Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Neuroinflammation ; 15(1): 30, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29391061

RESUMO

BACKGROUND: Iron regulation is essential for cellular energy production. Loss of cellular iron homeostasis has critical implications for both normal function and disease progression. The H63D variant of the HFE gene is the most common gene variant in Caucasians. The resulting mutant protein alters cellular iron homeostasis and is associated with a number of neurological diseases and cancer. In the brain, microglial and infiltrating macrophages are critical to maintaining iron homeostasis and modulating inflammation associated with the pathogenic process in multiple diseases. This study addresses whether HFE genotype affects macrophage function and the implications of these findings for disease processes. METHODS: Bone marrow macrophages were isolated from wildtype and H67D HFE knock-in mice. The H67D gene variant in mice is the human equivalent of the H63D variant. Upon differentiation, the macrophages were used to analyze iron regulatory proteins, cellular iron release, migration, phagocytosis, and cytokine expression. RESULTS: The results of this study demonstrate that the H67D HFE genotype significantly impacts a number of critical macrophage functions. Specifically, fundamental activities such as proliferation in response to iron exposure, L-ferritin expression in response to iron loading, secretion of BMP6 and cytokines, and migration and phagocytic activity were all found to be impacted by genotype. Furthermore, we demonstrated that exposure to apo-Tf (iron-poor transferrin) can increase the release of iron from macrophages. In normal conditions, 70% of circulating transferrin is unsaturated. Therefore, the ability of apo-Tf to induce iron release could be a major regulatory mechanism for iron release from macrophages. CONCLUSIONS: These studies demonstrate that the HFE genotype impacts fundamental components of macrophage phenotype that could alter their role in degenerative and reparative processes in neurodegenerative disorders.


Assuntos
Genótipo , Proteína da Hemocromatose/genética , Proteína da Hemocromatose/metabolismo , Macrófagos/metabolismo , Animais , Células da Medula Óssea/metabolismo , Proliferação de Células/fisiologia , Células Cultivadas , Técnicas de Introdução de Genes , Humanos , Ferro/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
2.
J Neurochem ; 145(4): 299-311, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29315562

RESUMO

Parkinson's disease is marked clinically by motor dysfunction and pathologically by dopaminergic cell loss in the substantia nigra and iron accumulation in the substantia nigra. The driver underlying iron accumulation remains unknown and could be genetic or environmental. The HFE protein is critical for the regulation of cellular iron uptake. Mutations within this protein are associated with increased iron accumulation including in the brain. We have focused on the commonly occurring H63D variant of the HFE gene as a disease modifier in a number of neurodegenerative diseases. To investigate the role of H63D HFE genotype, we generated a mouse model in which the wild-type (WT) HFE gene is replaced by the H67D gene variant (mouse homolog of the human H63D gene variant). Using paraquat toxicity as the model for Parkinson's disease, we found that WT mice responded as expected with significantly greater motor function, loss of tyrosine hydroxylase staining and increase microglial staining in the substantia nigra, and an increase in R2 relaxation rate within the substantia nigra of the paraquat-treated mice compared to their saline-treated counterparts. In contrast, the H67D mice showed a remarkable resistance to paraquat treatment; specifically differing from the WT mice with no changes in motor function or changes in R2 relaxation rates following paraquat exposure. At baseline, there were differences between the H67D HFE mice and WT mice in gut microbiome profile and increased L-ferritin staining in the substantia nigra that could account for the resistance to paraquat. Of particular note, the H67D HFE mice regardless of whether or not they were treated with paraquat had significantly less tyrosine hydroxylase immunostaining than WT. Our results clearly demonstrate that the HFE genotype impacts the expression of tyrosine hydroxylase in the substantia nigra, the gut microbiome and the response to paraquat providing additional support that the HFE genotype is a disease modifier for Parkinson's disease. Moreover, the finding that the HFE mutant mice are resistant to paraquat may provide a model in which to study resistant mechanisms to neurotoxicants.


Assuntos
Encéfalo/metabolismo , Proteína da Hemocromatose/genética , Síndromes Neurotóxicas/genética , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Microbioma Gastrointestinal/fisiologia , Interação Gene-Ambiente , Variação Genética , Genótipo , Herbicidas/toxicidade , Camundongos , Camundongos Transgênicos , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Paraquat/toxicidade , Doença de Parkinson/genética
3.
Cancer Cell ; 28(4): 441-455, 2015 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-26461092

RESUMO

Glioblastomas display hierarchies with self-renewing cancer stem-like cells (CSCs). RNA sequencing and enhancer mapping revealed regulatory programs unique to CSCs causing upregulation of the iron transporter transferrin, the top differentially expressed gene compared with tissue-specific progenitors. Direct interrogation of iron uptake demonstrated that CSCs potently extract iron from the microenvironment more effectively than other tumor cells. Systematic interrogation of iron flux determined that CSCs preferentially require transferrin receptor and ferritin, two core iron regulators, to propagate and form tumors in vivo. Depleting ferritin disrupted CSC mitotic progression, through the STAT3-FoxM1 regulatory axis, revealing an iron-regulated CSC pathway. Iron is a unique, primordial metal fundamental for earliest life forms, on which CSCs have an epigenetically programmed, targetable dependence.


Assuntos
Neoplasias Encefálicas/patologia , Ferritinas/metabolismo , Glioblastoma/patologia , Ferro/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptores da Transferrina/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Células Cultivadas , Células-Tronco Embrionárias , Epigênese Genética , Ferritinas/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Camundongos , Transplante de Neoplasias , Células-Tronco Neoplásicas/patologia , Receptores da Transferrina/genética , Análise de Sequência de RNA , Transdução de Sinais , Transferrina/metabolismo
4.
PLoS One ; 10(5): e0125272, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25973894

RESUMO

BACKGROUND: There is a significant clinical need for effective treatment of iron deficiency. A number of compounds that can be administered intravenously have been developed. This study examines how the compounds are handled by macrophages and their relative potential to provoke oxidative stress. METHODS: Human kidney (HK-2) cells, rat peritoneal macrophages and renal cortical homogenates were exposed to pharmaceutical iron preparations. Analyses were performed for indices of oxidative stress and cell integrity. In addition, in macrophages, iron uptake and release and cytokine secretion was monitored. RESULTS: HK-2 cell viability was decreased by iron isomaltoside and ferumoxytol and all compounds induced lipid peroxidation. In the renal cortical homogenates, lipid peroxidation occurred at lowest concentrations with ferric carboxymaltose, iron dextran, iron sucrose and sodium ferric gluconate. In the macrophages, iron sucrose caused loss of cell viability. Iron uptake was highest for ferumoxytol and iron isomaltoside and lowest for iron sucrose and sodium ferric gluconate. Iron was released as secretion of ferritin or as ferrous iron via ferroportin. The latter was blocked by hepcidin. Exposure to ferric carboxymaltose and iron dextran resulted in release of tumor necrosis factor α. CONCLUSIONS: Exposure to iron compounds increased cell stress but was tissue and dose dependent. There was a clear difference in the handling of iron from the different compounds by macrophages that suggests in vivo responses may differ.


Assuntos
Dissacarídeos/farmacologia , Células Epiteliais/efeitos dos fármacos , Compostos Férricos/farmacologia , Óxido Ferroso-Férrico/farmacologia , Ferro/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Animais , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Óxido de Ferro Sacarado , Ferritinas/metabolismo , Ácido Glucárico/farmacologia , Hepcidinas/metabolismo , Humanos , Complexo Ferro-Dextran/farmacologia , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Peroxidação de Lipídeos , Macrófagos Peritoneais/citologia , Macrófagos Peritoneais/metabolismo , Maltose/análogos & derivados , Maltose/farmacologia , Estresse Oxidativo , Cultura Primária de Células , Ratos , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA