Hepcidin (HAMP) negatively regulates iron absorption, degrading the iron exporter ferroportin at the level of enterocytes and macrophages. We showed that mice with beta-thalassemia intermedia (th3/+) have increased anemia and iron overload. However, their hepcidin expression is relatively low compared to their iron burden. We also showed that the iron metabolism gene Hfe is down-regulated in concert with hepcidin in th3/+ mice. These observations suggest that low hepcidin levels are responsible for abnormal iron absorption in thalassemic mice and that down-regulation of Hfe might be involved in the pathway that controls hepcidin synthesis in beta-thalassemia. Therefore, these studies suggest that increasing hepcidin and/or Hfe expression could be a strategy to reduces iron overload in these animals. The goal of this paper is to review recent findings that correlate hepcidin, Hfe, and iron metabolism in beta-thalassemia and to discuss potential novel therapeutic approaches based on these recent discoveries.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Sobrecarga de Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Talasemia beta/metabolismo , Anemia/etiología , Anemia/metabolismo , Animales , Antibacterianos/metabolismo , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Terapia Genética , Vectores Genéticos/genética , Proteína de la Hemocromatosis , Hepcidinas , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/uso terapéutico , Humanos , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/terapia , Proteínas de la Membrana/genética , Proteínas de la Membrana/uso terapéutico , Talasemia beta/complicaciones , Talasemia beta/genética , Talasemia beta/terapia
18.
J Clin Invest ; 120(12): 4466-77, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21099112

RESUMEN

Excessive iron absorption is one of the main features of ß-thalassemia and can lead to severe morbidity and mortality. Serial analyses of ß-thalassemic mice indicate that while hemoglobin levels decrease over time, the concentration of iron in the liver, spleen, and kidneys markedly increases. Iron overload is associated with low levels of hepcidin, a peptide that regulates iron metabolism by triggering degradation of ferroportin, an iron-transport protein localized on absorptive enterocytes as well as hepatocytes and macrophages. Patients with ß-thalassemia also have low hepcidin levels. These observations led us to hypothesize that more iron is absorbed in ß-thalassemia than is required for erythropoiesis and that increasing the concentration of hepcidin in the body of such patients might be therapeutic, limiting iron overload. Here we demonstrate that a moderate increase in expression of hepcidin in ß-thalassemic mice limits iron overload, decreases formation of insoluble membrane-bound globins and reactive oxygen species, and improves anemia. Mice with increased hepcidin expression also demonstrated an increase in the lifespan of their red cells, reversal of ineffective erythropoiesis and splenomegaly, and an increase in total hemoglobin levels. These data led us to suggest that therapeutics that could increase hepcidin levels or act as hepcidin agonists might help treat the abnormal iron absorption in individuals with ß-thalassemia and related disorders.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/uso terapéutico , Sobrecarga de Hierro/tratamiento farmacológico , Talasemia beta/tratamiento farmacológico , Animales , Péptidos Catiónicos Antimicrobianos/genética , Secuencia de Bases , Cartilla de ADN/genética , Modelos Animales de Enfermedad , Eritropoyesis/efectos de los fármacos , Expresión Génica , Hepcidinas , Humanos , Hierro/metabolismo , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/metabolismo , Hierro de la Dieta/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Proteínas Recombinantes/genética , Proteínas Recombinantes/uso terapéutico , Talasemia beta/sangre , Talasemia beta/metabolismo
19.
J Bone Miner Res ; 24(3): 543-57, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18505376

RESUMEN

Adults with beta thalassemia major frequently have low BMD, fractures, and bone pain. The purpose of this study was to determine the prevalence of low BMD, fractures, and bone pain in all thalassemia syndromes in childhood, adolescence, and adulthood, associations of BMD with fractures and bone pain, and etiology of bone disease in thalassemia. Patients of all thalassemia syndromes in the Thalassemia Clinical Research Network, > or =6 yr of age, with no preexisting medical condition affecting bone mass or requiring steroids, participated. We measured spine and femur BMD and whole body BMC by DXA and assessed vertebral abnormalities by morphometric X-ray absorptiometry (MXA). Medical history by interview and review of medical records, physical examinations, and blood and urine collections were performed. Three hundred sixty-one subjects, 49% male, with a mean age of 23.2 yr (range, 6.1-75 yr), were studied. Spine and femur BMD Z-scores < -2 occurred in 46% and 25% of participants, respectively. Greater age, lower weight, hypogonadism, and increased bone turnover were strong independent predictors of low bone mass regardless of thalassemia syndrome. Peak bone mass was suboptimal. Thirty-six percent of patients had a history of fractures, and 34% reported bone pain. BMD was negatively associated with fractures but not with bone pain. Nine percent of participants had uniformly decreased height of several vertebrae by MXA, which was associated with the use of iron chelator deferoxamine before 6 yr of age. In patients with thalassemia, low BMD and fractures occur frequently and independently of the particular syndrome. Peak bone mass is suboptimal. Low BMD is associated with hypogonadism, increased bone turnover, and an increased risk for fractures.


Asunto(s)
Enfermedades Óseas/complicaciones , Talasemia/complicaciones , Absorciometría de Fotón , Adolescente , Adulto , Distribución por Edad , Envejecimiento/patología , Biomarcadores/metabolismo , Densidad Ósea , Enfermedades Óseas/epidemiología , Enfermedades Óseas/fisiopatología , Remodelación Ósea , Huesos/anatomía & histología , Huesos/patología , Niño , Femenino , Humanos , Articulaciones/patología , Masculino , Tamaño de los Órganos , Dolor/complicaciones , Dolor/epidemiología , Prevalencia , Análisis de Regresión , Fracturas de la Columna Vertebral/complicaciones , Fracturas de la Columna Vertebral/epidemiología , Fracturas de la Columna Vertebral/patología , Fracturas de la Columna Vertebral/fisiopatología , Talasemia/epidemiología , Talasemia/fisiopatología , Estados Unidos/epidemiología
20.
Br J Haematol ; 138(3): 374-81, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17614825

RESUMEN

The most widely used drug for iron chelation is deferoxamine (DFO) mesylate. While effective in promoting iron excretion, it requires prolonged daily infusions, often resulting in poor compliance. A clinical trial was conducted using starch-conjugated DFO (S-DFO; 40SD02), a high-molecular-weight iron chelator possessing prolonged vascular retention. Single doses of S-DFO were infused intravenously into groups of four transfusion-dependent patients with beta-thalassaemia at doses of 150, 300, 600 and 900 mg/kg. Urinary iron excretion and various pharmacologic parameters were evaluated for 1 week and safety for 3 weeks. No drug-related effects were observed on clinical chemistries, haematological and coagulation parameters, urinalyses, vital signs or electrocardiograms. Drug-related adverse events were limited to four urticarial reactions, none requiring termination of the infusion. The drug stimulated clinically significant urinary iron excretion, with the highest dose (900 mg/kg) inducing excretion of 1.31 mg of iron/kg (range 0.79-1.90 mg/kg) over 1 week, with residual iron-binding capacity present in the plasma for over 6 d. In summary, treatment with S-DFO, administered weekly, has the potential to achieve iron balance in the poorly compliant patient.


Asunto(s)
Terapia por Quelación/métodos , Deferoxamina/uso terapéutico , Quelantes del Hierro/uso terapéutico , Talasemia beta/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Infusiones Intravenosas , Hierro/sangre , Hierro/orina , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Talasemia beta/sangre , Talasemia beta/orina
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda