Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oxid Med Cell Longev ; 2019: 4721950, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781336

RESUMO

Several neurodegenerative disorders exhibit selective vulnerability, with subsets of neurons more affected than others, possibly because of the high expression of an altered gene or the presence of particular features that make them more susceptible to insults. On the other hand, resilient neurons may display the ability to develop antioxidant defenses, particularly in diseases of mitochondrial origin, where oxidative stress might contribute to the neurodegenerative process. In this work, we investigated the oxidative stress response of embryonic fibroblasts and cortical neurons obtained from Afg3l2-KO mice. AFG3L2 encodes a subunit of a protease complex that is expressed in mitochondria and acts as both quality control and regulatory enzyme affecting respiration and mitochondrial dynamics. When cells were subjected to an acute oxidative stress protocol, the survival of AFG3L2-KO MEFs was not significantly influenced and was comparable to that of WT; however, the basal level of the antioxidant molecule glutathione was higher. Indeed, glutathione depletion strongly affected the viability of KO, but not of WT MEF, thereby indicating that oxidative stress is more elevated in KO MEF even though well controlled by glutathione. On the other hand, when cortical KO neurons were put in culture, they immediately appeared more vulnerable than WT to the acute oxidative stress condition, but after few days in vitro, the situation was reversed with KO neurons being more resistant than WT to acute stress. This compensatory, protective competence was not due to the upregulation of glutathione, rather of two mitochondrial antioxidant proteins: superoxide dismutase 2 and, at an even higher level, peroxiredoxin 3. This body of evidence sheds light on the capability of neurons to activate neuroprotective pathways and points the attention to peroxiredoxin 3, an antioxidant enzyme that might be critical for neuronal survival also in other disorders affecting mitochondria.


Assuntos
Proteases Dependentes de ATP/deficiência , ATPases Associadas a Diversas Atividades Celulares/deficiência , Córtex Cerebral/enzimologia , Regulação Enzimológica da Expressão Gênica , Doenças Neurodegenerativas/enzimologia , Neurônios/enzimologia , Estresse Oxidativo , Peroxirredoxina III/biossíntese , Regulação para Cima , Proteases Dependentes de ATP/metabolismo , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Animais , Sobrevivência Celular/genética , Córtex Cerebral/patologia , Camundongos , Camundongos Knockout , Mitocôndrias/enzimologia , Mitocôndrias/genética , Mitocôndrias/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Peroxirredoxina III/genética
2.
Hum Mol Genet ; 25(22): 4847-4855, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28175303

RESUMO

We employed induced pluripotent stem cell (iPSC)-derived neurons obtained from Friedreich ataxia (FRDA) patients and healthy subjects, FRDA neurons and CT neurons, respectively, to unveil phenotypic alterations related to frataxin (FXN) deficiency and investigate if they can be reversed by treatments that upregulate FXN. FRDA and control iPSCs were equally capable of differentiating into a neuronal or astrocytic phenotype. FRDA neurons showed lower levels of iron­sulfur (Fe­S) and lipoic acid-containing proteins, higher labile iron pool (LIP), higher expression of mitochondrial superoxide dismutase (SOD2), increased reactive oxygen species (ROS) and lower reduced glutathione (GSH) levels, and enhanced sensitivity to oxidants compared with CT neurons, indicating deficient Fe­S cluster biogenesis, altered iron metabolism, and oxidative stress. Treatment with the benzamide HDAC inhibitor 109 significantly upregulated FXN expression and increased Fe­S and lipoic acid-containing protein levels, downregulated SOD2 levels, normalized LIP and ROS levels, and almost fully protected FRDA neurons from oxidative stress-mediated cell death. Our findings suggest that correction of FXN deficiency may not only stop disease progression, but also lead to clinical improvement by rescuing still surviving, but dysfunctional neurons.


Assuntos
Inibidores de Histona Desacetilases/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Benzamidas/farmacologia , Ataxia de Friedreich/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas Ferro-Enxofre/metabolismo , Mitocôndrias/metabolismo , Neurônios/citologia , Estresse Oxidativo/fisiologia , Fenótipo , Superóxido Dismutase/metabolismo , Ácido Tióctico/metabolismo , Frataxina
3.
Front Mol Neurosci ; 8: 18, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26089776

RESUMO

Iron plays a fundamental role in the development of the central nervous system (CNS) as well as in several neuronal functions including synaptic plasticity. Accordingly, neuronal iron supply is tightly controlled: it depends not only on transferrin-bound iron but also on non-transferrin-bound iron (NTBI), which represents a relevant quote of the iron physiologically present in the cerebrospinal fluid (CSF). Different calcium permeable channels as well as the divalent metal transporter 1 (DMT1) have been proposed to sustain NTBI entry in neurons and astrocytes even though it remains an open issue. In both cases, it emerges that the control of iron entry is tightly linked to synaptic activity. The iron-induced oxidative tone can, in physiological conditions, positively influence the calcium levels and thus the synaptic plasticity. On the other hand, an excess of iron, with the ensuing uncontrolled production of reactive oxygen species (ROS), is detrimental for neuronal survival. A protective mechanism can be played by astrocytes that, more resistant to oxidative stress, can uptake iron, thereby buffering its concentration in the synaptic environment. This competence is potentiated when astrocytes undergo activation during neuroinflammation and neurodegenerative processes. In this minireview we focus on the mechanisms responsible for NTBI entry in neurons and astrocytes and on how they can be modulated during synaptic activity. Finally, we speculate on the relevance they may have in both physiological and pathological conditions.

4.
J Neuroinflammation ; 10: 130, 2013 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-24160637

RESUMO

BACKGROUND: Astrocytes respond to local insults within the brain and the spinal cord with important changes in their phenotype. This process, overall known as "activation", is observed upon proinflammatory stimulation and leads astrocytes to acquire either a detrimental phenotype, thereby contributing to the neurodegenerative process, or a protective phenotype, thus supporting neuronal survival. Within the mechanisms responsible for inflammatory neurodegeneration, oxidative stress plays a major role and has recently been recognized to be heavily influenced by changes in cytosolic iron levels. In this work, we investigated how activation affects the competence of astrocytes to handle iron overload and the ensuing oxidative stress. METHODS: Cultures of pure cortical astrocytes were preincubated with proinflammatory cytokines (interleukin-1ß and tumor necrosis factor α) or conditioned medium from lipopolysaccharide-activated microglia to promote activation and then exposed to a protocol of iron overload. RESULTS: We demonstrate that activated astrocytes display an efficient protection against iron-mediated oxidative stress and cell death. Based on this evidence, we performed a comprehensive biochemical and molecular analysis, including a transcriptomic approach, to identify the molecular basis of this resistance. CONCLUSIONS: We propose the protective phenotype acquired after activation not to involve the most common astrocytic antioxidant pathway, based on the Nrf2 transcription factor, but to result from a complex change in the expression and activity of several genes involved in the control of cellular redox state.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Estresse Oxidativo/fisiologia , Animais , Western Blotting , Ferro/metabolismo , Fenótipo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
5.
Biochim Biophys Acta ; 1832(8): 1326-33, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23583428

RESUMO

Astrocytes play a crucial role in proper iron handling within the central nervous system. This competence can be fundamental, particularly during neuroinflammation, and neurodegenerative processes, where an increase in iron content can favor oxidative stress, thereby worsening disease progression. Under these pathological conditions, astrocytes undergo a process of activation that confers them either a beneficial or a detrimental role on neuronal survival. Our work investigates the mechanisms of iron entry in cultures of quiescent and activated hippocampal astrocytes. Our data confirm that the main source of iron is the non-transferrin-bound iron (NTBI) and show the involvement of two different routes for its entry: the resident transient receptor potential (TRP) channels in quiescent astrocytes and the de novo expressed divalent metal transporter 1 (DMT1) in activated astrocytes, which accounts for a potentiation of iron entry. Overall, our data suggest that at rest, but even more after activation, astrocytes have the potential to buffer the excess of iron, thereby protecting neurons from iron overload. These findings further extend our understanding of the protective role of astrocytes under the conditions of iron-mediated oxidative stress observed in several neurodegenerative conditions.


Assuntos
Astrócitos/metabolismo , Compostos Férricos/farmacocinética , Compostos Ferrosos/farmacocinética , Inflamação/metabolismo , Ferro/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Hipocampo/metabolismo , Neurônios/metabolismo , Estresse Oxidativo/fisiologia , Ratos , Ratos Sprague-Dawley , Transferrina/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo
6.
J Neurochem ; 120(2): 269-78, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22121954

RESUMO

The divalent metal transporter 1 (DMT1) is the best characterized Fe²âº transporter involved in cellular iron uptake in mammals. Four possible isoforms have been identified as a result of alternative promoter (DMT1-1A and DMT1-1B) and alternative splicing involving the C-terminus and producing transcripts with or without an iron responsive element [DMT1-IRE⁺ and DMT1-IRE⁻, respectively]. Despite the general importance of DMT1 in controlling iron homeostasis, the distribution and the role of the transporter in the CNS is still controversial. In this study, we characterize the expression of DMT1 in hippocampal neurons and astrocytes. We found that the main isoform endogenously expressed is DMT1-1B/IRE⁺, which shows cytoplasmic distribution, colocalization with late endosome/lysosome markers and iron regulation, as expected from the presence of an iron responsive element. Our results also show that DMT1-1B/IRE⁺ isoform does not sustain iron entry, even after its neuronal over-expression. Overall, our results argue against a physiological role of the endogenous DMT1 in neuronal iron uptake but do not exclude that, under pathological conditions, the expression of other DMT1 isoforms might contribute to iron overload.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Regulação da Expressão Gênica/fisiologia , Hipocampo/citologia , Ferro/metabolismo , Neurônios/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Fracionamento Celular/métodos , Células Cultivadas , Desferroxamina/farmacologia , Duodeno/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Ferro/farmacologia , Proteínas de Membrana Lisossomal/metabolismo , Microscopia Confocal , RNA Mensageiro , Ratos , Ratos Sprague-Dawley , Receptores da Transferrina/metabolismo , Sideróforos/farmacologia , Transfecção/métodos , Transferrina/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Gravação em Vídeo
7.
J Neurosci ; 31(50): 18568-77, 2011 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-22171055

RESUMO

Parkinson's disease is a neurodegenerative disorder characterized by oxidative stress and CNS iron deposition. Ceruloplasmin is an extracellular ferroxidase that regulates cellular iron loading and export, and hence protects tissues from oxidative damage. Using two-dimensional electrophoresis, we investigated ceruloplasmin patterns in the CSF of human Parkinson's disease patients. Parkinson's disease ceruloplasmin profiles proved more acidic than those found in healthy controls and in other human neurological diseases (peripheral neuropathies, amyotrophic lateral sclerosis, and Alzheimer's disease); degrees of acidity correlated with patients' pathological grading. Applying an unsupervised pattern recognition procedure to the two-dimensional electrophoresis images, we identified representative pathological clusters. In vitro oxidation of CSF in two-dimensional electrophoresis generated a ceruloplasmin shift resembling that observed in Parkinson's disease and co-occurred with an increase in protein carbonylation. Likewise, increased protein carbonylation was observed in Parkinson's disease CSF, and the same modification was directly identified in these samples on ceruloplasmin. These results indicate that ceruloplasmin oxidation contributes to pattern modification in Parkinson's disease. From the functional point of view, ceruloplasmin oxidation caused a decrease in ferroxidase activity, which in turn promotes intracellular iron retention in neuronal cell lines as well as in primary neurons, which are more sensitive to iron accumulation. Accordingly, the presence of oxidized ceruloplasmin in Parkinson's disease CSF might be used as a marker for oxidative damage and might provide new insights into the underlying pathological mechanisms.


Assuntos
Ceruloplasmina/líquido cefalorraquidiano , Ceruloplasmina/metabolismo , Ferro/metabolismo , Estresse Oxidativo/fisiologia , Doença de Parkinson/líquido cefalorraquidiano , Idoso , Idoso de 80 Anos ou mais , Animais , Astrócitos/metabolismo , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurônios/metabolismo , Oxirredução , Ratos , Ratos Sprague-Dawley
8.
Eur J Neurosci ; 33(2): 236-43, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21073551

RESUMO

BACE1 and BACE2 are two closely related membrane-bound aspartic proteases. BACE1 is widely recognized as the neuronal ß-secretase that cleaves the amyloid-ß precursor protein, thus allowing the production of amyloid-ß, i.e. the peptide that has been proposed to trigger the neurodegenerative process in Alzheimer's disease. BACE2 has ubiquitous expression and its physiological and pathological role is still unclear. In light of a possible role of glial cells in the accumulation of amyloid-ß in brain, we have investigated the expression of these two enzymes in primary cultures of astrocytes. We show that astrocytes possess ß-secretase activity and produce amyloid-ß because of the activity of BACE2, but not BACE1, the expression of which is blocked at the translational level. Finally, our data demonstrate that changes in the astrocytic phenotype during neuroinflammation can produce both a negative as well as a positive modulation of ß-secretase activity, also depending on the differential responsivity of the brain regions.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Astrócitos/enzimologia , Regulação da Expressão Gênica , Biossíntese de Proteínas , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Astrócitos/citologia , Células Cultivadas , Hipocampo/citologia , Humanos , Neurônios/citologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley
9.
Aging Cell ; 10(1): 172-83, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21108725

RESUMO

The characterization of iron handling in neurons is still lacking, with contradictory and incomplete results. In particular, the relevance of non-transferrin-bound iron (NTBI), under physiologic conditions, during aging and in neurodegenerative disorders, is undetermined. This study investigates the mechanisms underlying NTBI entry into primary hippocampal neurons and evaluates the consequence of iron elevation on neuronal viability. Fluorescence-based single cell analysis revealed that an increase in extracellular free Fe(2+) (the main component of NTBI pool) is sufficient to promote Fe(2+) entry and that activation of either N-methyl-d-aspartate receptors (NMDARs) or voltage operated calcium channels (VOCCs) significantly potentiates this pathway, independently of changes in intracellular Ca(2+) concentration ([Ca(2+) ](i) ). The enhancement of Fe(2+) influx was accompanied by a corresponding elevation of reactive oxygen species (ROS) production and higher susceptibility of neurons to death. Interestingly, iron vulnerability increased in aged cultures. Scavenging of mitochondrial ROS was the most powerful protective treatment against iron overload, being able to preserve the mitochondrial membrane potential and to safeguard the morphologic integrity of these organelles. Overall, we demonstrate for the first time that Fe(2+) and Ca(2+) compete for common routes (i.e. NMDARs and different types of VOCCs) to enter primary neurons. These iron entry pathways are not controlled by the intracellular iron level and can be harmful for neurons during aging and in conditions of elevated NTBI levels. Finally, our data draw the attention to mitochondria as a potential target for the treatment of the neurodegenerative processes induced by iron dysmetabolism.


Assuntos
Transporte de Íons/fisiologia , Ferro/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Técnicas de Cultura de Células , Morte Celular/fisiologia , Células Cultivadas , Hipocampo/citologia , Hipocampo/metabolismo , Distúrbios do Metabolismo do Ferro/prevenção & controle , Potencial da Membrana Mitocondrial , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Análise de Célula Única
10.
Biochem Soc Trans ; 36(Pt 6): 1309-12, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021546

RESUMO

Iron and calcium are required for general cellular functions, as well as for specific neuronal-related activities. However, a pathological increase in their levels favours oxidative stress and mitochondrial damage, leading to neuronal death. Neurodegeneration can thus be determined by alterations in ionic homoeostasis and/or pro-oxidative-antioxidative equilibrium, two conditions that vary significantly in different kinds of brain cell and also with aging. In the present review, we re-evaluate recent data on NTBI (non-transferrin bound iron) uptake that suggest a strict interplay with the mechanisms of calcium control. In particular, we focus on the use of common entry pathways and on the way cytosolic calcium can modulate iron entry and determine its intracellular accumulation.


Assuntos
Cálcio/metabolismo , Sistema Nervoso Central/metabolismo , Doença , Saúde , Ferro/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA