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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 421(3): 449-55, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22503983

RESUMO

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.


Assuntos
Mucosa Intestinal/metabolismo , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Sequência de Aminoácidos , Animais , Biotinilação , Células CACO-2 , Polaridade Celular , Cães , Humanos , Dados de Sequência Molecular , ATPase Trocadora de Sódio-Potássio/metabolismo
2.
J Nutr ; 140(10): 1728-35, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20685892

RESUMO

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."


Assuntos
Ceruloplasmina/química , Ceruloplasmina/genética , Cobre/análise , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Ceruloplasmina/análise , Cobre/metabolismo , Feminino , Expressão Gênica , Humanos , Ferro/metabolismo , Glândulas Mamárias Animais/enzimologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Modelos Moleculares , Especificidade de Órgãos , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Fragmentos de Peptídeos/química , Placenta/enzimologia , Gravidez , RNA Interferente Pequeno/farmacologia , Ratos , Homologia de Sequência
3.
J Cell Biochem ; 107(4): 803-8, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19452451

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Ferro/metabolismo , Proteínas de Membrana/análise , Transporte Biológico , Células CACO-2 , Proteínas de Transporte de Cátions/análise , Diferenciação Celular , Cobre/deficiência , Enterócitos/metabolismo , Humanos , Proteínas de Membrana/metabolismo
4.
Biometals ; 22(5): 827-34, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19330300

RESUMO

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.


Assuntos
Envelhecimento/fisiologia , Ceruloplasmina/genética , Homeostase , Ferro/metabolismo , Mutação/genética , Anemia Ferropriva/genética , Anemia Ferropriva/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Northern Blotting , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Hepcidinas , Homeostase/efeitos dos fármacos , Immunoblotting , Técnicas In Vitro , Sobrecarga de Ferro/metabolismo , Ferro da Dieta/farmacologia , Fígado/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
J Nutr ; 136(5): 1236-41, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16614410

RESUMO

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.


Assuntos
Anemia Ferropriva/etiologia , Cobre/deficiência , Proteínas de Membrana/deficiência , Animais , Linhagem Celular Tumoral , Neoplasias do Colo , Feminino , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Valores de Referência , Superóxido Dismutase/metabolismo
6.
Blood ; 102(5): 1893-9, 2003 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12730111

RESUMO

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.


Assuntos
Anemia Ferropriva/genética , Anemia Ferropriva/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Especificidade de Anticorpos , Dieta , Enterócitos/metabolismo , Ferritinas/sangue , Expressão Gênica , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Sobrecarga de Ferro/genética , Sobrecarga de Ferro/metabolismo , Ferro da Dieta/sangue , Ferro da Dieta/farmacocinética , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Masculino , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fenômenos Fisiológicos da Nutrição , RNA Mensageiro/análise
7.
Blood ; 103(10): 3933-9, 2004 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-14751926

RESUMO

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.


Assuntos
Anemia/genética , Ceruloplasmina/metabolismo , Proteínas de Membrana/metabolismo , Amina Oxidase (contendo Cobre)/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Células Cultivadas , Enterócitos/enzimologia , Enterócitos/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X , Ferro/metabolismo , Masculino , Proteínas de Membrana/análise , Proteínas de Membrana/química , Camundongos , Camundongos Mutantes , Modelos Moleculares , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa