Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 293(20): 7853-7863, 2018 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-29610275

RESUMO

Microglia are immune cells of the central nervous system and are implicated in brain inflammation. However, how brain microglia modulate transport and metabolism of the essential metal iron in response to pro- and anti-inflammatory environmental cues is unclear. Here, we characterized uptake of transferrin (Tf)-bound iron (TBI) and non-Tf-bound iron (NTBI) by immortalized microglial (IMG) cells. We found that these cells preferentially take up NTBI in response to the proinflammatory stimulus lipopolysaccharide (LPS) or ß-amyloid (Aß). In contrast, the anti-inflammatory cytokine interleukin 4 (IL-4) promoted TBI uptake. Concordant with these functional data, levels of the Tf receptor (TfR) in IMG cells were up-regulated in response to IL-4, whereas divalent metal transporter-1 (DMT1) and ferritin levels increased in response to LPS or Aß. Similar changes in expression were confirmed in isolated primary adult mouse microglia treated with pro- or anti-inflammatory inducers. LPS-induced changes in IMG cell iron metabolism were accompanied by notable metabolic changes, including increased glycolysis and decreased oxidative respiration. Under these conditions, the extracellular acidification rate was increased, compatible with changes in the cellular microenvironment that would support the pH-dependent function of DMT1. Moreover, LPS increased heme oxygenase-1 (HO1) expression in IMG cells, and iron released because of HO1 activity increased the intracellular labile free-iron pool. Together, this evidence indicates that brain microglia preferentially acquire iron from Tf or from non-Tf sources, depending on their polarization state; that NTBI uptake is enhanced by the proinflammatory response; and that under these conditions microglia sequester both extra- and intracellular iron.


Assuntos
Proteínas de Transporte de Cátions/genética , Ferro/metabolismo , Microglia/metabolismo , Receptores da Transferrina/genética , Transferrina/genética , Peptídeos beta-Amiloides/farmacologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Transformada , Microambiente Celular , Ferritinas/genética , Ferritinas/metabolismo , Regulação da Expressão Gênica , Glicólise/efeitos dos fármacos , Glicólise/genética , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Concentração de Íons de Hidrogênio , Inflamação , Transporte de Íons , Lipopolissacarídeos/farmacologia , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Cultura Primária de Células , Receptores da Transferrina/metabolismo , Transdução de Sinais , Transferrina/metabolismo
2.
J Neuroinflammation ; 13: 21, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26819091

RESUMO

BACKGROUND: Alzheimer's disease is associated with amyloid-beta (Aß)-induced microglia activation. This pro-inflammatory response promotes neuronal damage, and therapies are sought to limit microglial activation. Screening efforts to develop new pharmacological inhibitors require a robust in vitro cell system. Current models lack significant responses to Aß, and their use in examining age-related neurodegenerative diseases is questionable. For example, the commonly used BV-2 microglial line was derived from embryonic mononuclear cells and its activation by various stimuli is limited. To this end, we have established a new immortalized microglial (IMG) cell line from adult murine brain. The objective of this study was to characterize Aß-induced activation of IMG cells, and here, we demonstrate the ability of cannabinoids to significantly reduce this inflammatory response. METHODS: Microglial cells derived from adult murine brain were immortalized via infection with the v-raf/v-myc retrovirus under conditions that selectively promote microglia growth. The presence or absence of markers CD11b and F4/80 (microglial), NeuN (neuronal), and GFAP (astrocytic) was assessed by immunofluorescence microscopy and western blotting. Using IMG and BV-2 cells, levels of pro- and anti-inflammatory transcripts in response to extracellular stimuli were determined by quantitative PCR (qPCR). Phagocytosis of fluorescent beads and fluorescein isothiocyanate (FITC)-labeled Aß oligomers was assessed using flow cytometry and fluorescence microscopy. FITC-Aß uptake was quantified using a fluorescence plate reader. The ability of cannabinoids to mitigate Aß-induced expression of inducible nitric oxide synthase (iNOS) was evaluated. RESULTS: IMG cells express the microglial markers CD11b and F4/80 but not NeuN or GFAP. Relative to BV-2 cells, IMG cells increased iNOS (>200-fold) and Arg-1 (>100-fold) in response to pro- and anti-inflammatory stimuli. IMG cells phagocytose foreign particles and Aß oligomers, with the latter trafficked to phagolysosomes. Aß-induced activation of IMG cells was suppressed by delta-9-tetrahydrocannabinol and the CB2-selective agonist JWH-015 in a time- and concentration-dependent manner. CONCLUSIONS: IMG cells recapitulate key features of microglial cell activation. As an example of their potential pharmacological use, cannabinoids were shown to reduce activation of Aß-induced iNOS gene expression. IMG cells hold promising potential for drug screening, mechanistic studies, and functional investigations directed towards understanding how Aß interacts with microglia.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Microglia/metabolismo , Análise de Variância , Animais , Antígenos de Diferenciação/metabolismo , Antígeno CD11b/metabolismo , Células Cultivadas , Citometria de Fluxo , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Técnicas In Vitro , Interleucina-1beta/metabolismo , Microglia/efeitos dos fármacos , Fagócitos/metabolismo , RNA Mensageiro/metabolismo
3.
EMBO Rep ; 15(7): 809-15, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24867889

RESUMO

A sequence within the E2 domain of soluble amyloid precursor protein (sAPP) stimulates iron efflux. This activity has been attributed to a ferroxidase activity suggested for this motif. We demonstrate that the stimulation of efflux supported by this peptide and by sAPPα is due to their stabilization of the ferrous iron exporter, ferroportin (Fpn), in the plasma membrane of human brain microvascular endothelial cells (hBMVEC). The peptide does not bind ferric iron explaining why it does not and thermodynamically cannot promote ferrous iron autoxidation. This peptide specifically pulls Fpn down from the plasma membrane of hBMVEC; based on these results, FTP, for ferroportin-targeting peptide, correctly identifies the function of this peptide. The data suggest that in stabilizing Fpn via the targeting due to the FTP sequence, sAPP will increase the flux of iron into the cerebral interstitium. This inference correlates with the observation of significant iron deposition in the amyloid plaques characteristic of Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Células Endoteliais/metabolismo , Ferro/metabolismo , Sequência de Aminoácidos , Peptídeos beta-Amiloides/metabolismo , Peptídeos beta-Amiloides/farmacologia , Precursor de Proteína beta-Amiloide/química , Ceruloplasmina/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana , Humanos , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas
4.
Cell Mol Life Sci ; 72(4): 709-27, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25355056

RESUMO

There are two barriers for iron entry into the brain: (1) the brain-cerebrospinal fluid (CSF) barrier and (2) the blood-brain barrier (BBB). Here, we review the literature on developmental iron accumulation by the brain, focusing on the transport of iron through the brain microvascular endothelial cells (BMVEC) of the BBB. We review the iron trafficking proteins which may be involved in the iron flux across BMVEC and discuss the plausible mechanisms of BMVEC iron uptake and efflux. We suggest a model for how BMVEC iron uptake and efflux are regulated and a mechanism by which the majority of iron is trafficked across the developing BBB under the direct guidance of neighboring astrocytes. Thus, we place brain iron uptake in the context of the neurovascular unit of the adult brain. Last, we propose that BMVEC iron is involved in the aggregation of amyloid-ß peptides leading to the progression of cerebral amyloid angiopathy which often occurs prior to dementia and the onset of Alzheimer's disease.


Assuntos
Barreira Hematoencefálica/metabolismo , Ferro/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Astrócitos/metabolismo , Encéfalo/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Angiopatia Amiloide Cerebral/patologia , Células Endoteliais/metabolismo , Humanos , Transporte de Íons , Receptores da Transferrina/metabolismo , Transferrina/metabolismo
5.
J Biol Chem ; 288(24): 17932-40, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640881

RESUMO

The mechanism(s) of iron flux across the brain microvasculature endothelial cells (BMVEC) of the blood-brain barrier remains unknown. Although both hephaestin (Hp) and the ferrous iron permease ferroportin (Fpn) have been identified in BMVEC, their roles in iron efflux have not been examined. Using a human BMVEC line (hBMVEC), we have demonstrated that these proteins are required for iron efflux from these cells. Expression of both Hp and Fpn protein was confirmed in hBMVEC by immunoblot and indirect immunofluorescence; we show that hBMVEC express soluble ceruloplasmin (Cp) transcript as well. Depletion of endogenous Hp and Cp via copper chelation leads to the reduction of hBMVEC Fpn protein levels as well as a complete inhibition of (59)Fe efflux. Both hBMVEC Fpn protein and (59)Fe efflux activity are restored upon incubation with 6.6 nm soluble plasma Cp. These results are independent of the source of cell iron, whether delivered as transferrin- or non-transferrin-bound (59)Fe. Our results demonstrate that iron efflux from hBMVEC Fpn requires the action of an exocytoplasmic ferroxidase, which can be either endogenous Hp or extracellular Cp.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Ceruloplasmina/metabolismo , Células Endoteliais/enzimologia , Ferro/metabolismo , Microvasos/citologia , Encéfalo/irrigação sanguínea , Células CACO-2 , Membrana Celular/metabolismo , Quelantes/farmacologia , Cobre/metabolismo , Meios de Cultivo Condicionados , Células Endoteliais/efeitos dos fármacos , Células Hep G2 , Humanos , Proteínas de Membrana/metabolismo , Fenantrolinas/farmacologia , Ligação Proteica , Transporte Proteico
6.
Cell Commun Signal ; 12: 65, 2014 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-25311416

RESUMO

BACKGROUND: Iron transport across the blood-brain barrier (BBB) involves the cooperation of brain microvascular endothelial cells (BMVEC) and their neighboring astrocytes. Astrocytes secrete a soluble form of ceruloplasmin (sCp) which, in turn, acts to export iron from ferroportin (Fpn) on the basolateral surface of BMVEC. Although regulation of astrocyte sCp gene expression has been demonstrated to be influenced by interleukin-1 beta (IL-1ß) and interleukin-6 (IL-6), the role of neighboring BMVEC in this regulation has yet to be determined and is the basis for this work. RESULTS: We provide evidence that human BMVEC (hBMVEC) IL-1ß and IL-6 positively influence the expression of sCp transcript by neighboring C6 glioma cells (astrocytes). The effect of hBMVEC on C6 glioma sCp expression at the level of transcript and protein was repressed via the addition of IL-1ß and IL-6 pathway inhibitors (IL-1 receptor antagonist protein and SC144, respectively). Stimulation of hBMVEC interleukin gene expression by apical exposure to bacterial endotoxin lipopolysaccharide significantly enhanced hBMVEC-mediated C6 glioma sCp gene expression. CONCLUSION: hBMVEC influence the gene expression of neighboring C6 glioma sCp. This change in gene expression is mediated by the secretion of IL-1ß and IL-6 from hBMVEC. Furthermore, the hBMVEC-induced increase in neighboring C6 glioma sCp gene expression leads to an increased rate of hBMVEC iron efflux. Taken together, our results indicate that hBMVEC-secreted cytokine activity increases the gene expression of neighboring C6 glioma sCp, which reciprocally acts on basolateral hBMVEC Fpn to enhance brain iron import.


Assuntos
Neoplasias Encefálicas/metabolismo , Ceruloplasmina/genética , Células Endoteliais/metabolismo , Glioblastoma/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/citologia , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Ceruloplasmina/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Imidazóis/farmacologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Ferro/metabolismo , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Ratos
7.
Biometals ; 25(4): 665-75, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22434419

RESUMO

The mechanism(s) by which iron in blood is transported across the blood-brain barrier (BBB) remains controversial. Here we have examined the first step of this trans-cellular pathway, namely the mechanism(s) of iron uptake into human brain microvascular endothelial cells (hBMVEC). We show that hBMVEC actively reduce non-transferrin bound Fe(III) (NTBI) and transferrin-bound Fe(III) (TBI); this activity is associated with one or more ferrireductases. Efficient, exo-cytoplasmic ferri-reduction from TBI is dependent upon transferrin receptor (TfR), also. Blocking holo-Tf binding with an anti-TfR antibody significantly decreases the reduction of iron from transferrin by hBMVEC, suggesting that holo-Tf needs to bind to TfR in order for efficient reduction to occur. Ferri-reduction from TBI significantly decreases when hBMVEC are pre-treated with Pt(II), an inhibitor of cell surface reductase activity. Uptake of (59)Fe from (59)Fe-Tf by endothelial cells is inhibited by 50 % when ferrozine is added to solution; in contrast, no inhibition occurs when cells are alkalinized with NH(4)Cl. This indicates that the iron reduced from holo-transferrin at the plasma membrane accounts for at least 50 % of the iron uptake observed. hBMVEC-dependent reduction and uptake of NTBI utilizes a Pt(II)-insensitive reductase. Reductase-independent uptake of Fe(II) by hBMVEC is inhibited up to 50 % by Zn(II) and/or Mn(II) by a saturable process suggesting that redundant Fe(II) transporters exist in the hBMVEC plasma membrane. These results are the first to demonstrate multiple mechanism(s) of TBI and NTBI reduction and uptake by endothelial cells (EC) of the BBB.


Assuntos
Células Endoteliais/metabolismo , Ferro/metabolismo , Barreira Hematoencefálica/metabolismo , Western Blotting , Células CACO-2 , Proteínas de Transporte de Cátions/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Grupo dos Citocromos b/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Oxirredutases/metabolismo , Transferrina/metabolismo
8.
Front Mol Neurosci ; 8: 31, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26236187

RESUMO

The transcellular trafficking of iron from the blood into the brain interstitium depends on iron uptake proteins in the apical membrane of brain microvascular capillary endothelial cells and efflux proteins at the basolateral, abluminal membrane. In this review, we discuss the three mechanisms by which these cells take-up iron from the blood and the sole mechanism by which they efflux this iron into the abluminal space. We then focus on the regulation of this efflux pathway by exocrine factors that are released from neighboring astrocytes. Also discussed are the cytokines secreted by capillary cells that regulate the expression of these glial cell signals. Among the interstitial factors that regulate iron efflux into the brain is the Amyloid precursor protein (APP). The role of this amyliodogenic species in brain iron metabolism is discussed. Last, we speculate on the potential relationship between iron transport at the blood-brain barrier and neurological disorders associated with iron mismanagement.

9.
PLoS One ; 9(2): e89003, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24533165

RESUMO

We have used an in vitro model system to probe the iron transport pathway across the brain microvascular endothelial cells (BMVEC) of the blood-brain barrier (BBB). This model consists of human BMVEC (hBMVEC) and C6 glioma cells (as an astrocytic cell line) grown in a transwell, a cell culture system commonly used to quantify metabolite flux across a cell-derived barrier. We found that iron efflux from hBMVEC through the ferrous iron permease ferroportin (Fpn) was stimulated by secretion of the soluble form of the multi-copper ferroxidase, ceruloplasmin (sCp) from the co-cultured C6 cells. Reciprocally, expression of sCp mRNA in the C6 cells was increased by neighboring hBMVEC. In addition, data indicate that C6 cell-secreted hepcidin stimulates internalization of hBMVEC Fpn but only when the end-feet projections characteristic of this glia-derived cell line are proximal to the endothelial cells. This hepcidin-dependent loss of Fpn correlated with knock-down of iron efflux from the hBMVEC; this result was consistent with the mechanism by which hepcidin regulates iron efflux in mammalian cells. In summary, the data support a model of iron trafficking across the BBB in which the capillary endothelium induce the underlying astrocytes to produce the ferroxidase activity needed to support Fpn-mediated iron efflux. Reciprocally, astrocyte proximity modulates the effective concentration of hepcidin at the endothelial cell membrane and thus the surface expression of hBMVEC Fpn. These results are independent of the source of hBMVEC iron (transferrin or non-transferrin bound) indicating that the model developed here is broadly applicable to brain iron homeostasis.


Assuntos
Encéfalo/irrigação sanguínea , Ceruloplasmina/metabolismo , Células Endoteliais/metabolismo , Hepcidinas/metabolismo , Ferro/metabolismo , Microvasos/citologia , Neuroglia/metabolismo , Astrócitos/citologia , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Ceruloplasmina/química , Células Endoteliais/citologia , Hepcidinas/genética , Humanos , Neuroglia/citologia , Comunicação Parácrina , Transporte Proteico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Solubilidade , Ubiquitinação , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA