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.
Biochim Biophys Acta Mol Basis Dis ; 1867(7): 166142, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33839281

RESUMEN

BACKGROUND & AIMS: Iron has been proposed as influencing the progression of liver disease in subjects with non-alcoholic fatty liver disease (NAFLD). We have previously shown that, in the Hfe-/- mouse model of hemochromatosis, feeding of a high-calorie diet (HCD) leads to increased liver injury. In this study we investigated whether the feeding of an iron deficient/HCD to Hfe-/- mice influenced the development of NAFLD. METHODS: Liver histology was assessed in Hfe-/- mice fed a standard iron-containing or iron-deficient diet plus or minus a HCD. Hepatic iron concentration, serum transferrin saturation and free fatty acid were measured. Expression of genes implicated in iron regulation and fatty liver disease was determined by quantitative real-time PCR (qRT-PCR). RESULTS: Standard iron/HCD-fed mice developed severe steatosis whereas NAS score was reduced in mice fed iron-deficient HCD. Mice fed iron-deficient HCD had lower liver weights, lower transferrin saturation and decreased ferroportin and hepcidin gene expression than HCD-fed mice. Serum non-esterified fatty acids were increased in iron-deficient HCD-fed mice compared with standard iron HCD. Expression analysis indicated that genes involved in fatty-acid binding and mTOR pathways were regulated by iron depletion. CONCLUSIONS: Our results indicate that decreasing iron intake attenuates the development of steatosis resulting from a high calorie diet. These results also suggest that human studies of agents that modify iron balance in patients with NAFLD should be revisited.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Hígado Graso/prevención & control , Proteína de la Hemocromatosis/fisiología , Deficiencias de Hierro , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Animales , Ácidos Grasos no Esterificados/metabolismo , Hígado Graso/etiología , Hígado Graso/metabolismo , Hígado Graso/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800732

RESUMEN

Matriptase-2, a serine protease expressed in hepatocytes, is a negative regulator of hepcidin expression. The purpose of the study was to investigate the interaction of matriptase-2 with hemojuvelin protein in vivo. Mice lacking the matriptase-2 proteolytic activity (mask mice) display decreased content of hemojuvelin protein. Vice versa, the absence of hemojuvelin results in decreased liver content of matriptase-2, indicating that the two proteins interact. To further characterize the role of matriptase-2, we investigated iron metabolism in mask mice fed experimental diets. Administration of iron-enriched diet increased liver iron stores as well as hepcidin expression. Treatment of iron-overloaded mask mice with erythropoietin increased hemoglobin and hematocrit, indicating that the response to erythropoietin is intact in mask mice. Feeding of an iron-deficient diet to mask mice significantly increased spleen weight as well as the splenic content of erythroferrone and transferrin receptor proteins, indicating stress erythropoiesis. Liver hepcidin expression was decreased; expression of Id1 was not changed. Overall, the results suggest a complex interaction between matriptase-2 and hemojuvelin, and demonstrate that hepcidin can to some extent be regulated even in the absence of matriptase-2 proteolytic activity.


Asunto(s)
Proteínas Ligadas a GPI/fisiología , Proteína de la Hemocromatosis/fisiología , Sobrecarga de Hierro/metabolismo , Proteínas de la Membrana/fisiología , Serina Endopeptidasas/fisiología , Animales , Proteína Morfogenética Ósea 6/biosíntesis , Proteína Morfogenética Ósea 6/genética , Eritropoyetina/farmacología , Femenino , Proteínas Ligadas a GPI/biosíntesis , Proteínas Ligadas a GPI/deficiencia , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteína de la Hemocromatosis/biosíntesis , Proteína de la Hemocromatosis/deficiencia , Proteína de la Hemocromatosis/genética , Hepcidinas/biosíntesis , Hepcidinas/genética , Proteína 1 Inhibidora de la Diferenciación/biosíntesis , Proteína 1 Inhibidora de la Diferenciación/genética , Deficiencias de Hierro , Hierro de la Dieta/farmacología , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Especificidad de Órganos , Regiones Promotoras Genéticas/genética , Dominios Proteicos , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Bazo/metabolismo
3.
Hepatology ; 72(2): 642-655, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31778583

RESUMEN

BACKGROUND AND AIMS: Bone morphogenetic proteins BMP2 and BMP6 play key roles in systemic iron homeostasis by regulating production of the iron hormone hepcidin. The homeostatic iron regulator (HFE) also regulates hepcidin through a mechanism that intersects with the BMP-mothers against decapentaplegic homolog 1/5/8 (SMAD1/5/8) pathway. However, the relative roles of BMP2 compared with BMP6 and whether HFE regulates hepcidin through a BMP2-dependent mechanism remain uncertain. APPROACH AND RESULTS: We therefore examined the iron phenotype of mice deficient for both Bmp2 and Bmp6 or both Bmp2 and Hfe compared with single knockout (KO) mice and littermate controls. Eight-week-old double endothelial Bmp6/Bmp2 KO mice exhibited a similar degree of hepcidin deficiency, serum iron overload, and tissue iron overload compared with single KO mice. Notably, dietary iron loading still induced liver SMAD5 phosphorylation and hepcidin in double Bmp6/endothelial Bmp2 KO mice, although no other BMP ligand mRNAs were increased in the livers of double KO mice, and only Bmp6 and Bmp2 mRNA were induced by dietary iron loading in wild-type mice. In contrast, double Hfe/endothelial Bmp2 KO mice exhibited reduced hepcidin and increased extrahepatic iron loading compared to single Hfe or endothelial Bmp2 KO mice. Liver phosphorylated SMAD5 and the SMAD1/5/8 target inhibitor of DNA binding 1 (Id1) mRNA were also reduced in double Hfe/endothelial Bmp2 KO compared with single endothelial Bmp2 KO female mice. Finally, hepcidin and Id1 mRNA induction by homodimeric BMP2, homodimeric BMP6, and heterodimeric BMP2/6 were blunted in Hfe KO primary hepatocytes. CONCLUSIONS: These data suggest that BMP2 and BMP6 work collaboratively to regulate hepcidin expression, that BMP2-independent and BMP6-independent SMAD1/5/8 signaling contributes a nonredundant role to hepcidin regulation by iron, and that HFE regulates hepcidin at least in part through a BMP2-independent but SMAD1/5/8-dependent mechanism.


Asunto(s)
Proteína Morfogenética Ósea 2/fisiología , Proteína Morfogenética Ósea 6/fisiología , Proteína de la Hemocromatosis/fisiología , Hemocromatosis/etiología , Animales , Endotelio , Femenino , Masculino , Ratones , Ratones Noqueados
4.
Transfus Clin Biol ; 26(4): 249-252, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31679808

RESUMEN

INTRODUCTION: Mutations in the HFE gene have been shown to be associated with hemochromatosis which is observed in beta-thalassemia major. In this study, we determined the HFE gene mutations (C282Y and H63D) among b-thalassemia major patients to investigate the effect of these mutations on serum Ferritin levels. MATERIAL AND METHODS: In this cross-sectional study, a total of 105 b-thalassemia subjects with a history of regular blood transfusion were selected. They divided into two distinct groups according cut off 1000ng/ml of serum Ferritin levels. The HFE gene mutant allele detected by RFLP-PCR. RESULTS: Of 105 thalassemia patients, 29 patients (14 male and 15 female) were heterozygote for H63D mutation, and just one male was homozygote, but for C282Y mutation just one heterozygote and one homozygote was detected, and overall 31% had coexistence of b-thal and HFE gene mutations. As expected, Ferritin levels significantly differed between groups (P=0.001). CONCLUSION: The impact of detection of HFE mutations could prognosis the likelihood of iron overload in multi-transfused patients, and allowing early diagnosis and proper management to overcome complications of iron overload in beta-thalassemia patients.


Asunto(s)
Ferritinas/sangre , Proteína de la Hemocromatosis/genética , Hemocromatosis/genética , Sobrecarga de Hierro/etiología , Mutación Puntual , Reacción a la Transfusión , Talasemia beta/sangre , Alelos , Transfusión Sanguínea , Estudios Transversales , Femenino , Frecuencia de los Genes , Hemocromatosis/epidemiología , Proteína de la Hemocromatosis/fisiología , Humanos , Irán/epidemiología , Sobrecarga de Hierro/genética , Sobrecarga de Hierro/prevención & control , Masculino , Pronóstico , Receptores de Transferrina/metabolismo , Transferrina/metabolismo , Microglobulina beta-2/metabolismo , Talasemia beta/terapia
5.
FASEB J ; 33(2): 2460-2471, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30277817

RESUMEN

Iron deficiency is closely associated with altered GABA metabolism and affective behavior. While mutation in the hemochromatosis ( HFE) gene disrupts iron homeostasis and promotes oxidative stress that increases the risk of neurodegeneration, it is largely unknown whether HFE mutation modifies GABAergic homeostasis and emotional behavior. The goal of our study was to investigate the impact of HFE on GABAergic neurochemistry and redox-epigenetic regulation in the brain using H67D HFE-mutant mice that recapitulates the H63D-HFE mutation in humans. H67D mice displayed elevated redox-active iron levels in the brain by 32% compared to age-matched wild-type mice. Moreover, the H67D brain had increased isoprostane and decreased glutathione, indicating elevated oxidative stress. Additionally, the H67D brain had decreased global methylation and attenuated DNA methyltransferase (DNMT) activity. Direct addition of iron to purified DNMT in vitro decreased enzyme activity in a concentration-dependent manner. Last, H67D mice exhibited decreased anxiety-like behavior, which was associated with increased expression of the GABAA receptor α2 subunits by 93%, and these changes were also observed in H67D mice fed a low-iron diet. Taken together, our results suggest a putative role of HFE in regulating labile iron status in the brain, and mutation in H67D perturbs redox-methylation status, contributing to GABAergic dysfunction.-Ye, Q., Trivedi, M., Zhang, Y., Böhlke, M., Alsulimani, H., Chang, J., Maher, T., Deth, R., Kim, J. Brain iron loading impairs DNA methylation and alters GABAergic function in mice.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Metilación de ADN , Proteína de la Hemocromatosis/fisiología , Hierro/metabolismo , Mutación , Receptores de GABA-A/metabolismo , Animales , Epigénesis Genética , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Subunidades de Proteína , Receptores de GABA-A/genética
6.
Am J Hematol ; 93(3): 383-393, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29194702

RESUMEN

We investigated the role of HFE C282Y, H63D, and TMPRSS6 A736V variants in the pathogenesis of iron deficiency anemia (IDA) in celiac disease (CD) patients, at diagnosis and after 1 year of gluten-free diet (GFD). Demographic and clinical features were prospectively recorded for all CD patients between 2013 and 2017. C282Y, H63D, and A736V variants were evaluated for CD patients and controls. Finally, 505 consecutive CD patients and 539 age-matched control subjects were enrolled. At diagnosis, 229 CD subjects had IDA (45.3%), with a subgroup of anemic patients (45.4%) presented persistent IDA at follow-up. C282Y allele frequency was significantly increased in CD compared with controls (1.1% vs 0.2%, P = .001), whereas H63D and A736V allele frequencies were similar among patients and controls (P = .92 and .84, respectively). At diagnosis, C282Y variant in anemic CD patients was significantly increased compared to nonanemic group (2% and 0.5%, P = .04). At follow-up, A736V was significantly increased in IDA persistent than in IDA not persistent (57.7% vs 35.2%, P < .0001). CD patients with H63D mutation showed higher Hb, MCV, serum iron, and ferritin levels than subjects without HFE mutations. Decreased hepcidin values were observed in anemic compared to nonanemic subjects at follow-up (1.22 ± 1.14 vs 2.08 ± 2.15, P < .001). This study suggests a protective role of HFE in IDA CD patients and confirms the role of TMPRSS6 in predicting oral iron response modulating hepcidin action on iron absorption. Iron supplementation therapeutic management in CD could depend on TMPRSS6 genotype that could predict persistent IDA despite iron supplementation and GFD.


Asunto(s)
Anemia Ferropénica/genética , Enfermedad Celíaca/genética , Proteína de la Hemocromatosis/fisiología , Proteínas de la Membrana/fisiología , Mutación Missense , Serina Endopeptidasas/fisiología , Adulto , Alelos , Anemia Ferropénica/etiología , Autoanticuerpos/sangre , Enfermedad Celíaca/complicaciones , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/fisiopatología , Dieta Sin Gluten , Índices de Eritrocitos , Femenino , Ferritinas/sangre , Frecuencia de los Genes , Proteína de la Hemocromatosis/genética , Hemoglobinas/análisis , Hepcidinas/sangre , Humanos , Absorción Intestinal , Hierro/sangre , Hierro de la Dieta/farmacocinética , Masculino , Proteínas de la Membrana/genética , Estudios Prospectivos , Serina Endopeptidasas/genética , Resultado del Tratamiento , Adulto Joven
7.
Mol Psychiatry ; 21(11): 1599-1607, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26728570

RESUMEN

The 'neurodegeneration with brain iron accumulation' (NBIA) disease family entails movement or cognitive impairment, often with psychiatric features. To understand how iron loading affects the brain, we studied mice with disruption of two iron regulatory genes, hemochromatosis (Hfe) and transferrin receptor 2 (Tfr2). Inductively coupled plasma atomic emission spectroscopy demonstrated increased iron in the Hfe-/- × Tfr2mut brain (P=0.002, n ≥5/group), primarily localized by Perls' staining to myelinated structures. Western immunoblotting showed increases of the iron storage protein ferritin light polypeptide and microarray and real-time reverse transcription-PCR revealed decreased transcript levels (P<0.04, n ≥5/group) for five other NBIA genes, phospholipase A2 group VI, fatty acid 2-hydroxylase, ceruloplasmin, chromosome 19 open reading frame 12 and ATPase type 13A2. Apart from the ferroxidase ceruloplasmin, all are involved in myelin homeostasis; 16 other myelin-related genes also showed reduced expression (P<0.05), although gross myelin structure and integrity appear unaffected (P>0.05). Overlap (P<0.0001) of differentially expressed genes in Hfe-/- × Tfr2mut brain with human gene co-expression networks suggests iron loading influences expression of NBIA-related and myelin-related genes co-expressed in normal human basal ganglia. There was overlap (P<0.0001) of genes differentially expressed in Hfe-/- × Tfr2mut brain and post-mortem NBIA basal ganglia. Hfe-/- × Tfr2mut mice were hyperactive (P<0.0112) without apparent cognitive impairment by IntelliCage testing (P>0.05). These results implicate myelin-related systems involved in NBIA neuropathogenesis in early responses to iron loading. This may contribute to behavioral symptoms in NBIA and hemochromatosis and is relevant to patients with abnormal iron status and psychiatric disorders involving myelin abnormalities or resistant to conventional treatments.


Asunto(s)
Trastornos del Metabolismo del Hierro/metabolismo , Trastornos del Metabolismo del Hierro/fisiopatología , Hierro/efectos adversos , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/fisiopatología , Animales , Encéfalo/metabolismo , Expresión Génica , Hemocromatosis/genética , Hemocromatosis/metabolismo , Proteína de la Hemocromatosis/metabolismo , Proteína de la Hemocromatosis/fisiología , Hierro/sangre , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos AKR , Vaina de Mielina/metabolismo , Distrofias Neuroaxonales/genética , Linaje , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...