Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 503(3): 1905-1910, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30060949

RESUMEN

Multi-copper ferroxidases (MCFs) play important roles in cellular iron metabolism and homeostasis. In this study, we generated the hephaestin (Heph), ceruloplasmin (Cp) single and Heph/Cp double knockout (KO) mice to investigate the roles of MCFs in iron transport among system and vital organs in mice at 4 weeks and 6 months of age. Compared with wild-type (WT) mice, Heph/Cp mice at both ages presented with severe anemia and significantly lower iron level in the serum and spleen, but with significantly higher iron level in the liver, heart, kidney, and duodenal enterocytes. Furthermore, Heph/Cp mice displayed significantly lower level of hepcidin mRNA and transferrin receptor 1 (TFR1) protein expression, but significantly higher level of ferroportin 1 (FPN1) protein expression in the liver than WT mice at 6 months of age. Liver superoxide dismutase (SOD) and glutathione peroxidase (GPx) enzyme activities were significantly lower in Heph/Cp KO mice than WT mice at 6 months of age. Together, our results suggest that ablation of HEPH and CP could lead to severe systemic iron deficiency and local tissue iron overload, which disrupt the whole body iron homeostasis and impact on tissue functions.


Asunto(s)
Anemia Ferropénica/genética , Anemia Ferropénica/metabolismo , Ceruloplasmina/deficiencia , Eliminación de Gen , Homeostasis/genética , Hierro/metabolismo , Proteínas de la Membrana/deficiencia , Animales , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
J Nutr ; 148(4): 643-649, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29659961

RESUMEN

Background: The accumulation of iron occurs in the central nervous system (CNS) in several neurodegenerative diseases. Although multi-copper ferroxidases (MCFs) play an important role in cellular iron metabolism and homeostasis, the mechanism of MCFs in the CNS remains unclear. Objective: The aim was to study the role of MCFs in CNS iron metabolism and homeostasis by using hephaestin/ceruloplasmin (Heph/Cp) double knockout (KO) mice. Methods: Heph/Cp double KO male mice were generated by crossing both single KO mice. In Heph/Cp KO and wild-type (WT) control mice at 4 wk and 6 mo of age, iron concentrations of selected brain regions were measured by atomic absorption spectrophotometry, and gene expressions of Heph, Cp, ferroportin 1 (Fpn1) [+ iron responsive element (IRE)], L-ferritin, H-ferritin, transferrin receptor 1 (Tfrc), and divalent metal transporter 1 (Dmt1) (+IRE) were quantitated by quantitative reverse transcriptase-polymerase chain reaction. Brain region L-ferritin protein concentration, superoxide dismutase (SOD), and glutathione peroxidase (GPx) activities and malondialdehyde (MDA) concentration were also determined. Learning and memory abilities in Heph/Cp KO and WT control mice at 6 mo of age were tested by the IntelliCage system (New Behavior). Results: Iron concentration was significantly higher in Heph/Cp KO mice than in WT control mice at 4 wk of age in the cortex (50%), hippocampus (120%), brainstem (35%), and cerebellum (220%) and at 6 mo of age in the cortex (140%), hippocampus (420%), brainstem (560%), and cerebellum (340%). L-Ferritin and MDA concentrations were significantly higher and SOD and GPx activities were significantly lower in the cortex, hippocampus, brainstem, and cerebellum of KO mice than in those of WT controls at both 4 wk and 6 mo of age. Iron-related gene expressions also differed significantly between groups. Learning and memory deficits occurred in Heph/Cp KO mice at 6 mo of age. Conclusion: Mutation of both MCFs in mice induces iron accumulation in brain regions, oxidative damage, and learning and memory defects.


Asunto(s)
Encéfalo/metabolismo , Ceruloplasmina/deficiencia , Cobre/metabolismo , Hierro/metabolismo , Discapacidades para el Aprendizaje/etiología , Trastornos de la Memoria/etiología , Estrés Oxidativo , Animales , Conducta Animal , Proteínas de Transporte de Catión/metabolismo , Ceruloplasmina/metabolismo , Ferritinas/metabolismo , Glutatión Peroxidasa/metabolismo , Aprendizaje , Masculino , Malondialdehído/metabolismo , Memoria , Ratones Noqueados , Receptores de Transferrina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/metabolismo
3.
Neurosci Lett ; 708: 134346, 2019 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-31229624

RESUMEN

AIMS: This study aimed to explore the synergistic effects of nitrogen-doped carbon nanocages (NCNCs) and human umbilical cord mesenchymal stem cells (HUC-MSCs) on ischemic stroke and investigate the potential underlying mechanisms. MAIN METHODS: The properties of NCNCs were analyzed by transmission electron microscopy, and the markers of HUC-MSCs were detected by flow cytometry. The cell toxicity of NCNCs was evaluated by MTT. Mice were induced cerebral infarction by transient middle cerebral artery occlusion (MCAO). NCNCs or HUC-MSCs or HUC-MSCs-NCNCs were intravenously injected thirty minutes after reperfusion. The infarct volume was examined by 2,3,5-triphenyltetrazolium chloride (TTC) staining, and behavior tests were evaluated by the modified Neurological Severity Score (mNSS) and rotarod test. The mRNA levels of TNF-α and IL-10 were detected by real-time PCR. The protein levels of TNF-α stimulated gene/protein 6 (TSG-6) and prostaglandin 2 (PGE2) were detected by ELISA. The microglia markers (CD86 and CD206) and the protein levels of TNF-α and IL-10 were examined by flow cytometry. The protein levels of Iba1 and CD16 were determined by immunostaining. KEY FINDINGS: NCNCs enhanced the therapeutic effects of HUC-MSCs on MCAO mice, including reducing infarct volume, improving behavior scores and inhibiting inflammation response. In addition, NCNCs and HUC-MSCs cooperatively inhibit the mRNA and protein levels of TNF-α, and increased the mRNA and protein levels of IL-10 and protein levels of PGE2 and TSG-6 in LPS-treated microglia. Furthermore, NCNCs exerted synergistic effects with HUC-MSCs on remodeling microglia polarization. SIGNIFICANCE: NCNCs enhance the therapeutic effects of HUC-MSCs on cerebral infarction in a mouse MCAO model, and inhibit the microglia reactivation and neuroinflammation, which indicates it as a potential treatment for ischemic stroke.


Asunto(s)
Isquemia Encefálica/prevención & control , Carbono , Trasplante de Células Madre de Sangre del Cordón Umbilical , Trasplante de Células Madre Mesenquimatosas , Nanoestructuras , Accidente Cerebrovascular/prevención & control , Animales , Isquemia Encefálica/etiología , Isquemia Encefálica/metabolismo , Carbono/química , Polaridad Celular , Células Cultivadas , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Inflamación/metabolismo , Inflamación/terapia , Masculino , Ratones Endogámicos C57BL , Microglía/fisiología , Nanoestructuras/química , Nanoestructuras/toxicidad , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo
4.
Sci Rep ; 9(1): 9437, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31263155

RESUMEN

Accumulation of iron has been associated with the pathobiology of various disorders of the central nervous system. Our previous work has shown that hephaestin (Heph) and ceruloplasmin (Cp) double knockout (KO) mice induced iron accumulation in multiple brain regions and that this was paralleled by increased oxidative damage and deficits in cognition and memory. In this study, we enriched astrocytes and oligodendrocytes from the cerebral cortex of neonatal wild-type (WT), Heph KO and Cp KO mice. We demonstrated that Heph is highly expressed in oligodendrocytes, while Cp is mainly expressed in astrocytes. Iron efflux was impaired in Cp KO astrocytes and Heph KO oligodendrocytes and was associated with increased oxidative stress. The expression of Heph, Cp, and other iron-related genes was examined in astrocytes and oligodendrocytes both with and without iron treatment. Interestingly, we found that the expression of the mRNA encoding ferroportin 1, a transmembrane protein that cooperates with CP and HEPH to export iron from cells, was positively correlated with Cp expression in astrocytes, and with Heph expression in oligodendrocytes. Our findings collectively demonstrate that HEPH and CP are important for the prevention of glial iron accumulation and thus may be protective against oxidative damage.


Asunto(s)
Ceruloplasmina/genética , Hierro/metabolismo , Proteínas de la Membrana/genética , Estrés Oxidativo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Ceruloplasmina/deficiencia , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodendroglía/citología , Oligodendroglía/metabolismo , Estrés Oxidativo/genética
5.
Sci Rep ; 6: 39470, 2016 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-27991585

RESUMEN

Multicopper ferroxidases (MCFs) play an important role in cellular iron homeostasis. However, the role of MCFs in renal metabolism remains unclear. We used Hephaestin (Heph) and Ceruloplasmin (Cp) single or double (Heph/Cp) knockout (KO) mice to study the roles of MCFs in the kidney. Renal iron levels and the expression of iron metabolism genes were examined. The non-heme iron content both in the renal cortex and medulla of Heph/Cp KO mice was significantly increased. Perls' Prussian blue staining showed iron accumulation on the apical side of renal tubular cells in Heph/Cp KO mice. A significant increase in ferritin protein expression was also observed in the renal medulla and cortex of Heph/Cp KO mice. Both DMT1 and TfR1 protein expression were significantly decreased in the renal medulla of Heph/Cp KO mice, while the expression of DMT1 protein was significantly increased in the renal cortex of these animals. Significant increase in proteinuria and total urinary iron was observed in the double knockout mice, and this was associated with compromised structural integrity. These results suggest that KO of both the HEPH and CP genes leads to kidney iron deposition and toxicity, MCFs could protect kidney against a damage from iron excess.


Asunto(s)
Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Hierro/metabolismo , Riñón/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Animales , Cobre/química , Ferritinas/metabolismo , Ferrocianuros , Genotipo , Homeostasis , Corteza Renal , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA