Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Glia ; 68(12): 2631-2642, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32585762

RESUMO

Cortical spreading depression (CSD) is a pathological neural excitation that underlies migraine pathophysiology. Since glutamate receptor antagonists impair CSD propagation, susceptibility to CSD might be determined by any of the neuronal (excitatory amino acid carrier 1 [EAAC1]) and glial (GLutamate ASpartate Transporter [GLAST] and glial glutamate transporter 1 [GLT-1]) glutamate transporters, which are responsible for clearing extracellular glutamate. To investigate this hypothesis, we performed electrophysiological, hemodynamic, and electrochemical analyses using EAAC1- (EAAC1 KO), GLAST- (GLAST KO), and conditional GLT1-1-knockout mice (GLT-1 cKO) to assess altered susceptibility to CSD. Despite the incomplete deletion of the gene in the cerebral cortex, GLT-1 cKO mice exhibited significant reduction of GLT-1 protein in the brain without apparent alteration of the cytoarchitecture in the cerebral cortex. Physiological analysis revealed that GLT-1 cKO showed enhanced susceptibility to CSD elicited by chemical stimulation with increased CSD frequency and velocity compared to GLT-1 control. In contrast, the germ-line EAAC1 and GLAST KOs showed no such effect. Intriguingly, both field potential and cerebral blood flow showed faster dynamics with narrower CSD than the controls. An enzyme-based biosensor revealed more rapid accumulation of glutamate in the extracellular space in GLT-1 cKO mice during the early phase of CSD than in GLT-1 control, resulting in an increased susceptibility to CSD. These results provided the first evidence for a novel role of GLT-1 in determining susceptibility to CSD.


Assuntos
Depressão , Animais , Córtex Cerebral/metabolismo , Transportador 2 de Aminoácido Excitatório , Ácido Glutâmico , Camundongos
2.
Biochem Biophys Res Commun ; 529(4): 943-949, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32819603

RESUMO

Glaucoma is one of the leading causes of blindness characterized by progressive loss of retinal ganglion cells (RGCs) and their axons. We reported that glutamate/aspartate transporter (GLAST) knockout mice showed progressive RGC loss and optic nerve degeneration that are similar to glaucoma. To explore the possibility that rare variants in the EAAT1 gene (the human homolog of GLAST) cause susceptibility to glaucoma, we performed targeted sequencing of EAAT1 in 440 patients with glaucoma and 450 control subjects. We identified 8 rare variants in 20 out of 440 patients, including 4 synonymous and 4 missense variants located at protein coding regions. One of these rare variants (rs117295512) showed significant association with the risk of glaucoma (OR = 10.44, P = 0.005). Furthermore, the allele frequency for loss-of-function EAAT1 variants, pAla169Gly and pAla329Thr, was 5.5 folds higher in the glaucoma (1.1%) compared with the control cohort (0.2%). These findings suggest that these rare variants may contribute to the pathogenesis of glaucoma and that loss-of-function variants in EAAT1 are present in a small number of patients with glaucoma.


Assuntos
Transportador 1 de Aminoácido Excitatório/genética , Glaucoma de Ângulo Aberto/genética , Glaucoma de Baixa Tensão/genética , Mutação de Sentido Incorreto , Mutação Silenciosa , Alelos , Sequência de Aminoácidos , Animais , Estudos de Casos e Controles , Linhagem Celular , Transportador 1 de Aminoácido Excitatório/deficiência , Expressão Gênica , Frequência do Gene , Glaucoma de Ângulo Aberto/metabolismo , Glaucoma de Ângulo Aberto/patologia , Humanos , Pressão Intraocular , Glaucoma de Baixa Tensão/metabolismo , Glaucoma de Baixa Tensão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nervo Óptico/metabolismo , Nervo Óptico/patologia , Células Ganglionares da Retina/metabolismo , Células Ganglionares da Retina/patologia , Fatores de Risco , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
3.
J Neurosci ; 34(49): 16273-85, 2014 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-25471567

RESUMO

The lateral habenula (LHb) regulates the activity of monoaminergic neurons in the brainstem. This area has recently attracted a surge of interest in psychiatry because studies have reported the pathological activation of the habenula in patients with major depression and in animal models. The LHb plays a significant role in the pathophysiology of depression; however, how habenular neurons are activated to cause various depression symptoms, such as reduced motivation and sleep disturbance, remain unclear. We hypothesized that dysfunctional astrocytes may cause LHb hyperactivity due to the defective uptake activity of extracellular glutamate, which induces depressive-like behaviors. We examined the activity of neurons in habenular pathways and performed behavioral and sleep analyses in mice with pharmacological and genetic inhibition of the activity of the glial glutamate transporter GLT-1 in the LHb. The habenula-specific inhibition of GLT-1 increased the neuronal firing rate and the level of c-Fos expression in the LHb. Mice with reduced GLT-1 activity in the habenula exhibited a depressive-like phenotype in the tail suspension and novelty-suppressed feeding tests. These animals also displayed increased susceptibility to chronic stress, displaying more frequent avoidant behavior without affecting locomotor activity in the open-field test. Intriguingly, the mice showed disinhibition of rapid eye movement sleep, which is a characteristic sleep pattern in patients with depression. These results provide evidence that disrupting glutamate clearance in habenular astrocytes increases neuronal excitability and depressive-like phenotypes in behaviors and sleep.


Assuntos
Astrócitos/patologia , Depressão/patologia , Depressão/fisiopatologia , Habenula/fisiopatologia , Sono REM , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Astrócitos/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Depressão/genética , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Transportador 2 de Aminoácido Excitatório/genética , Habenula/efeitos dos fármacos , Habenula/patologia , Camundongos , Camundongos Transgênicos , Microinjeções , Neurônios/fisiologia , Inibidores da Captação de Neurotransmissores/administração & dosagem , Inibidores da Captação de Neurotransmissores/farmacologia , Pironas/administração & dosagem , Pironas/farmacologia , Sono REM/genética
4.
Exp Cell Res ; 317(2): 221-33, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20965167

RESUMO

A high concentration of bone morphogenetic proteins (BMPs) stimulates myogenic progenitor cells to undergo heterotopic osteogenic differentiation. However, the physiological role of the Smad signaling pathway during terminal muscle differentiation has not been resolved. We report here that Smad1/5/8 was phosphorylated and activated in undifferentiated growing mouse myogenic progenitor Ric10 cells without exposure to any exogenous BMPs. The amount of phosphorylated Smad1/5/8 was severely reduced during precocious myogenic differentiation under the high cell density culture condition even in growth medium supplemented with a high concentration of serum. Inhibition of the Smad signaling pathway by dorsomorphin, an inhibitor of Smad activation, or noggin, a specific antagonist of BMP, induced precocious terminal differentiation of myogenic progenitor cells in a cell density-dependent fashion even in growth medium. In addition, Smad1/5/8 was transiently activated in proliferating myogenic progenitor cells during muscle regeneration in rats. The present results indicate that the Smad signaling pathway is involved in a critical switch between growth and differentiation of myogenic progenitor cells both in vitro and in vivo. Furthermore, precocious cell density-dependent myogenic differentiation suggests that a community effect triggers the terminal muscle differentiation of myogenic cells by quenching the Smad signaling.


Assuntos
Diferenciação Celular/fisiologia , Mioblastos/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Transdução de Sinais/fisiologia , Proteínas Smad/metabolismo , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Morfogenéticas Ósseas/farmacologia , Proteínas Morfogenéticas Ósseas/fisiologia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Camundongos , Camundongos Endogâmicos ICR , Células Musculares/citologia , Células Musculares/metabolismo , Células Musculares/fisiologia , Fosforilação , Ratos , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
5.
Neuropsychopharmacology ; 40(7): 1569-79, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25662838

RESUMO

An increase in the ratio of cellular excitation to inhibition (E/I ratio) has been proposed to underlie the pathogenesis of neuropsychiatric disorders, such as autism spectrum disorders (ASD), obsessive-compulsive disorder (OCD), and Tourette's syndrome (TS). A proper E/I ratio is achieved via factors expressed in neuron and glia. In astrocytes, the glutamate transporter GLT1 is critical for regulating an E/I ratio. However, the role of GLT1 dysfunction in the pathogenesis of neuropsychiatric disorders remains unknown because mice with a complete deficiency of GLT1 exhibited seizures and premature death. Here, we show that astrocyte-specific GLT1 inducible knockout (GLAST(CreERT2/+)/GLT1(flox/flox), iKO) mice exhibit pathological repetitive behaviors including excessive and injurious levels of self-grooming and tic-like head shakes. Electrophysiological studies reveal that excitatory transmission at corticostriatal synapse is normal in a basal state but is increased after repetitive stimulation. Furthermore, treatment with an N-methyl-D-aspartate (NMDA) receptor antagonist memantine ameliorated the pathological repetitive behaviors in iKO mice. These results suggest that astroglial GLT1 has a critical role in controlling the synaptic efficacy at corticostriatal synapses and its dysfunction causes pathological repetitive behaviors.


Assuntos
Córtex Cerebral/patologia , Transtornos Traumáticos Cumulativos/genética , Transtornos Traumáticos Cumulativos/patologia , Transportador 1 de Aminoácido Excitatório/deficiência , Transportador 2 de Aminoácido Excitatório/deficiência , Sinapses/genética , Animais , Animais Recém-Nascidos , Ansiedade/genética , Transtornos Traumáticos Cumulativos/complicações , Transtornos Traumáticos Cumulativos/tratamento farmacológico , Modelos Animais de Doenças , Inibidores Enzimáticos/uso terapêutico , Transportador 1 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório/genética , Potenciais Pós-Sinápticos Excitadores/genética , Feminino , Regulação da Expressão Gênica/genética , Hiperalgesia/genética , Masculino , Camundongos , Camundongos Transgênicos , Degeneração Neural/etiologia , Degeneração Neural/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas/genética , Convulsões/genética
6.
Mol Brain ; 6: 34, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23902942

RESUMO

BACKGROUND: Loss of retinal ganglion cells (RGCs) is a hallmark of various retinal diseases including glaucoma, retinal ischemia, and diabetic retinopathy. N-methyl-D-aspartate (NMDA)-type glutamate receptor (NMDAR)-mediated excitotoxicity is thought to be an important contributor to RGC death in these diseases. Native NMDARs are heterotetramers that consist of GluN1 and GluN2 subunits, and GluN2 subunits (GluN2A-D) are major determinants of the pharmacological and biophysical properties of NMDARs. All NMDAR subunits are expressed in RGCs in the retina. However, the relative contribution of the different GluN2 subunits to RGC death by excitotoxicity remains unclear. RESULTS: GluN2B- and GluN2D-deficiency protected RGCs from NMDA-induced excitotoxic retinal cell death. Pharmacological inhibition of the GluN2B subunit attenuated RGC loss in glutamate aspartate transporter deficient mice. CONCLUSIONS: Our data suggest that GluN2B- and GluN2D-containing NMDARs play a critical role in NMDA-induced excitotoxic retinal cell death and RGC degeneration in glutamate aspartate transporter deficient mice. Inhibition of GluN2B and GluN2D activity is a potential therapeutic strategy for the treatment of several retinal diseases.


Assuntos
N-Metilaspartato/toxicidade , Neurotoxinas/toxicidade , Subunidades Proteicas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Retina/metabolismo , Retina/patologia , Animais , Morte Celular/efeitos dos fármacos , Transportador 1 de Aminoácido Excitatório/deficiência , Transportador 1 de Aminoácido Excitatório/metabolismo , Deleção de Genes , Marcação In Situ das Extremidades Cortadas , Isoquinolinas/farmacologia , Camundongos , Subunidades Proteicas/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Retina/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA