Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
3.
Proc Natl Acad Sci U S A ; 113(13): 3428-35, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26929359

RESUMEN

Disrupted brain iron homeostasis is a common feature of neurodegenerative disease. To begin to understand how neuronal iron handling might be involved, we focused on dopaminergic neurons and asked how inactivation of transport proteins affected iron homeostasis in vivo in mice. Loss of the cellular iron exporter, ferroportin, had no apparent consequences. However, loss of transferrin receptor 1, involved in iron uptake, caused neuronal iron deficiency, age-progressive degeneration of a subset of dopaminergic neurons, and motor deficits. There was gradual depletion of dopaminergic projections in the striatum followed by death of dopaminergic neurons in the substantia nigra. Damaged mitochondria accumulated, and gene expression signatures indicated attempted axonal regeneration, a metabolic switch to glycolysis, oxidative stress, and the unfolded protein response. We demonstrate that loss of transferrin receptor 1, but not loss of ferroportin, can cause neurodegeneration in a subset of dopaminergic neurons in mice.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Hierro/metabolismo , Degeneración Nerviosa/etiología , Degeneración Nerviosa/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Neuronas Dopaminérgicas/patología , Femenino , Homeostasis , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Degeneración Nerviosa/patología , Receptores de Transferrina/deficiencia , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo
4.
Nat Genet ; 48(1): 74-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26642240

RESUMEN

Patients with a combined immunodeficiency characterized by normal numbers but impaired function of T and B cells had a homozygous p.Tyr20His substitution in transferrin receptor 1 (TfR1), encoded by TFRC. The substitution disrupts the TfR1 internalization motif, resulting in defective receptor endocytosis and markedly increased TfR1 expression on the cell surface. Iron citrate rescued the lymphocyte defects, and expression of wild-type but not mutant TfR1 rescued impaired transferrin uptake in patient-derived fibroblasts. Tfrc(Y20H/Y20H) mice recapitulated the immunological defects of patients. Despite the critical role of TfR1 in erythrocyte development and function, patients had only mild anemia and only slightly increased TfR1 expression in erythroid precursors. We show that STEAP3, a metalloreductase expressed in erythroblasts, associates with TfR1 and partially rescues transferrin uptake in patient-derived fibroblasts, suggesting that STEAP3 may provide an accessory TfR1 endocytosis signal that spares patients from severe anemia. These findings demonstrate the importance of TfR1 in adaptive immunity.


Asunto(s)
Antígenos CD/genética , Antígenos CD/inmunología , Síndromes de Inmunodeficiencia/genética , Mutación Missense , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología , Inmunidad Adaptativa/genética , Anemia/genética , Animales , Antígenos CD/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Endocitosis , Femenino , Fibroblastos/fisiología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Oxidorreductasas , Linaje , Receptores de Transferrina/metabolismo
5.
Cell Rep ; 13(3): 533-545, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26456827

RESUMEN

Both iron overload and iron deficiency have been associated with cardiomyopathy and heart failure, but cardiac iron utilization is incompletely understood. We hypothesized that the transferrin receptor (Tfr1) might play a role in cardiac iron uptake and used gene targeting to examine the role of Tfr1 in vivo. Surprisingly, we found that decreased iron, due to inactivation of Tfr1, was associated with severe cardiac consequences. Mice lacking Tfr1 in the heart died in the second week of life and had cardiomegaly, poor cardiac function, failure of mitochondrial respiration, and ineffective mitophagy. The phenotype could only be rescued by aggressive iron therapy, but it was ameliorated by administration of nicotinamide riboside, an NAD precursor. Our findings underscore the importance of both Tfr1 and iron in the heart, and may inform therapy for patients with heart failure.


Asunto(s)
Cardiomiopatías/genética , Miocardio/metabolismo , Receptores de Transferrina/genética , Animales , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/patología , Respiración de la Célula , Hierro/metabolismo , Ratones , Mitofagia , Miocardio/patología , Niacinamida/análogos & derivados , Niacinamida/uso terapéutico , Compuestos de Piridinio , Receptores de Transferrina/metabolismo
6.
Proc Natl Acad Sci U S A ; 112(37): 11714-9, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26324903

RESUMEN

Transferrin receptor 1 (Tfr1) facilitates cellular iron uptake through receptor-mediated endocytosis of iron-loaded transferrin. It is expressed in the intestinal epithelium but not involved in dietary iron absorption. To investigate its role, we inactivated the Tfr1 gene selectively in murine intestinal epithelial cells. The mutant mice had severe disruption of the epithelial barrier and early death. There was impaired proliferation of intestinal epithelial cell progenitors, aberrant lipid handling, increased mRNA expression of stem cell markers, and striking induction of many genes associated with epithelial-to-mesenchymal transition. Administration of parenteral iron did not improve the phenotype. Surprisingly, however, enforced expression of a mutant allele of Tfr1 that is unable to serve as a receptor for iron-loaded transferrin appeared to fully rescue most animals. Our results implicate Tfr1 in homeostatic maintenance of the intestinal epithelium, acting through a role that is independent of its iron-uptake function.


Asunto(s)
Homeostasis , Intestinos/embriología , Receptores de Transferrina/fisiología , Alelos , Animales , Encéfalo/embriología , Proliferación Celular , Transición Epitelial-Mesenquimal , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genotipo , Mucosa Intestinal/metabolismo , Hierro/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Fenotipo , Recombinación Genética , Células Madre/citología
7.
J Clin Invest ; 125(9): 3422-3, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26301806

RESUMEN

Iron-deficient individuals experience a loss of appetite that can be restored with iron supplementation. It has been proposed that iron influences the satiety hormone leptin; however, a direct link between iron and leptin has remained elusive. In this issue of the JCI, Gao and colleagues demonstrate an inverse relationship between adipocyte iron and leptin that is mediated by iron-dependent activation of cAMP-responsive element binding protein (CREB), the transcription factor that represses leptin transcription. Together, the results of this study provide a mechanistic connection between dietary iron and the appetite-regulating hormone leptin.


Asunto(s)
Adipocitos/metabolismo , Suplementos Dietéticos , Ingestión de Alimentos/efectos de los fármacos , Hemocromatosis/metabolismo , Hierro , Leptina/metabolismo , Animales
9.
EBioMedicine ; 2(11): 1705-17, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26870796

RESUMEN

Transferrin receptor (Tfr1) is ubiquitously expressed, but its roles in non-hematopoietic cells are incompletely understood. We used a tissue-specific conditional knockout strategy to ask whether skeletal muscle required Tfr1 for iron uptake. We found that iron assimilation via Tfr1 was critical for skeletal muscle metabolism, and that iron deficiency in muscle led to dramatic changes, not only in muscle, but also in adipose tissue and liver. Inactivation of Tfr1 incapacitated normal energy production in muscle, leading to growth arrest and a muted attempt to switch to fatty acid ß oxidation, using up fat stores. Starvation signals stimulated gluconeogenesis in the liver, but amino acid substrates became limiting and hypoglycemia ensued. Surprisingly, the liver was also iron deficient, and production of the iron regulatory hormone hepcidin was depressed. Our observations reveal a complex interaction between iron homeostasis and metabolism that has implications for metabolic and iron disorders.


Asunto(s)
Músculos/metabolismo , Receptores de Transferrina/deficiencia , Animales , Análisis por Conglomerados , Regulación de la Expresión Génica , Genes Letales , Deficiencias de Hierro , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/metabolismo , Trastornos del Metabolismo del Hierro/patología , Hígado/metabolismo , Metaboloma , Metabolómica/métodos , Ratones , Ratones Noqueados , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculos/patología , Fosforilación Oxidativa , Fenotipo , Receptores de Transferrina/genética
10.
Cell Metab ; 17(3): 319-28, 2013 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-23473029

RESUMEN

Transition metals are frequently used as cofactors for enzymes and oxygen-carrying proteins that take advantage of their propensity to gain and lose single electrons. Metals are particularly important in mitochondria, where they play essential roles in the production of ATP and detoxification of reactive oxygen species. At the same time, transition metals (particularly Fe and Cu) can promote the formation of harmful radicals, necessitating meticulous control of metal concentration and subcellular compartmentalization. We summarize our current understanding of Fe and Cu in mammalian mitochondrial biology and discuss human diseases associated with aberrations in mitochondrial metal homeostasis.


Asunto(s)
Hemo/biosíntesis , Homeostasis/fisiología , Proteínas Hierro-Azufre/biosíntesis , Hierro/metabolismo , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Modelos Biológicos , Humanos
11.
Cell Metab ; 16(4): 449-61, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23000401

RESUMEN

Reactive oxygen species (ROS) contribute to target-cell damage in inflammatory and iron-overload diseases. Little is known about iron transport regulation during inflammatory attack. Through a combination of in vitro and in vivo studies, we show that the proinflammatory cytokine IL-1ß induces divalent metal transporter 1 (DMT1) expression correlating with increased ß cell iron content and ROS production. Iron chelation and siRNA and genetic knockdown of DMT1 expression reduce cytokine-induced ROS formation and cell death. Glucose-stimulated insulin secretion in the absence of cytokines in Dmt1 knockout islets is defective, highlighting a physiological role of iron and ROS in the regulation of insulin secretion. Dmt1 knockout mice are protected against multiple low-dose streptozotocin and high-fat diet-induced glucose intolerance, models of type 1 and type 2 diabetes, respectively. Thus, ß cells become prone to ROS-mediated inflammatory damage via aberrant cellular iron metabolism, a finding with potential general cellular implications.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Transporte de Catión/metabolismo , Células Secretoras de Insulina/metabolismo , Interleucina-1beta/farmacología , Hierro/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Proteínas de Transporte de Catión/antagonistas & inhibidores , Proteínas de Transporte de Catión/genética , Diabetes Mellitus Experimental , Dieta Alta en Grasa , Intolerancia a la Glucosa , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Secretoras de Insulina/citología , Ratones , Ratones Noqueados , Modelos Biológicos , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transactivadores/genética , Transactivadores/metabolismo
12.
Haematologica ; 97(11): 1648-56, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22581006

RESUMEN

BACKGROUND: We and others have shown previously that over-expression of hepcidin antimicrobial peptide, independently of inflammation, induces several features of anemia of inflammation and chronic disease, including hypoferremia, sequestration of iron stores and iron-restricted erythropoiesis. Because the iron-restricted erythropoiesis evident in hepcidin transgenic mice differs from the normocytic, normochromic anemia most often observed in anemia of inflammation, we tested the hypothesis that chronic inflammation may contribute additional features to anemia of inflammation which continue to impair erythropoiesis following the acute phase of inflammation in which hepcidin is active. DESIGN AND METHODS: We compared erythropoiesis and iron handling in mice with turpentine-induced sterile abscesses with erythropoiesis and iron handling in hepcidin transgenic mice. We compared erythrocyte indices, expression of genes in the hepcidin regulatory pathway, tissue iron distribution, expression of heme and iron transport genes in splenic macrophages, the phenotype of erythroid maturation and chloromethyl dichlorodihydrofluorescein diacetate, acetyl ester fluorescence. RESULTS: Mice with sterile abscesses exhibited an intense, acute inflammatory phase followed by a mild to moderate chronic inflammatory phase. We found that erythrocytes in mice with sterile abscesses were normocytic and normochromic in contrast to those in hepcidin transgenic mice. We also observed that although hypoferremia resolved in the late phases of inflammation, erythropoiesis remained suppressed, with evidence of inefficient maturation of erythroid precursors in the bone marrow of mice with sterile abscesses. Finally, we observed increased oxidative stress in erythroid progenitors and circulating erythrocytes of mice with sterile abscesses which was not evident in hepcidin transgenic mice. CONCLUSIONS: Our results suggest that chronic inflammation inhibits late stages of erythroid production in the turpentine-induced sterile abscess model and induces features of impaired erythropoiesis which are distinct from those in hepcidin transgenic mice.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Células Precursoras Eritroides/metabolismo , Eritropoyesis , Animales , Péptidos Catiónicos Antimicrobianos/genética , Enfermedad Crónica , Células Precursoras Eritroides/patología , Hepcidinas , Inflamación/sangre , Inflamación/inducido químicamente , Inflamación/genética , Mediadores de Inflamación/sangre , Irritantes/efectos adversos , Irritantes/farmacología , Ratones , Ratones Transgénicos , Trementina/efectos adversos , Trementina/farmacología
14.
PLoS One ; 7(1): e29495, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253730

RESUMEN

BACKGROUND: The vestibular apparatus of the vertebrate inner ear uses three fluid-filled semicircular canals to sense angular acceleration of the head. Malformation of these canals disrupts the sense of balance and frequently causes circling behavior in mice. The Epistatic circler (Ecl) is a complex mutant derived from wildtype SWR/J and C57L/J mice. Ecl circling has been shown to result from the epistatic interaction of an SWR-derived locus on chromosome 14 and a C57L-derived locus on chromosome 4, but the causative genes have not been previously identified. METHODOLOGY/PRINCIPAL FINDINGS: We developed a mouse chromosome substitution strain (CSS-14) that carries an SWR/J chromosome 14 on a C57BL/10J genetic background and, like Ecl, exhibits circling behavior due to lateral semicircular canal malformation. We utilized CSS-14 to identify the chromosome 14 Ecl gene by positional cloning. Our candidate interval is located upstream of bone morphogenetic protein 4 (Bmp4) and contains an inner ear-specific, long non-coding RNA that we have designated Rubie (RNA upstream of Bmp4 expressed in inner ear). Rubie is spliced and polyadenylated, and is expressed in developing semicircular canals. However, we discovered that the SWR/J allele of Rubie is disrupted by an intronic endogenous retrovirus that causes aberrant splicing and premature polyadenylation of the transcript. Rubie lies in the conserved gene desert upstream of Bmp4, within a region previously shown to be important for inner ear expression of Bmp4. We found that the expression patterns of Bmp4 and Rubie are nearly identical in developing inner ears. CONCLUSIONS/SIGNIFICANCE: Based on these results and previous studies showing that Bmp4 is essential for proper vestibular development, we propose that Rubie is the gene mutated in Ecl mice, that it is involved in regulating inner ear expression of Bmp4, and that aberrant Bmp4 expression contributes to the Ecl phenotype.


Asunto(s)
Proteína Morfogenética Ósea 4/genética , Mutación/genética , ARN no Traducido/genética , Vestíbulo del Laberinto/anomalías , Animales , Conducta Animal , Cromosomas de los Mamíferos/genética , Clonación Molecular , Retrovirus Endógenos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Empalme del ARN/genética , ARN no Traducido/metabolismo , Canales Semicirculares/anomalías , Canales Semicirculares/embriología , Canales Semicirculares/metabolismo , Canales Semicirculares/patología , Especificidad de la Especie , Vestíbulo del Laberinto/metabolismo , Vestíbulo del Laberinto/patología
15.
Proc Natl Acad Sci U S A ; 109(5): E225-33, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22203997

RESUMEN

Global disruption of transient receptor potential-melastatin-like 7 (Trpm7) in mice results in embryonic lethality before embryonic day 7. Using tamoxifen-inducible disruption of Trpm7 and multiple Cre recombinase lines, we show that Trpm7 deletion before and during organogenesis results in severe tissue-specific developmental defects. We find that Trpm7 is essential for kidney development from metanephric mesenchyme but not ureteric bud. Disruption of neural crest Trpm7 at early stages results in loss of pigment cells and dorsal root ganglion neurons. In contrast, late disruption of brain-specific Trpm7 after embryonic day 10.5 does not alter normal brain development. We developed induced pluripotent stem cells and neural stem (NS) cells in which Trpm7 disruption could be induced. Trpm7(-/-) NS cells retained the capacities of self-renewal and differentiation into neurons and astrocytes. During in vitro differentiation of induced pluripotent stem cells to NS cells, Trpm7 disruption prevents the formation of the NS cell monolayer. The in vivo and in vitro results demonstrate a temporal requirement for the Trpm7 channel kinase during embryogenesis.


Asunto(s)
Desarrollo Embrionario/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Diferenciación Celular , Femenino , Proteínas de Filamentos Intermediarios/fisiología , Masculino , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/fisiología , Nestina , Células Madre Pluripotentes/citología
16.
Cell Metab ; 14(3): 281-2, 2011 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-21907132
17.
Blood ; 117(23): 6319-25, 2011 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-21493799

RESUMEN

Hepcidin, a hormone produced mainly by the liver, has been shown to inhibit both intestinal iron absorption and iron release from macrophages. Hemojuvelin, a glycophosphatidyl inositol-linked membrane protein, acts as a bone morphogenetic protein coreceptor to activate hepcidin expression through a SMAD signaling pathway in hepatocytes. In the present study, we show in mice that loss of hemojuvelin specifically in the liver leads to decreased liver hepcidin production and increased tissue and serum iron levels. Although it does not have any known function outside of the liver, hemojuvelin is expressed at very high levels in cardiac and skeletal muscle. To explore possible roles for hemojuvelin in skeletal muscle, we analyzed conditional knockout mice that lack muscle hemojuvelin. The mutant animals had no apparent phenotypic abnormalities. We found that systemic iron homeostasis and liver hepcidin expression were not affected by loss of hemojuvelin in skeletal muscle regardless of dietary iron content. We conclude that, in spite of its expression pattern, hemojuvelin is primarily important in the liver.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hierro/metabolismo , Hígado/metabolismo , Proteínas de la Membrana/biosíntesis , Proteínas Musculares/biosíntesis , Músculo Esquelético/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas Ligadas a GPI , Proteína de la Hemocromatosis , Hepcidinas , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Musculares/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo
18.
Blood ; 117(17): 4590-9, 2011 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-21355094

RESUMEN

The hereditary hemochromatosis protein HFE promotes the expression of hepcidin, a circulating hormone produced by the liver that inhibits dietary iron absorption and macrophage iron release. HFE mutations are associated with impaired hepatic bone morphogenetic protein (BMP)/SMAD signaling for hepcidin production. TMPRSS6, a transmembrane serine protease mutated in iron-refractory iron deficiency anemia, inhibits hepcidin expression by dampening BMP/SMAD signaling. In the present study, we used genetic approaches in mice to examine the relationship between Hfe and Tmprss6 in the regulation of systemic iron homeostasis. Heterozygous loss of Tmprss6 in Hfe(-/-) mice reduced systemic iron overload, whereas homozygous loss caused systemic iron deficiency and elevated hepatic expression of hepcidin and other Bmp/Smad target genes. In contrast, neither genetic loss of Hfe nor hepatic Hfe overexpression modulated the hepcidin elevation and systemic iron deficiency of Tmprss6(-/-) mice. These results indicate that genetic loss of Tmprss6 increases Bmp/Smad signaling in an Hfe-independent manner that can restore Bmp/Smad signaling in Hfe(-/-) mice. Furthermore, these results suggest that natural genetic variation in the human ortholog TMPRSS6 might modify the clinical penetrance of HFE-associated hereditary hemochromatosis, raising the possibility that pharmacologic inhibition of TMPRSS6 could attenuate iron loading in this disorder.


Asunto(s)
Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Hierro/metabolismo , Proteínas de la Membrana/genética , Serina Endopeptidasas/genética , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Femenino , Genotipo , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hemocromatosis/fisiopatología , Proteína de la Hemocromatosis , Hepcidinas , Heterocigoto , Antígenos de Histocompatibilidad Clase I/metabolismo , Homocigoto , Humanos , Hígado/fisiología , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Serina Endopeptidasas/metabolismo , Transducción de Señal/fisiología , Regulación hacia Arriba/fisiología
19.
Blood ; 117(2): 630-7, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20956801

RESUMEN

As a central regulator of iron metabolism, hepcidin inhibits dietary iron absorption and macrophage iron recycling. Its expression is regulated by multiple factors including iron availability and erythropoietic activity. To investigate the role of transferrin (Tf) in the regulation of hepcidin expression by these factors in vivo, we employed the hypotransferrinemic (hpx) mouse. These Tf-deficient mice have severe microcytic anemia, tissue iron overload, and hepcidin deficiency. To determine the relationship of Tf levels and erythropoiesis to hepcidin expression, we subjected hpx mutant and control mice to a number of experimental manipulations. Treatment of hpx mice with Tf injections corrected their anemia and restored hepcidin expression. To investigate the effect of erythropoiesis on hepcidin expression, we suppressed erythropoiesis with blood transfusions or myeloablation with chemotherapeutic drugs. Transfusion of hpx animals with wild-type red blood cells led to increased hepcidin expression, while hepcidin expression in myeloablated hpx mice increased only if Tf was administered postablation. These results suggest that hepcidin expression in hpx mice is regulated both by Tf-restricted erythropoiesis and by Tf through a mechanism independent of its role in erythropoiesis.


Asunto(s)
Anemia/fisiopatología , Péptidos Catiónicos Antimicrobianos/metabolismo , Eritropoyesis/fisiología , Transferrina/metabolismo , Anemia/metabolismo , Animales , Modelos Animales de Enfermedad , Hepcidinas , Humanos , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...