Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 25(5)2024 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-38474295

RESUMO

Astroglia constitute the largest group of glial cells and are involved in numerous actions that are critical to neuronal development and functioning, such as maintaining the blood-brain barrier, forming synapses, supporting neurons with nutrients and trophic factors, and protecting them from injury. These properties are deeply affected in the course of many neurodegenerative diseases, including tauopathies, often before the onset of the disease. In this respect, the transfer of essential amino acids such as glutamate and glutamine between neurons and astrocytes in the glutamate-glutamine cycle (GGC) is one example. In this review, we focus on the GGC and the disruption of this cycle in tau-dependent neurodegeneration. A profound understanding of the complex functions of the GGC and, in the broader context, searching for dysfunctions in communication pathways between astrocytes and neurons via GGC in health and disease, is of critical significance for the development of novel mechanism-based therapies for neurodegenerative disorders.


Assuntos
Astrócitos , Ácido Glutâmico , Glutamina , Doenças Neurodegenerativas , Neurônios , Humanos , Astrócitos/metabolismo , Barreira Hematoencefálica/metabolismo , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Comunicação Celular
2.
Int J Mol Sci ; 24(17)2023 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-37685956

RESUMO

Glutamine (Gln), glutamate (Glu), and γ-amino butyric acid (GABA) are essential amino acids for brain metabolism and function. Astrocyte-derived Gln is the precursor for the two most important neurotransmitters in the central nervous system (CNS), which are the excitatory neurotransmitter Glu and the inhibitory neurotransmitter GABA. In addition to their roles in neurotransmission, these amino acids can be used as alternative substrates in brain metabolism that enable metabolic coupling between astrocytes and neurons in the glutamate-glutamine cycle (GGC). The disturbed homeostasis of these amino acids within the tripartite synapse may be involved in the pathogenesis of various neurological diseases. Interactions between astrocytes and neurons in terms of Gln, Glu, and GABA homeostasis were studied in different phases of experimental allergic encephalomyelitis (EAE) in Lewis rats. The results of the study showed a decrease in the transport (uptake and release) of Gln and GABA in both neuronal and astrocyte-derived fractions. These effects were fully or partially reversed when the EAE rats were treated with memantine, a NMDA receptor antagonist. Changes in the expression and activity of selected glutamine/glutamate metabolizing enzymes, such as glutamine synthase (GS) and phosphate-activated glutaminase (PAG), which were affected by memantine, were observed in different phases of EAE. The results suggested perturbed homeostasis of Gln, Glu, and GABA during EAE, which may indicate alterations in neuron-astrocyte coupling and dysfunction of the tripartite synapse. Memantine appears to partially regulate the disturbed relationships between Gln, Glu, and GABA.


Assuntos
Antifibrinolíticos , Encefalomielite Autoimune Experimental , Animais , Ratos , Ratos Endogâmicos Lew , Encefalomielite Autoimune Experimental/tratamento farmacológico , Glutamina , Memantina/farmacologia , Memantina/uso terapêutico , Encéfalo , Ácido Glutâmico , Ácido gama-Aminobutírico , Aminoácidos , Homeostase
3.
Neurochem Int ; 168: 105551, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37295680

RESUMO

Tau-dependent neurodegeneration is accompanied by astrocytosis in a mouse trans-genic model, which replicates the neuropathological characteristic of tauopathy and other human neurodegenerative disorders where astrocyte activation precedes neuronal loss and is associated with disease progression. This indicates an important role of astrocytes in the development of the disease. Astrocytes derived from a transgenic mouse model expressing human Tau, exhibit changes in cellular markers of astrocyte neuroprotective function related to the glutamate-glutamine cycle (GGC), representing a key part of astrocyte-neuron integrity. Here, we focused on investigating the functional properties of key GGC components involved in the astrocyte-neuron network associated with Tau pathology in vitro. Mutant recombinant Tau (rTau) carrying the P301L mutation was added to the neuronal cultures, with or without control astrocyte-conditioned medium (ACM), to study glutamine translocation through the GGC. We demonstrated that mutant Tau in vitro induces neuronal degeneration, while control astrocytes response in neuroprotective way by preventing neurodegeneration. In parallel with this observation, we noticed the Tau-dependent decline of neuronal microtubule associated protein 2 (MAP2), followed by changes in glutamine (Gln) transport. Exposure to rTau decreases sodium-dependent Gln uptake in neurons and that effect was reversed when cells were co-incubated with control ACM after induction of rTau dependent pathology. Further, we found that neuronal Na+-dependent system A is the most specific system that is affected under rTau exposure. In addition, in rTau-treated astrocytes total Na+-dependent uptake of Gln, which is mediated by the N system, increases. Altogether, our study suggest mechanisms operating in Tau pathology may be related to the alterations in glutamine transport and recycling that affect neuronal-astrocytic integrity.


Assuntos
Astrócitos , Tauopatias , Animais , Camundongos , Astrócitos/metabolismo , Modelos Animais de Doenças , Glutamina/metabolismo , Camundongos Transgênicos , Neurônios/metabolismo , Proteínas tau/metabolismo , Tauopatias/metabolismo
4.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34768760

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is an animal model most commonly used in research on the pathomechanisms of multiple sclerosis (MS). The inflammatory processes, glutamate excitotoxicity, and oxidative stress have been proposed as determinants accompanying demyelination and neuronal degeneration during the course of MS/EAE. The aim of the current study was to characterize the role of NMDA receptors in the induction of oxidative stress during the course of EAE. The effect of memantine, the uncompetitive NMDA receptor antagonist, on modulation of neurological deficits and oxidative stress in EAE rats was analyzed using several experimental approaches. We demonstrated that the expression of antioxidative enzymes (superoxide dismutases SOD1 and SOD2) were elevated in EAE rat brains. Under the same experimental conditions, we observed alterations in oxidative stress markers such as increased levels of malondialdehyde (MDA) and decreased levels of sulfhydryl (-SH) groups, both protein and non-protein (indicating protein damage), and a decline in reduced glutathione. Importantly, pharmacological inhibition of ionotropic NMDA glutamate receptors by their antagonist memantine improved the physical activity of EAE rats, alleviated neurological deficits such as paralysis of tail and hind limbs, and modulated oxidative stress parameters (MDA, -SH groups, SOD's). Furthermore, the current therapy aiming to suppress NMDAR-induced oxidative stress was partially effective when NMDAR's antagonist was administered at an early (asymptomatic) stage of EAE.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encefalomielite Autoimune Experimental/tratamento farmacológico , Antagonistas de Aminoácidos Excitatórios/farmacologia , Memantina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Feminino , Memantina/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Ratos Endogâmicos Lew , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
5.
Eur J Neurosci ; 54(9): 7377-7404, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34561918

RESUMO

Multiple sclerosis (MS) is an immune-mediated demyelinating disease of the central nervous system which leads to neurological dysfunctions and severe disabilities. MS pathology is characterised by damage of the blood-brain barrier and infiltration of autoreactive T cells that overactivate glial cells, thereby initiating neuroinflammation accompanied by the formation of demyelinating plaques and neurodegeneration. Clinical deficits in this multifactorial disease depend on the progression of myelin loss, the stage of inflammation, the status of axons and the activity of oligodendrocyte precursor cells (OPCs). Despite significant progress in the treatment of MS, current therapies remain limited and new approaches are highly desirable. Nanosystems based on liposomes and nanoparticles are among some of the more noteworthy therapeutic strategies being investigated. Applications of nanosystems alone or as drug carriers in animal models of MS have been found to successfully alleviate the symptoms of the disease and exert anti-inflammatory potential. Exosomes are a specific type of nanosystem based on nanometre-sized extracellular vesicles released by different cells which exhibit important healing features. Exosomes contain an array of anti-inflammatory and neuroprotective agents which may contribute to modulation of the immune system as well as promoting remyelination and tissue repair. In this review, opportunities to use nanosystems against progression of MS will be discussed in context of cell-specific pathologies associated with MS.


Assuntos
Exossomos , Esclerose Múltipla , Remielinização , Animais , Sistema Nervoso Central , Esclerose Múltipla/tratamento farmacológico , Bainha de Mielina , Neuroglia , Oligodendroglia
6.
Int J Mol Sci ; 22(16)2021 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-34445109

RESUMO

Multiple sclerosis (MS) is an autoimmune inflammatory disease of the central nervous system that leads to the progressive disability of patients. A characteristic feature of the disease is the presence of focal demyelinating lesions accompanied by an inflammatory reaction. Interactions between autoreactive immune cells and glia cells are considered as a central mechanism underlying the pathology of MS. A glia-mediated inflammatory reaction followed by overproduction of free radicals and generation of glutamate-induced excitotoxicity promotes oligodendrocyte injury, contributing to demyelination and subsequent neurodegeneration. Activation of purinergic signaling, in particular P2X7 receptor-mediated signaling, in astrocytes and microglia is an important causative factor in these pathological processes. This review discusses the role of astroglial and microglial cells, and in particular glial P2X7 receptors, in inducing MS-related neuroinflammatory events, highlighting the importance of P2X7R-mediated molecular pathways in MS pathology and identifying these receptors as a potential therapeutic target.


Assuntos
Astrócitos/metabolismo , Sistema Nervoso Central/metabolismo , Inflamação/metabolismo , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Receptores Purinérgicos P2X7/metabolismo , Animais , Humanos , Neuroglia/metabolismo , Transdução de Sinais/fisiologia
7.
Neural Regen Res ; 16(2): 218-222, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32859767

RESUMO

Astrocytes are integral components of the central nervous system, where they are involved in numerous functions critical for neuronal development and functioning, including maintenance of blood-brain barrier, formation of synapses, supporting neurons with nutrients and trophic factors, and protecting them from injury. These roles are markedly affected in the course of chronic neurodegenerative disorders, often before the onset of the disease. In this review, we summarize the recent findings supporting the hypothesis that astrocytes play a fundamental role in the processes contributing to neurodegeneration. We focus on α-synucleinopathies and tauopathies as the most common neurodegenerative diseases. The mechanisms implicated in the development and progression of these disorders appear not to be exclusively neuronal, but are often related to the astrocytic-neuronal integrity and the response of astrocytes to the altered microglial function. A profound understanding of the multifaceted functions of astrocytes and identification of their communication pathways with neurons and microglia in health and in the disease is of critical significance for the development of novel mechanism-based therapies against neurodegenerative disorders.

8.
Int J Mol Sci ; 21(23)2020 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-33256007

RESUMO

Due to strong antimicrobial properties, silver nanoparticles (AgNPs) are used in a wide range of medical and consumer products, including those dedicated for infants and children. While AgNPs are known to exert neurotoxic effects, current knowledge concerning their impact on the developing brain is scarce. During investigations of mechanisms of neurotoxicity in immature rats, we studied the influence of AgNPs on glutamate transporter systems which are involved in regulation of extracellular concentration of glutamate, an excitotoxic amino acid, and compared it with positive control-Ag citrate. We identified significant deposition of AgNPs in brain tissue of exposed rats over the post-exposure time. Ultrastructural alterations in endoplasmic reticulum (ER) and Golgi complexes were observed in neurons of AgNP-exposed rats, which are characteristics of ER stress. These changes presumably underlie substantial long-lasting downregulation of neuronal glutamate transporter EAAC1, which was noted in AgNP-exposed rats. Conversely, the expression of astroglial glutamate transporters GLT-1 and GLAST was not affected by exposure to AgNPs, but the activity of the transporters was diminished. These results indicate that even low doses of AgNPs administered during an early stage of life create a substantial risk for health of immature organisms. Hence, the safety of AgNP-containing products for infants and children should be carefully considered.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Encéfalo/metabolismo , Nanopartículas Metálicas/toxicidade , Prata/toxicidade , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/ultraestrutura , Encéfalo/efeitos dos fármacos , Transportador 3 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Ratos , Prata/sangue , Sódio/metabolismo , Fatores de Tempo
9.
Biochem J ; 476(22): 3493-3504, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31774919

RESUMO

Astrocytes, by maintaining an optimal environment for neuronal function, play a critical role in proper function of mammalian nervous system. They regulate synaptic transmission and plasticity and protect neurons against toxic insults. Astrocytes and neurons interact actively via glutamine-glutamate cycle (GGC) that supports neuronal metabolic demands and neurotransmission. GGC deficiency may be involved in different diseases of the brain, where impaired astrocytic control of glutamate homeostasis contributes to neuronal dysfunction. This includes tau-dependent neurodegeneration, where astrocytes lose key molecules involved in regulation of glutamate/glutamine homeostasis, neuronal survival and synaptogenesis. Astrocytic dysfunction in tauopathy appears to precede neurodegeneration and overt tau neuropathology such as phosphorylation, aggregation and formation of neurofibrillary tangles. In this review, we summarize recent studies demonstrating that activation of astrocytes is strictly associated with neurodegenerative processes including those involved in tau related pathology. We propose that astrocytic dysfunction, by disrupting the proper neuron-glia signalling early in the disease, significantly contributes to tauopathy pathogenesis.


Assuntos
Astrócitos/metabolismo , Doenças Neurodegenerativas/metabolismo , Proteínas tau/metabolismo , Animais , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Humanos , Doenças Neurodegenerativas/genética , Neurônios/metabolismo
10.
Neurochem Res ; 44(11): 2449-2459, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31571097

RESUMO

Manganese (Mn) overexposure is a public health concern due to its widespread industrial usage and the risk for environmental contamination. The clinical symptoms of Mn neurotoxicity, or manganism, share several pathological features of Parkinson's disease (PD). Biologically, Mn is an essential trace element, and Mn in the brain is preferentially localized in astrocytes. This review summarizes the role of astrocytes in Mn-induced neurotoxicity, specifically on the role of neurotransmitter recycling, neuroinflammation, and genetics. Mn overexposure can dysregulate astrocytic cycling of glutamine (Gln) and glutamate (Glu), which is the basis for Mn-induced excitotoxic neuronal injury. In addition, reactive astrocytes are important mediators of Mn-induced neuronal damage by potentiating neuroinflammation. Genetic studies, including those with Caenorhabditis elegans (C. elegans) have uncovered several genes associated with Mn neurotoxicity. Though we have yet to fully understand the role of astrocytes in the pathologic changes characteristic of manganism, significant strides have been made over the last two decades in deciphering the role of astrocytes in Mn-induced neurotoxicity and neurodegeneration.


Assuntos
Astrócitos/metabolismo , Intoxicação por Manganês/fisiopatologia , Manganês/toxicidade , Animais , Astrócitos/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Humanos , Neurônios/metabolismo
11.
Acta Neuropathol Commun ; 5(1): 89, 2017 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-29187256

RESUMO

Microtubule-associated protein tau aggregates constitute the characteristic neuropathological features of several neurodegenerative diseases grouped under the name of tauopathies. It is now clear that the process of tau aggregation is associated with neurodegeneration. Several transgenic tau mouse models have been developed where tau progressively aggregates, causing neuronal death. Previously we have shown that transplantation of astrocytes in P301S tau transgenic mice rescues cortical neuron death, implying that the endogenous astrocytes are deficient in survival support. We now show that the gliosis markers Glial fibrillary acidic protein (GFAP) and S100 calcium-binding protein B (S100ß) are elevated in brains from P301S tau mice compared to control C57Bl/6 mice whereas the expression of proteins involved in glutamine/glutamate metabolism are reduced, pointing to a functional deficit. To test whether astrocytes from P301S mice are intrinsically deficient, we co-cultured astrocytes and neurons from control and P301S mice. Significantly more C57-derived and P301S-derived neurons survived when cells were cultured with C57-derived astrocytes or astrocyte conditioned medium (C57ACM) than with P301S-derived astrocytes or astrocyte conditioned medium (P301SACM), or ACM from P301L tau mice, where the transgene is also specifically expressed in neurons. The astrocytic alterations developed in mice during the first postnatal week of life. In addition, P301SACM significantly decreased presynaptic (synaptophysin, SNP) and postsynaptic (postsynaptic density protein 95, PSD95) protein expression in cortical neuron cultures whereas C57ACM enhanced these markers. Since thrombospondin 1 (TSP-1) is a major survival and synaptogenic factor, we examined whether TSP-1 is deficient in P301S mouse brains and ACM. Significantly less TSP-1 was expressed in the brains of P301S tau mice or produced by P301S-derived astrocytes, whereas supplementation of P301SACM with TSP-1 increased its neurosupportive capacity. Our results demonstrate that P301S-derived astrocytes acquire an early functional deficiency that may explain in part the loss of cortical neurons in the P301S tau mice.


Assuntos
Astrócitos/fisiologia , Encéfalo/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Tauopatias/patologia , Animais , Animais Recém-Nascidos , Astrócitos/química , Astrócitos/patologia , Encéfalo/metabolismo , Proliferação de Células/fisiologia , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Neurônios/fisiologia , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Tauopatias/genética , Tubulina (Proteína)/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
12.
BMC Pharmacol Toxicol ; 14: 23, 2013 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-23594835

RESUMO

Astrocytes are responsible for numerous aspects of metabolic support, nutrition, control of the ion and neurotransmitter environment in central nervous system (CNS). Failure by astrocytes to support essential neuronal metabolic requirements plays a fundamental role in the pathogenesis of brain injury and the ensuing neuronal death. Astrocyte-neuron interactions play a central role in brain homeostasis, in particular via neurotransmitter recycling functions. Disruption of the glutamine (Gln)/glutamate (Glu) -γ-aminobutyric acid (GABA) cycle (GGC) between astrocytes and neurons contributes to changes in Glu-ergic and/or GABA-ergic transmission, and is associated with several neuropathological conditions, including manganese (Mn) toxicity. In this review, we discuss recent advances in support of the important roles for astrocytes in normal as well as neuropathological conditions primarily those caused by exposure to Mn.


Assuntos
Astrócitos/metabolismo , Manganês/toxicidade , Síndromes Neurotóxicas/etiologia , Animais , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Humanos , Síndromes Neurotóxicas/metabolismo
13.
Lab Chip ; 13(6): 1093-101, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23344641

RESUMO

Current techniques for mimicking the Blood-Brain Barrier (BBB) largely use incubation chambers (Transwell) separated with a filter and matrix coating to represent and to study barrier permeability. These devices have several critical shortcomings: (a) they do not reproduce critical microenvironmental parameters, primarily anatomical size or hemodynamic shear stress, (b) they often do not provide real-time visualization capability, and (c) they require a large amount of consumables. To overcome these limitations, we have developed a microfluidics based Synthetic Microvasculature model of the Blood-Brain Barrier (SyM-BBB). The SyM-BBB platform is comprised of a plastic, disposable and optically clear microfluidic chip with a microcirculation sized two-compartment chamber. The chamber is designed in such a way as to permit the realization of side-by-side apical and basolateral compartments, thereby simplifying fabrication and facilitating integration with standard instrumentation. The individually addressable apical side is seeded with endothelial cells and the basolateral side can support neuronal cells or conditioned media. In the present study, an immortalized Rat Brain Endothelial cell line (RBE4) was cultured in SyM-BBB with a perfusate of Astrocyte Conditioned Media (ACM). Biochemical analysis showed upregulation of tight junction molecules while permeation studies showed an intact BBB. Finally, transporter assay was successfully demonstrated in SyM-BBB indicating a functional model.


Assuntos
Barreira Hematoencefálica/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Linhagem Celular , Rastreamento de Células , Meios de Cultivo Condicionados/química , Dextranos/química , Técnicas Analíticas Microfluídicas/instrumentação , Modelos Biológicos , Permeabilidade , Ratos , Rodamina 123/química
14.
Neurotox Res ; 23(2): 124-30, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22878846

RESUMO

The astrocytic glutamate transporters (GLT-1, GLAST) are critical for removing excess glutamate from synaptic sites, thereby maintaining glutamate homeostasis within the brain. 17ß-Estradiol (E2) is one of the most active estrogen hormones possessing neuroprotective effects both in in vivo and in vitro models, and it has been shown to enhance astrocytic glutamate transporter function (Liang et al. in J Neurochem 80:807-814, 2002; Pawlak et al. in Brain Res Mol Brain Res 138:1-7, 2005). However, E2 is not clinically optimal for neuroprotection given its peripheral feminizing and proliferative effects; therefore, brain selective estrogen receptor modulators (neuro SERMs) (Zhao et al. in Neuroscience 132:299-311, 2005) that specifically target estrogenic mechanisms, but lack the systemic estrogen side effects offer more promising therapeutic modality for the treatment of conditions associated with excessive synaptic glutamate levels. This review highlights recent studies from our laboratory showing that E2 and SERMs effectively reverse glutamate transport inhibition in a manganese (Mn)-induced model of glutamatergic deregulation. Specifically, we discuss mechanisms by which E2 restores the expression and activity of glutamate uptake. We advance the hypothesis that E2 and related compounds, such as tamoxifen may offer a potential therapeutic modality in neurodegenerative disorders, which are characterized by altered glutamate homeostasis.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Astrócitos/efeitos dos fármacos , Estrogênios/farmacologia , Manganês/toxicidade , Animais , Astrócitos/metabolismo , Interações Medicamentosas , Regulação da Expressão Gênica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Ratos , Receptores Acoplados a Proteínas G/metabolismo
15.
J Neurochem ; 122(4): 856-67, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22708868

RESUMO

Manganese (Mn) has been implicated in the impairment of the glutamate-glutamine cycling (GGC) by deregulation of Glu and glutamine (Gln) turnover in astrocytes. Here, we have examined possible mechanisms involved in the Mn(II)-mediated disruption of Glu turnover, including those related to protein degradation, such as the proteasomal and lysosomal machinery. Our study revealed that lysosome but not proteasomal inhibition is responsible for down-regulation of the Glu transporter after Mn(II) treatment. Because protein kinase C (PKC) activation leads to the down-regulation of Glu carriers, and Mn(II) increases PKC activity, we hypothesized that the PKC signaling contributes to the Mn(II)-mediated disruption of Glu turnover. Our results show that PKC activation causes a decrease in Glu uptake and that inhibition of PKC reverses Mn(II)-dependent down-regulation of Glu influx as well as glutamate transporter 1 (GLT1) and glutamate-aspartate transporter (GLAST) protein level. Co-immunoprecipitation studies show association of GLT1 with the PKCδ and PKCα isoforms and Mn(II)-induced specific increase in PKCδ-GLT1 interaction. In addition, astrocytes transfected with shRNA against PKCδ show decreased sensitivity to Mn(II) compared with those transfected with control shRNA or shRNA targeted against PKCα. Taken together, these findings demonstrate that PKCδ signaling is involved in the Mn(II)-induced deregulation of Glu turnover in astrocytes.


Assuntos
Cloretos/toxicidade , Ácido Glutâmico/metabolismo , Glutamina/metabolismo , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Animais Recém-Nascidos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Western Blotting , Inibidores de Caspase , Caspases/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Endocitose/efeitos dos fármacos , Imunoprecipitação , Indicadores e Reagentes , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Compostos de Manganês , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Quinase C/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteína Quinase C-delta/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Translocação Genética/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos
16.
Cardiovasc Res ; 95(3): 375-84, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22673370

RESUMO

AIMS: Pulmonary arterial endothelial cells (PAECs) express the enzymes needed for generation of l-arginine from intracellular l-citrulline but do not express the enzymes needed for de novo l-citrulline synthesis. Hence, l-citrulline levels in PAECs are dependent on l-citrulline transport. Once generated, l-arginine can be converted to l-citrulline and nitric oxide (NO) by the enzyme NO synthase. We sought to determine whether hypoxia, a condition aetiologically linked to pulmonary hypertension, alters the transport of l-citrulline and the expression of the sodium-coupled neutral amino acid transporters (SNATs) in PAECs from newborn piglets. METHODS AND RESULTS: PAECs isolated from newborn piglets were cultured under normoxic and hypoxic conditions and used to measure SNAT1, 2, 3, and 5 protein expression and (14)C-l-citrulline uptake. SNAT1 protein expression was increased, while SNAT2, SNAT3, and SNAT5 expression was unaltered in hypoxic PAECs. (14)C-l-citrulline uptake was increased in hypoxic PAECs. Studies with inhibitors of System A (SNAT1/2) and System N (SNAT3/5) revealed that the increased (14)C-l-citrulline uptake was largely due to System A-mediated transport. Additional studies were performed to evaluate SNAT protein expression and l-citrulline levels in lungs of piglets with chronic hypoxia-induced pulmonary hypertension and comparable age controls. Lungs from piglets raised in chronic hypoxia exhibited greater SNAT1 expression and higher l-citrulline levels than lungs from controls. CONCLUSION: Increased SNAT1 expression and the concomitant enhanced ability to transport l-citrulline in PAECs could represent an important regulatory mechanism to counteract NO signalling impairments known to occur during the development of chronic hypoxia-induced pulmonary hypertension in newborns.


Assuntos
Sistema A de Transporte de Aminoácidos/metabolismo , Citrulina/metabolismo , Células Endoteliais/metabolismo , Hipóxia/metabolismo , Artéria Pulmonar/metabolismo , Circulação Pulmonar , Sistema A de Transporte de Aminoácidos/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Transporte Biológico , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipóxia/complicações , Hipóxia/fisiopatologia , Moduladores de Transporte de Membrana/farmacologia , Óxido Nítrico/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/fisiopatologia , Suínos , Fatores de Tempo
17.
J Biol Chem ; 287(32): 26817-28, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22645130

RESUMO

The G protein-coupled estrogen receptor GPR30 contributes to the neuroprotective effects of 17ß-estradiol (E2); however, the mechanisms associated with this protection have yet to be elucidated. Given that E2 increases astrocytic expression of glutamate transporter-1 (GLT-1), which would prevent excitotoxic-induced neuronal death, we proposed that GPR30 mediates E2 action on GLT-1 expression. To investigate this hypothesis, we examined the effects of G1, a selective agonist of GPR30, and GPR30 siRNA on astrocytic GLT-1 expression, as well as glutamate uptake in rat primary astrocytes, and explored potential signaling pathways linking GPR30 to GLT-1. G1 increased GLT-1 protein and mRNA levels, subject to regulation by both MAPK and PI3K signaling. Inhibition of TGF-α receptor suppressed the G1-induced increase in GLT-1 expression. Silencing GPR30 reduced the expression of both GLT-1 and TGF-α and abrogated the G1-induced increase in GLT-1 expression. Moreover, the G1-induced increase in GLT-1 protein expression was abolished by a protein kinase A inhibitor and an NF-κB inhibitor. G1 also enhanced cAMP response element-binding protein (CREB), as well as both NF-κB p50 and NF-κB p65 binding to the GLT-1 promoter. Finally, to model dysfunction of glutamate transporters, manganese was used, and G1 was found to attenuate manganese-induced impairment in GLT-1 protein expression and glutamate uptake. Taken together, the present data demonstrate that activation of GPR30 increases GLT-1 expression via multiple pathways, suggesting that GPR30 is worthwhile as a potential target to be explored for developing therapeutics of excitotoxic neuronal injury.


Assuntos
Astrócitos/metabolismo , Transportador 2 de Aminoácido Excitatório/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Animais , Sequência de Bases , Western Blotting , Células Cultivadas , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Primers do DNA , Inativação Gênica , Imuno-Histoquímica , Reação em Cadeia da Polimerase , Proteínas Quinases/metabolismo , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética
18.
Glia ; 60(7): 1024-36, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22488924

RESUMO

Glutamate transporter-1 (GLT-1) plays a central role in preventing excitotoxicity by removing excess glutamate from the synaptic clefts. 17ß-Estradiol (E2) and tamoxifen (TX), a selective estrogen receptor (ER) modulator, afford neuroprotection in a range of experimental models. However, the mechanisms that mediate E2 and TX neuroprotection have yet to be elucidated. We tested the hypothesis that E2 and TX enhance GLT-1 function by increasing transforming growth factor (TGF)-α expression and, thus, attenuate manganese (Mn)-induced impairment in astrocytic GLT-1 expression and glutamate uptake in rat neonatal primary astrocytes. The results showed that E2 (10 nM) and TX (1 µM) increased GLT-1 expression and reversed the Mn-induced reduction in GLT-1, both at the mRNA and protein levels. E2/TX also concomitantly reversed the Mn-induced inhibition of astrocytic glutamate uptake. E2/TX activated the GLT-1 promoter and attenuated the Mn-induced repression of the GLT-1 promoter in astrocytes. TGF-α knockdown (siRNA) abolished the E2/TX effect on GLT-1 expression, and inhibition of epidermal growth factor receptor (TGF-α receptor) suppressed the effect of E2/TX on GLT-1 expression and GLT-1 promoter activity. E2/TX also increased TGF-α mRNA and protein levels with a concomitant increase in astrocytic glutamate uptake. All ERs (ER-α, ER-ß, and G protein-coupled receptor 30) were involved in mediating E2 effects on the regulation of TGF-α, GLT-1, and glutamate uptake. These results indicate that E2/TX increases GLT-1 expression in astrocytes via TGF-α signaling, thus offering an important putative target for the development of novel therapeutics for neurological disorders.


Assuntos
Astrócitos/efeitos dos fármacos , Estradiol/farmacologia , Transportador 2 de Aminoácido Excitatório/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Transportador 2 de Aminoácido Excitatório/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno , Ratos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Fator de Crescimento Transformador alfa/genética , Regulação para Cima/fisiologia
19.
Free Radic Biol Med ; 52(6): 1067-74, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22245093

RESUMO

Excessive exposure to manganese (Mn) increases levels of oxidative stressors and proinflammatory mediators, such as cyclooxygenase-2 and prostaglandin E(2). Mn also activates nuclear factor-κB (NF-κB), an important mediator of inflammation. The signaling molecule 15-deoxy-Δ12,14-prostaglandin J(2) (15 d-PGJ(2)) is an anti-inflammatory prostaglandin. Here, we tested the hypothesis that 15 d-PGJ(2) modulates Mn-induced activation of astrocytic intracellular signaling, including NF-κB and nuclear factor erythroid 2-related factor (Nrf2), a master regulator of antioxidant transcriptional responses. The results establish that 15 d-PGJ(2) suppresses Mn-induced NF-κB activation by interacting with several signaling pathways. The PI3K/Akt pathway, which is upstream of NF-κB, plays a role in this activation, because (i) pretreatment with 15 d-PGJ(2) (10 µM for 1h) significantly (p<0.01) inhibited Mn (500 µM)-induced PI3K/Akt activation and (ii) inhibition of the PI3K/Akt pathway with LY29004 significantly (p<0.05) decreased NF-κB activation. 15 d-PGJ(2) also significantly (p<0.05) attenuated Mn-induced astrocytic NF-κB activation by inhibiting the Mn-induced phosphorylation of IκB kinase and subsequent IκB-α degradation. Because Mn-induced oxidative stress is also associated with Nrf2 activation, additional studies addressed the ability of 15 d-PGJ(2) to modulate the Nrf2 pathway. 15 d-PGJ(2) significantly (p<0.01) increased Nrf2 expression in whole-cell lysates. Consistent with its pro-oxidant properties, Mn also increased Nrf2 expression. Nevertheless, cotreatment of whole-cell lysates with both Mn and 15 d-PGJ(2) partially suppressed (p<0.01) the 15 d-PGJ(2)-induced increase in astrocytic Nrf2 protein expression. Mn treatment also decreased (p<0.001) expression of DJ-1, a Parkinson disease-associated protein and a stabilizer of Nrf2, and 15 d-PGJ(2) attenuated Mn-induced astrocytic inhibition of DJ-1 expression. Collectively, these results demonstrate that 15d-PGJ(2) exerts a protective effect in astrocytes against Mn-induced inflammation and oxidative stress by modulating the activation of the NF-κB and Nrf2 signaling pathways.


Assuntos
Astrócitos/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Oncogênicas/metabolismo , Doença de Parkinson/metabolismo , Prostaglandina D2/análogos & derivados , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Células Cultivadas , Cromonas/farmacologia , Citoproteção/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Manganês/metabolismo , Morfolinas/farmacologia , Fator 2 Relacionado a NF-E2/genética , NF-kappa B/metabolismo , Proteínas Oncogênicas/genética , Estresse Oxidativo/efeitos dos fármacos , Doença de Parkinson/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Prostaglandina D2/farmacologia , Proteína Desglicase DJ-1 , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
20.
Glia ; 59(11): 1732-43, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21812036

RESUMO

Manganese (Mn) is a trace element essential for normal human development and is required for the proper functioning of a variety of physiological processes. Chronic exposure to Mn can cause manganism, a neurodegenerative disorder resembling idiopathic Parkinson's disease (PD). Mn(II) neurotoxicity is characterized by astrocytic impairment both in the expression and activity of glutamine (Gln) transporters. Because protein kinase C (PKC) activation leads to the downregulation of a number of neurotransmitter transporters and Mn(II) increases PKC activity, we hypothesized that the PKC signaling pathway contributes to the Mn(II)-mediated disruption of Gln turnover. Our results have shown that Mn exposure increases the phosphorylation of both the PKCα and PKCδ isoforms. PKC activity was also shown to be increased in response to Mn(II) treatment. Corroborating our earlier observations, Mn(II) also caused a decrease in Gln uptake. This effect was blocked by PKC inhibitors. Notably, PKC activation caused a decrease in Gln uptake mediated by systems ASC and N, but had no effect on the activities of systems A and L. Exposure to α-phorbol 12-myristate 13-acetate significantly decreased SNAT3 (system N) and ASCT2 (system ASC) protein levels. Additionally, a co-immunoprecipitation study demonstrated the association of SNAT3 and ASCT2 with the PKCδ isoform, and Western blotting revealed the Mn(II)-mediated activation of PKCδ by proteolytic cleavage. PKC activation was also found to increase SNAT3 and ubiquitin ligase Nedd4-2 binding and to induce hyperubiquitination. Taken together, these findings demonstrate that the Mn(II)-induced dysregulation of Gln homeostasis in astrocytes involves PKCδ signaling accompanied by an increase in ubiquitin-mediated proteolysis.


Assuntos
Astrócitos/metabolismo , Glutamina/metabolismo , Intoxicação por Manganês/metabolismo , Proteína Quinase C/fisiologia , Sistema ASC de Transporte de Aminoácidos/biossíntese , Sistema ASC de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Animais , Biotinilação , Western Blotting , Células Cultivadas , Cloretos/toxicidade , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Imunoprecipitação , Compostos de Manganês , Intoxicação por Manganês/enzimologia , Proteínas de Membrana/biossíntese , Antígenos de Histocompatibilidade Menor , Ubiquitina-Proteína Ligases Nedd4 , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...