Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 421(3): 449-55, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22503983

RESUMEN

While intestinal cellular iron entry in vertebrates employs multiple routes including heme and non-heme routes, iron egress from these cells is exclusively channeled through the only known transporter, ferroportin. Reduced intestinal iron export in sex-linked anemia mice implicates hephaestin, a ferroxidase, in this process. Polarized cells are exposed to two distinct environments. Enterocytes contact the gut lumen via the apical surface of the cell, and through the basolateral surface, to the body. Previous studies indicate both local and systemic control of iron uptake. We hypothesized that differences in iron availability at the apical and/or basolateral surface may modulate iron uptake via cellular localization of hephaestin. We therefore characterized the localization of hephaestin in two models of polarized epithelial cell lines, MDCK and Caco2, with varying iron availability at the apical and basolateral surfaces. Our results indicate that hephaestin is expressed in a supra-nuclear compartment in non-polarized cells regardless of the iron status of the cells and in iron deficient and polarized cells. In polarized cells, we found that both apical (as FeSO(4)) and basolateral iron (as the ratio of apo-transferrin to holo-transferrin) affect mobilization of hephaestin from the supra-nuclear compartment. We find that the presence of apical iron is essential for relocalization of hephaestin to a cellular compartment in close proximity but not overlapping with the basolateral surface. Surface biotinylation studies indicate that hephaestin in the peri-basolateral location is accessible to the extra-cellular environment. These results support the hypothesis that hephaestin is involved in iron mobilization of iron from the intestine to circulation.


Asunto(s)
Mucosa Intestinal/metabolismo , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Secuencia de Aminoácidos , Animales , Biotinilación , Células CACO-2 , Polaridad Celular , Perros , Humanos , Datos de Secuencia Molecular , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
2.
J Nutr ; 140(10): 1728-35, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20685892

RESUMEN

We previously detected a membrane-bound, copper-containing oxidase that may be involved in iron efflux in BeWo cells, a human placental cell line. We have now identified a gene encoding a predicted multicopper ferroxidase (MCF) with a putative C-terminal membrane-spanning sequence and high sequence identity to hephaestin (Heph) and ceruloplasmin (Cp), the other known vertebrate MCF. Molecular modeling revealed conservation of all type I, II, and III copper-binding sites as well as a putative iron-binding site. Protein expression was observed in multiple diverse mouse tissues, including placenta and mammary gland, and the expression pattern was distinct from that of Cp and Heph. The protein possessed ferroxidase activity, and protein levels decreased in cellular copper deficiency. Knockdown with small interfering RNA in BeWo cells indicates that this gene represents the previously detected oxidase. We propose calling this new member of the MCF family "zyklopen."


Asunto(s)
Ceruloplasmina/química , Ceruloplasmina/genética , Cobre/análisis , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Ceruloplasmina/análisis , Cobre/metabolismo , Femenino , Expresión Génica , Humanos , Hierro/metabolismo , Glándulas Mamarias Animales/enzimología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Modelos Moleculares , Especificidad de Órganos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Fragmentos de Péptidos/química , Placenta/enzimología , Embarazo , ARN Interferente Pequeño/farmacología , Ratas , Homología de Secuencia
3.
J Cell Biochem ; 107(4): 803-8, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19452451

RESUMEN

Iron is transported across intestinal brush border cells into the circulation in at least two distinct steps. Iron can enter the enterocyte via the apical surface through several paths. However, iron egress from the basolateral side of enterocytes converges on a single export pathway requiring the iron transporter, ferroportin1, and hephaestin, a ferroxidase. Copper deficiency leads to reduced hephaestin protein expression and activity in mouse enterocytes and intestinal cell lines. We tested the effect of copper deficiency on differentiated Caco2 cells grown in transwells and found decreased hephaestin protein expression and activity as well as reduced ferroportin1 protein levels. Furthermore, the decrease in hephaestin levels correlates with a decrease of (55)Fe release from the basolateral side of Caco2 cells. Presence of ceruloplasmin, apo-transferrin or holo-transferrin did not significantly alter the results observed. Repletion of copper in Caco2 cells leads to reconstitution of hephaestin protein expression, activity, and transepithelial iron transport.


Asunto(s)
Células Epiteliales/metabolismo , Hierro/metabolismo , Proteínas de la Membrana/análisis , Transporte Biológico , Células CACO-2 , Proteínas de Transporte de Catión/análisis , Diferenciación Celular , Cobre/deficiencia , Enterocitos/metabolismo , Humanos , Proteínas de la Membrana/metabolismo
4.
Biometals ; 22(5): 827-34, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19330300

RESUMEN

Disorders of iron metabolism are a significant problem primarily in young and old populations. In this study, We compared 1-year-old C57BL6/J mice on iron deficient, iron overload, or iron sufficient diets with two similarly aged genetic models of disturbed iron homeostasis, the sla (sex-linked anemia), and the ceruloplasmin knockout mice (Cp(-/-)) on iron sufficient diet. We found tissue specific changes in sla and nutritional iron deficiency including decreased liver Hamp1 expression and increased protein expression of the enterocyte basolateral iron transport components, hephaestin and ferroportin. In contrast, the Cp(-/-) mice did not show significantly increased Hamp1 expression despite increased liver iron suggesting that regulation is independent of liver iron levels. Together, these results suggest that older mice have a distinct response to alterations in iron metabolism and that age must be considered in future studies of iron metabolism.


Asunto(s)
Envejecimiento/fisiología , Ceruloplasmina/genética , Homeostasis , Hierro/metabolismo , Mutación/genética , Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/metabolismo , Northern Blotting , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Hepcidinas , Homeostasis/efectos de los fármacos , Immunoblotting , Técnicas In Vitro , Sobrecarga de Hierro/metabolismo , Hierro de la Dieta/farmacología , Hígado/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
J Nutr ; 136(5): 1236-41, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16614410

RESUMEN

Copper and iron metabolism intersect in mammals. Copper deficiency simultaneously leads to decreased iron levels in some tissues and iron deficiency anemia, whereas it results in iron overload in other tissues such as the intestine and liver. The copper requirement of the multicopper ferroxidases hephaestin and ceruloplasmin likely explains this link between copper and iron homeostasis in mammals. We investigated the effect of in vivo and in vitro copper deficiency on hephaestin (Heph) expression and activity. C57BL/6J mice were separated into 2 groups on the day of parturition. One group was fed a copper-deficient diet and another was fed a control diet for 6 wk. Copper-deficient mice had significantly lower hephaestin and ceruloplasmin (approximately 50% of controls) ferroxidase activity. Liver hepcidin expression was significantly downregulated by copper deficiency (approximately 60% of controls), and enterocyte mRNA and protein levels of ferroportin1 were increased to 2.5 and 10 times, respectively, relative to controls, by copper deficiency, indicating a systemic iron deficiency in the copper-deficient mice. Interestingly, hephaestin protein levels were significantly decreased to approximately 40% of control, suggesting that decreased enterocyte copper content leads to decreased hephaestin synthesis and/or stability. We also examined the effect of copper deficiency on hephaestin in vitro in the HT29 cell line and found dramatically decreased hephaestin synthesis and activity. Both in vivo and in vitro studies indicate that copper is required for the proper processing and/or stability of hephaestin.


Asunto(s)
Anemia Ferropénica/etiología , Cobre/deficiencia , Proteínas de la Membrana/deficiencia , Animales , Línea Celular Tumoral , Neoplasias del Colon , Femenino , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , Valores de Referencia , Superóxido Dismutasa/metabolismo
6.
Blood ; 103(10): 3933-9, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-14751926

RESUMEN

Hephaestin (Hp) plays an important role in intestinal iron absorption and is predicted to be a ferroxidase based on significant sequence identity to the serum multicopper ferroxidase ceruloplasmin. Here, we demonstrate that Hp has both amine oxidase and ferroxidase activity in cultured cells and primary intestinal enterocytes with the use of both gel and solution assays. The specificity of the activity is shown by immunoblotting, immunoprecipitation, and immunodepletion experiments. Surprisingly, the truncated hephaestin expressed in sex-linked anemia (sla) mice still has measurable, but decreased, oxidase activity. Molecular modeling of the truncated hephaestin suggests retention of a minimum catalytic core required for enzymatic activity. We suggest that hephaestin, by way of its ferroxidase activity, facilitates iron export from intestinal enterocytes, most likely in cooperation with the basolateral iron transporter, Ireg1.


Asunto(s)
Anemia/genética , Ceruloplasmina/metabolismo , Proteínas de la Membrana/metabolismo , Amina Oxidasa (conteniendo Cobre)/metabolismo , Secuencia de Aminoácidos , Animales , Dominio Catalítico , Células Cultivadas , Enterocitos/enzimología , Enterocitos/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X , Hierro/metabolismo , Masculino , Proteínas de la Membrana/análisis , Proteínas de la Membrana/química , Ratones , Ratones Mutantes , Modelos Moleculares , Eliminación de Secuencia
7.
Blood ; 102(5): 1893-9, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12730111

RESUMEN

Hephaestin is a membrane-bound multicopper ferroxidase necessary for iron egress from intestinal enterocytes into the circulation. Mice with sex-linked anemia (sla) have a mutant form of Hephaestin and a defect in intestinal basolateral iron transport, which results in iron deficiency and anemia. Ireg1 (SLC11A3, also known as Ferroportin1 or Mtp1) is the putative intestinal basolateral iron transporter. We compared iron levels and expression of genes involved in iron uptake and storage in sla mice and C57BL/6J mice fed iron-deficient, iron-overload, or control diets. Both iron-deficient wild-type mice and sla mice showed increased expression of Heph and Ireg1 mRNA, compared to controls, whereas only iron-deficient wild-type mice had increased expression of the brush border transporter Dmt1. Unlike iron-deficient mice, sla mouse enterocytes accumulated nonheme iron and ferritin. These results indicate that Dmt1 can be modulated by the enterocyte iron level, whereas Hephaestin and Ireg1 expression respond to systemic rather than local signals of iron status. Thus, the basolateral transport step appears to be the primary site at which the small intestine responds to alterations in body iron requirements.


Asunto(s)
Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Animales , Especificidad de Anticuerpos , Dieta , Enterocitos/metabolismo , Ferritinas/sangre , Expresión Génica , Intestino Delgado/citología , Intestino Delgado/metabolismo , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/metabolismo , Hierro de la Dieta/sangre , Hierro de la Dieta/farmacocinética , Proteínas de Unión a Hierro/genética , Proteínas de Unión a Hierro/metabolismo , Masculino , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Fenómenos Fisiológicos de la Nutrición , ARN Mensajero/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA