Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Biomolecules ; 11(2)2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33669305

RESUMO

This study demonstrates how exposure to psychosocial crowding stress (CS) for 3, 7, and 14 days affects glutamate synapse functioning and signal transduction in the frontal cortex (FC) of rats. CS effects on synaptic activity were evaluated in FC slices of the primary motor cortex (M1) by measuring field potential (FP) amplitude, paired-pulse ratio (PPR), and long-term potentiation (LTP). Protein expression of GluA1, GluN2B mGluR1a/5, VGLUT1, and VGLUT2 was assessed in FC by western blot. The body's response to CS was evaluated by measuring body weight and the plasma level of plasma corticosterone (CORT), adrenocorticotropic hormone (ACTH), and interleukin 1 beta (IL1B). CS 3 14d increased FP and attenuated LTP in M1, while PPR was augmented in CS 14d. The expression of GluA1, GluN2B, and mGluR1a/5 was up-regulated in CS 3d and downregulated in CS 14d. VGLUTs expression tended to increase in CS 7d. The failure to blunt the effects of chronic CS on FP and LTP in M1 suggests the impairment of habituation mechanisms by psychosocial stressors. PPR augmented by chronic CS with increased VGLUTs level in the CS 7d indicates that prolonged CS exposure changed presynaptic signaling within the FC. The CS bidirectional profile of changes in glutamate receptors' expression seems to be a common mechanism evoked by stress in the FC.


Assuntos
Lobo Frontal/metabolismo , Receptores de Glutamato/biossíntese , Hormônio Adrenocorticotrópico/biossíntese , Animais , Peso Corporal , Corticosterona/biossíntese , Aglomeração , Eletrofisiologia , Ácido Glutâmico , Interleucina-1beta/biossíntese , Potenciação de Longa Duração , Masculino , Modelos Animais , Córtex Motor , Tamanho do Órgão , Ratos , Ratos Wistar , Receptores de AMPA/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Baço/patologia , Estresse Psicológico , Transmissão Sináptica/efeitos dos fármacos , Proteína Vesicular 1 de Transporte de Glutamato/biossíntese , Proteína Vesicular 2 de Transporte de Glutamato/biossíntese
2.
Mol Cell ; 81(7): 1425-1438.e10, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33662272

RESUMO

Eukaryotic elongation factor 2 (eEF2) mediates translocation of peptidyl-tRNA from the ribosomal A site to the P site to promote translational elongation. Its phosphorylation on Thr56 by its single known kinase eEF2K inactivates it and inhibits translational elongation. Extensive studies have revealed that different signal cascades modulate eEF2K activity, but whether additional factors regulate phosphorylation of eEF2 remains unclear. Here, we find that the X chromosome-linked intellectual disability protein polyglutamine-binding protein 1 (PQBP1) specifically binds to non-phosphorylated eEF2 and suppresses eEF2K-mediated phosphorylation at Thr56. Loss of PQBP1 significantly reduces general protein synthesis by suppressing translational elongation. Moreover, we show that PQBP1 regulates hippocampal metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD) and mGluR-LTD-associated behaviors by suppressing eEF2K-mediated phosphorylation. Our results identify PQBP1 as a novel regulator in translational elongation and mGluR-LTD, and this newly revealed regulator in the eEF2K/eEF2 pathway is also an excellent therapeutic target for various disease conditions, such as neural diseases, virus infection, and cancer.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Hipocampo/metabolismo , Depressão Sináptica de Longo Prazo , Elongação Traducional da Cadeia Peptídica , Fator 2 de Elongação de Peptídeos/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Animais , Proteínas de Ligação a DNA/genética , Células HEK293 , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Fator 2 de Elongação de Peptídeos/genética , Fosforilação , Receptores de Glutamato Metabotrópico/genética
3.
J Neurosci ; 41(11): 2344-2359, 2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33500274

RESUMO

Metabotropic glutamate receptor 7 (mGlu7) is an inhibitory heterotrimeric G-protein-coupled receptor that modulates neurotransmitter release and synaptic plasticity at presynaptic terminals in the mammalian central nervous system. Recent studies have shown that rare mutations in glutamate receptors and synaptic scaffold proteins are associated with neurodevelopmental disorders (NDDs). However, the role of presynaptic mGlu7 in the pathogenesis of NDDs remains largely unknown. Recent whole-exome sequencing (WES) studies in families with NDDs have revealed that several missense mutations (c.1865G>A:p.R622Q; c.461T>C:p.I154T; c.1972C>T:p.R658W and c.2024C>A:p.T675K) or a nonsense mutation (c.1757G>A:p.W586X) in the GRM7 gene may be linked to NDDs. In the present study, we investigated the mechanistic links between GRM7 point mutations and NDD pathology. We find that the pathogenic GRM7 I154T and R658W/T675K mutations lead to the degradation of the mGlu7 protein. In particular, the GRM7 R658W/T675K mutation results in a lack of surface mGlu7 expression in heterologous cells and cultured neurons isolated from male and female rat embryos. We demonstrate that the expression of mGlu7 variants or exposure to mGlu7 antagonists impairs axon outgrowth through the mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling pathway during early neuronal development, which subsequently leads to a decrease in the number of presynaptic terminals in mature neurons. Treatment with an mGlu7 agonist restores the pathologic phenotypes caused by mGlu7 I154T but not by mGlu7 R658W/T675K because of its lack of neuronal surface expression. These findings provide evidence that stable neuronal surface expression of mGlu7 is essential for neural development and that mGlu7 is a promising therapeutic target for NDDs.SIGNIFICANCE STATEMENT Neurodevelopmental disorders (NDDs) affect brain development and function by multiple etiologies. Metabotropic glutamate receptor 7 (mGlu7) is a receptor that controls excitatory neurotransmission and synaptic plasticity. Since accumulating evidence indicates that the GRM7 gene locus is associated with NDD risk, we analyzed the functional effects of human GRM7 variants identified in patients with NDDs. We demonstrate that stable neuronal surface expression of mGlu7 is essential for axon outgrowth and presynaptic terminal development in neurons. We found that mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling and subsequent cytoskeletal dynamics are defective because of the degradation of mGlu7 variants. Finally, we show that the defects caused by mGlu7 I154T can be reversed by agonists, providing the rationale for proposing mGlu7 as a potential therapeutic target for NDDs.


Assuntos
Axônios/patologia , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Mutação Puntual/genética , Terminações Pré-Sinápticas , Receptores de Glutamato Metabotrópico/genética , Animais , Axônios/efeitos dos fármacos , Contagem de Células , Sobrevivência Celular , Proteínas Quinases Dependentes de AMP Cíclico/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/genética , Feminino , Regulação da Expressão Gênica , Masculino , Neurônios/metabolismo , Neurônios/patologia , Gravidez , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Transdução de Sinais/genética , Sinapses/patologia , Sequenciamento do Exoma
4.
Neurotox Res ; 39(3): 634-644, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33464538

RESUMO

Aluminum demonstrates clear neurotoxicity and can cause Alzheimer's disease (AD)-like symptoms, including cognitive impairment. One toxic effect of aluminum is a decrease in synaptic plasticity, but the specific mechanism remains unclear. In this study, PC12 cells were treated with Al(mal)3 to construct a toxic cell model. (S)-3,5-Dihydroxyphenylglycine (DHPG), α-methyl-4-carboxyphenylglycine (MCPG), and mGluR1-siRNA were used to interfere with the expression of metabotropic glutamate receptor subtype 1 (mGluR1). Polymerase chain reaction and western blotting were used to investigate the expression of mGluR1, protein kinase C (PKC), and N-methyl-D-aspartate receptor (NMDAR) subunits. ELISA was used to detect PKC enzyme activity. In PC12 cells, mRNA and protein expressions of PKC and NMDAR subunits were inhibited by Al(mal)3. Aluminum may further regulate the expression of NMDAR1 and NMDAR2B through mGluR1 to regulate PKC enzyme activity, thereby affecting learning and memory functions. Furthermore, the results implied that the mGluR1-PKC-NMDAR signaling pathway may predominately involve positive regulation. These findings provide new targets for studying the neurotoxic mechanism of aluminum.


Assuntos
Alumínio/toxicidade , Proteína Quinase C/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Animais , Expressão Gênica , Glicina/análogos & derivados , Glicina/farmacologia , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Células PC12 , Ratos , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores
5.
Toxicology ; 440: 152500, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32428529

RESUMO

Autism spectrum disorders (ASD) include neurodevelopmental disorders in which behavioral deficits can result from neuronal imbalance of excitation to inhibition (E/I) in the brain. Here we used RT-qPCR to screen for the expression of 99 genes associated with excitatory (glutamatergic) and inhibitory (GABAergic) neurotransmission in the cerebral cortex, hippocampus and cerebellum of rats in an established VPA model of ASD. The largest changes in the expression of glutamatergic genes were found in the cerebral cortex, where 12 genes including these encoding some of the subunits of the ionotropic glutamate receptors, were upregulated, while 2 genes were downregulated. The expression of genes encoding the presynaptic glutamatergic proteins vGluT1 and mGluR7 and PKA, involved in downstream glutamatergic signaling, was elevated more than 100-fold. Changes in GABAergic gene expression were found in the cortex, cerebellum and hippocampus; 3 genes were upregulated, and 3 were downregulated. In conclusion, these results revealed that, in the ASD model, several glutamatergic genes in the rat cerebral cortex were upregulated, which contrasts with small and balanced changes in the expression of GABAergic genes. The VPA rat model, useful in studying the molecular basis of ASD, may be suitable for testing experimental therapies in these disabilities.


Assuntos
Transtorno Autístico/induzido quimicamente , Transtorno Autístico/genética , Ácido Glutâmico/genética , Ácido Valproico , Ácido gama-Aminobutírico/genética , Animais , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Feminino , GABAérgicos , Perfilação da Expressão Gênica , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de Glutamato Metabotrópico/genética , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/biossíntese , Proteína Vesicular 1 de Transporte de Glutamato/genética
6.
Neurochem Int ; 129: 104515, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31369778

RESUMO

Lysophosphatidic acid (LPA) is a glycerophospholipid that can be detected in serum, saliva and cerebrospinal fluid. However, the effect of LPA on neuronal death and survival has not been fully determined. In the present study, we investigated the potential neurotoxic effect of LPA in primary cultured cortical neurons. Treatment with LPA (0.5, 1 and 5 µM) markedly decreased neuronal viability, increased lactate dehydrogenase (LDH) release and promoted apoptosis in cortical neurons. The results of western blot showed that LPA increased the expression of endoplasmic reticulum (ER) stress associated factors, and the protein misfolding inhibitor 4-phenylbutyric acid (4-PBA) attenuated LPA-induced toxicity. In addition, treatment with LPA did not alter the expression and distribution of Homer1 in cortical neurons. The protein levels of metabotropic glutamate receptor 1 (mGluR1), but not metabotropic glutamate receptor 5 (mGluR5), were significantly increased by LPA at 12 and 24 h after treatment. Knockdown of Homer1 using specific siRNA partially prevented the LPA-induced neurotoxicity and ER stress. Furthermore, the results of Ca2+ imaging showed that treatment with LPA induced intracellular Ca2+ release, which could be partially prevented by 4-PBA and downregulation of Homer1. The LPA-induced intracellular Ca2+ release was associated with ER Ca2+ release through the Homer1-mGluR1 pathway. In summary, our results showed that LPA treatment induced ER stress and apoptosis in cortical neurons, and its neurotoxicity was partially mediated by Ca2+ release from the ER via the Homer1/mGluR1 pathway.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Proteínas de Arcabouço Homer/fisiologia , Lisofosfolipídeos/toxicidade , Proteínas do Tecido Nervoso/fisiologia , Neurônios/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/fisiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Regulação para Baixo , Feminino , Proteínas de Arcabouço Homer/antagonistas & inibidores , Proteínas de Arcabouço Homer/biossíntese , Proteínas de Arcabouço Homer/genética , Proteínas do Tecido Nervoso/antagonistas & inibidores , Neurônios/metabolismo , Fenilbutiratos/farmacologia , Cultura Primária de Células , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/biossíntese , Receptor de Glutamato Metabotrópico 5/genética , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de Glutamato Metabotrópico/genética
7.
Neurosci Lett ; 705: 143-150, 2019 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-31029678

RESUMO

Hyperexcitability in the corticostriatal glutamatergic pathway may have a pivotal role in the pathogenesis of Parkinson's disease (PD). Metabotropic glutamate receptors (mGluRs) modulate glutamate transmission by both pre- and postsynaptic mechanisms, making them attractive targets for modifying pathological changes in the corticostriatal pathway. Exercise reportedly alleviates motor dysfunction and induced neuroplasticity in glutamatergic transmission. Here, the mGluR-mediated plasticity mechanism underlying behavioral improvement by exercise intervention was investigated. The experimental models were prepared by 6-hydroxydopamine injection into the right medial forebrain bundle. The models were evaluated with the apomorphine-induced rotation test. Starting 2 weeks postoperatively, exercise intervention was applied to the PD + Ex group for 4 weeks. The exercise-intervention effects on locomotor behavior, glutamate levels, and mGluR (mGluR2/3 and mGluR5) expression in hemiparkinsonian rats were investigated. The results showed that hemiparkinsonian rats have a significant increase in extracellular glutamate levels in the lesioned-lateral striatum. MGluR2/3 protein expression was reduced while mGluR5 protein expression was increased in the striatum. Notably, treadmill exercise markedly reversed these abnormal changes in the corticostriatal glutamate system and promoted motor performance in PD rats. These findings suggest that mGluR-mediated glutamatergic transmission in the corticostriatal pathway may serve as an attractive target for exercise-induced neuroplasticity in hemiparkinsonian rats.


Assuntos
Corpo Estriado/fisiopatologia , Terapia por Exercício , Locomoção/fisiologia , Doença de Parkinson/metabolismo , Receptor de Glutamato Metabotrópico 5/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Animais , Corpo Estriado/metabolismo , Ácido Glutâmico/metabolismo , Masculino , Feixe Prosencefálico Mediano/efeitos dos fármacos , Oxidopamina , Ratos
8.
Neurosci Lett ; 690: 76-82, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30315852

RESUMO

Drugs of abuse modify synaptic long-term potentiation and long-term depression (LTD) in the nucleus accumbens, and the impairment of synaptic plasticity in this brain region may be a universal feature of drug addiction. It is unknown whether metabotropic glutamate receptors (mGluRs) play a role in synaptic plasticity induced by drugs such as morphine. The neurochemical, electrophysiological, and Western blotting experiments reported here reveal a novel form of LTD in synapses of the shell region of the nucleus accumbens induced in vivo by low-frequency stimulation of the medial prefrontal cortex. This plasticity required the activation of N-methyl-d-aspartate receptors and mGluR2/3 but not mGluR5. The expression of mGluR2/3 was downregulated during withdrawal from repeated morphine exposure (10 days after the last injection), resulting in impaired low-frequency stimulation-induced LTD. These results indicate that withdrawal-induced mGluR2/3 downregulation alters neural plasticity after morphine exposure, which may be a mechanism contributing to drug addiction.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Morfina/efeitos adversos , Núcleo Accumbens/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Síndrome de Abstinência a Substâncias/metabolismo , 2-Amino-5-fosfonovalerato/farmacologia , Aminoácidos/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Estimulação Elétrica , Depressão Sináptica de Longo Prazo/fisiologia , Masculino , Microinjeções , Núcleo Accumbens/fisiopatologia , Córtex Pré-Frontal/fisiologia , Ratos , Receptor de Glutamato Metabotrópico 5/fisiologia , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Síndrome de Abstinência a Substâncias/fisiopatologia
9.
Psychopharmacology (Berl) ; 236(1): 265-272, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30215216

RESUMO

RATIONALE: Post-traumatic stress disorder (PTSD) is a devastating anxiety-related disorder which develops subsequent to a severe psychologically traumatic event. Only ~ 9% of people who experience such a trauma develop PTSD. It is clear that a number of factors, including genetics, influence whether an individual will develop PTSD subsequent to a trauma. The 129S1/SvImJ (S1) inbred mouse strain displays poor fear extinction and may be useful to model this specific aspect of PTSD. The metabotropic glutamate receptor 7 (mGlu7 receptor) has previously been shown to be involved in cognitive processes and anxiety-like behaviour placing it in a key position to regulate fear extinction processes. We sought to compare mGlu7 receptor mRNA levels in the S1 strain with those in the robustly extinguishing C57BL/6J (B6) inbred strain using in situ hybridisation (ISH) in three brain regions associated with fear extinction: the amygdala, hippocampus and prefrontal cortex (PFC). RESULTS: Compared to the B6 strain, S1 mice had increased mGlu7 receptor mRNA levels in the lateral amygdala (LA) and basolateral amygdala (BLA) subdivisions. An increase was also seen in the hippocampal CA1 and CA3 subregions of S1 mice. No difference in mGlu7 receptor levels were seen in the central nucleus (CeA) of the amygdala, dentate gyrus (DG) of the hippocampus or prefrontal cortex. CONCLUSIONS: These data show altered mGlu7 receptor expression in key brain regions associated with fear extinction in two different inbred mouse strains which differ markedly in their fear extinction behaviour. Altered mGlu7 receptor levels may contribute to the deficit fear extinction processes seen in fear extinction in the S1 strain.


Assuntos
Modelos Animais de Doenças , Extinção Psicológica/fisiologia , Medo/fisiologia , RNA Mensageiro/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Tonsila do Cerebelo/metabolismo , Animais , Transtornos de Ansiedade/genética , Transtornos de Ansiedade/metabolismo , Medo/psicologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Córtex Pré-Frontal/metabolismo , RNA Mensageiro/genética , Receptores de Glutamato Metabotrópico/genética
10.
Neuropharmacology ; 141: 223-237, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30170084

RESUMO

A specific activation of metabotropic glutamate receptor 7 (mGluR7) has been shown to be neuroprotective in various models of neuronal cell damage, however, its role in glia cell survival has not been studied, yet. Thus, we performed comparative experiments estimating protective effects of the mGluR7 allosteric agonist AMN082 in glia, neuronal and neuronal-glia cell cultures against various harmful stimuli. First, the transcript levels of mGluR7 and other subtypes of group II and III mGluRs in cortical neuronal, neuronal-glia and glia cell cultures have been measured by qPCR method. Next, we demonstrated that AMN082 with similar efficiency attenuated the glia cell damage evoked by staurosporine (St) and doxorubicin (Dox). The AMN082-mediated glioprotection was mGluR7-dependent and associated with decreased DNA fragmentation without involvement of caspase-3 inhibition. Moreover, the inhibitors of PI3K/Akt and MAPK/ERK1/2 pathways blocked the protective effect of AMN082. In neuronal and neuronal-glia cell cultures in the model of glutamate (Glu)- but not St-evoked cell damage, we showed a significant glia contribution to mGluR7-mediated neuroprotection. Finally, by using glia and neuronal cells derived from mGluR7+/+ and mGluR7-/- mice we demonstrated a higher cell-damaging effect of St and Dox in mGluR7-deficient glia but not in neurons (cerebellar granule cells). Our present data showed for the first time a glioprotective potential of AMN082 underlain by mechanisms involving the activation of PI3K/Akt and MAPK/ERK1/2 pathways and pro-survival role of mGluR7 in glia cells. These findings together with the confirmed neuroprotective properties of AMN082 justify further research on mGluR7-targeted therapies for various CNS disorders.


Assuntos
Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Compostos Benzidrílicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/agonistas , Animais , Compostos Benzidrílicos/antagonistas & inibidores , Caspase 3/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/metabolismo , Técnicas de Cocultura , Fragmentação do DNA/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Doxorrubicina/antagonistas & inibidores , Inibidores Enzimáticos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de Glutamato Metabotrópico/genética , Transdução de Sinais , Estaurosporina/antagonistas & inibidores
11.
Neurosci Lett ; 681: 56-61, 2018 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-29800675

RESUMO

The nucleus accumbens (NAc) plays a primary role in opioid reward. The actions of glutamate are mediated by the activation of ionotropic and metabotropic glutamate receptors (mGluRs). Previous documents have shown the extensive distributions of the different types of mGluRs, including mGluR7, in regions that are involved in opioid reward, such as the NAc. In this study, seventy male Wistar rats were used to investigate the role of mGluR7 receptors in the NAc on the acquisition and expression of morphine-induced conditioned place preference (CPP). In Experiment 1, to determine the effect of AMN082, a selective mGluR7 allosteric agonist, on the acquisition of morphine-induced conditioned place preference (CPP), the rats bilaterally received AMN082 (1, 3 and 5 µg/0.5 µL DMSO) during three-day conditioning by morphine (5 mg/kg). In Experiment 2, the rats bilaterally received AMN082 (5 µg/0.5 µL DMSO) 5 min prior to the post-conditioning test to investigate the effect of AMN082 on the expression of morphine-induced CPP. The results showed that the intra-accumbal injection of AMN082 prevents the acquisition of morphine-induced CPP in a dose-dependent manner. However, intra-accumbal injection of AMN082 had no effect on the expression of morphine-induced CPP. The findings propose that the mGluR7 in the NAc inhibits the acquisition of morphine-induced CPP that could be mediated by inhibition of NMDA receptors in the NAc.


Assuntos
Analgésicos Opioides/administração & dosagem , Compostos Benzidrílicos/administração & dosagem , Condicionamento Psicológico/fisiologia , Morfina/administração & dosagem , Núcleo Accumbens/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Regulação Alostérica/efeitos dos fármacos , Regulação Alostérica/fisiologia , Animais , Condicionamento Psicológico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Injeções Intravenosas , Masculino , Núcleo Accumbens/efeitos dos fármacos , Ratos , Ratos Wistar , Receptores de Glutamato Metabotrópico/agonistas
12.
J Psychiatry Neurosci ; 43(2): 102-110, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29481317

RESUMO

BACKGROUND: The nucleus accumbens (NAcc) has been implicated in the pathology and treatment of schizophrenia. Recent postmortem evidence suggests a hyperglutamatergic state in the NAcc. With the present study we aimed to explore possible glutamatergic alterations in the NAcc of a large schizophrenia cohort. METHODS: We performed immunoblots on postmortem NAcc samples from 30 individuals who had schizophrenia and 30 matched controls. We examined the protein expression of primary glutamatergic receptors, including the N-methyl-D-aspartate (NMDA) receptor (NR1, NR2A and NR2B subunits) and the group 1 metabotropic glutamate receptor (mGluR1 and mGluR5; dimeric and monomeric forms). In addition, we measured the group 1 mGluR endogenous regulators, neurochondrin and Homer1b/c, which have recently been implicated in the pathophysiology of schizophrenia. RESULTS: Protein levels of glutamatergic receptors and endogenous regulators were not significantly different between the controls and individuals who had schizophrenia. Furthermore, mGluR5, but not mGluR1, showed a positive association with NMDA receptor subunits, suggesting differential interactions between these receptors in this brain region. LIMITATIONS: Investigation of these proteins in antipsychotic-naive individuals, in addition to the subregions of the NAcc and subcellular fractions, will strengthen future studies. CONCLUSION: The present study does not provide evidence for glutamatergic abnormalities within the NAcc of individuals with schizophrenia. Taken together with the results of previous studies, these findings suggest NMDA receptors and group 1 mGluRs are altered in a brain region-dependent manner in individuals with schizophrenia. The differential associations between mGluR1, mGluR5 and NMDA receptors observed in this study warrant further research into the interactions of these proteins and the implications for the therapeutic and adverse effect profile of glutamatergic-based novel therapeutics.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Receptores de N-Metil-D-Aspartato/biossíntese , Esquizofrenia/metabolismo , Estudos de Casos e Controles , Feminino , Proteínas de Arcabouço Homer/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade
13.
Pigment Cell Melanoma Res ; 31(4): 534-540, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29453787

RESUMO

Studies demonstrate that GRM, expressed by >60% of human melanomas, may be a therapeutic target. We performed a phase II trial of 100 mg PO bid of riluzole, an inhibitor of GRM1 signaling, in patients with advanced melanoma with the primary endpoint of response rate. Thirteen patients with GRM1-positive tumors were enrolled. No objective responses were observed, and accrual was stopped. Stable disease was noted in six (46%) patients, with one patient on study for 42 weeks. Riluzole was well tolerated, with fatigue (62%) as the most common adverse event. Downregulation of MAPK and PI3K/AKT was noted in 33% of paired tumor biopsies. Hypothesis-generating correlative studies suggested that downregulation of angiogenic markers and increased leukocytes at the active edge of tumor correlate with clinical benefit. Pharmacokinetic analysis showed interpatient variability consistent with prior riluzole studies. Future investigations should interrogate mechanisms of biologic activity and advance the development of agents with improved bioavailability.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Riluzol/administração & dosagem , Adulto , Feminino , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Melanoma/patologia , Pessoa de Meia-Idade , Receptores de Glutamato Metabotrópico/biossíntese , Riluzol/efeitos adversos
14.
Mol Neurobiol ; 55(3): 1998-2012, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28265857

RESUMO

The serotonin 2A (5-HT2A) and metabotropic glutamate 2 (mGlu2) receptors regulate each other and are associated with schizophrenia. The Roman high- (RHA-I) and the Roman low- (RLA-I) avoidance rat strains present well-differentiated behavioral profiles, with the RHA-I strain emerging as a putative genetic rat model of schizophrenia-related features. The RHA-I strain shows increased 5-HT2A and decreased mGlu2 receptor binding levels in prefrontal cortex (PFC). Here, we looked for differences in gene expression and transcriptional regulation of these receptors. The striatum (STR) was included in the analysis. 5-HT2A, 5-HT1A, and mGlu2 mRNA and [3H]ketanserin binding levels were measured in brain homogenates. As expected, 5-HT2A binding was significantly increased in PFC in the RHA-I rats, while no difference in binding was observed in STR. Surprisingly, 5-HT2A gene expression was unchanged in PFC but significantly decreased in STR. mGlu2 receptor gene expression was significantly decreased in both PFC and STR. No differences were observed for the 5-HT1A receptor. Chromatin immunoprecipitation assay revealed increased trimethylation of histone 3 at lysine 27 (H3K27me3) at the promoter region of the HTR2A gene in the STR. We further looked at the Akt/GSK3 signaling pathway, a downstream point of convergence of the serotonin and glutamate system, and found increased phosphorylation levels of GSK3ß at tyrosine 216 and increased ß-catenin levels in the PFC of the RHA-I rats. These results reveal region-specific regulation of the 5-HT2A receptor in the RHA-I rats probably due to absence of mGlu2 receptor that may result in differential regulation of downstream pathways.


Assuntos
Aprendizagem da Esquiva/fisiologia , Epigênese Genética/fisiologia , Regiões Promotoras Genéticas/fisiologia , Receptor 5-HT2A de Serotonina/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Animais , Expressão Gênica , Masculino , Ratos , Ratos Transgênicos , Receptor 5-HT2A de Serotonina/genética , Receptores de Glutamato Metabotrópico/genética , Especificidade da Espécie
15.
Rom J Morphol Embryol ; 58(1): 67-72, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28523300

RESUMO

Several studies have indicated that abnormal prenatal changes in the circulating glucocorticoids (GCs), induced by either maternal stress or exogenous GC administration, significantly alter the development of Purkinje cells (PCs). Among the suggested mechanisms that could mediate this GC-dependent PC susceptibility are changes in the expression of type-1 metabotropic glutamate receptors (mGluR1). In the current study, we analyzed whether a single course of prenatally administered betamethasone phosphate (BET) in pregnant rats increased the immunohistochemical expression of mGluR1 in PCs and decreased PC dendritic growth. The data obtained showed that in utero BET exposure resulted in a significant immunohistochemical overexpression of mGluR1 and a significant reduction in Purkinje cell dendritic outgrowth during postnatal life.


Assuntos
Córtex Cerebelar/efeitos dos fármacos , Dendritos/efeitos dos fármacos , Glucocorticoides/farmacologia , Células de Purkinje/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/biossíntese , Animais , Córtex Cerebelar/citologia , Córtex Cerebelar/metabolismo , Dendritos/metabolismo , Feminino , Imuno-Histoquímica , Masculino , Células de Purkinje/citologia , Células de Purkinje/metabolismo , Ratos , Ratos Sprague-Dawley
16.
Neurochem Res ; 42(9): 2468-2478, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28365868

RESUMO

Studies by Bruce Ransom and colleagues have made a major contribution to show that white matter is susceptible to ischemia/hypoxia. White matter contains axons and the glia that support them, notably myelinating oligodendrocytes, which are highly vulnerable to ischemic-hypoxic damage. Previous studies have shown that metabotropic GluRs (mGluRs) are cytoprotective for oligodendrocyte precursor cells and immature oligodendrocytes, but their potential role in adult white matter was unresolved. Here, we report that group 1 mGluR1/5 and group 2 mGluR3 subunits are expressed in optic nerves from mice aged postnatal day (P)8-12 and P30-35. We demonstrate that activation of group 1 mGluR protects oligodendrocytes against oxygen-glucose deprivation (OGD) in developing and young adult optic nerves. In contrast, group 2 mGluR are shown to be protective for oligodendrocytes against OGD in postnatal but not young adult optic nerves. The cytoprotective effect of group 1 mGluR requires activation of PKC, whilst group 2 mGluR are dependent on negatively regulating adenylyl cyclase and cAMP. Our results identify a role for mGluR in limiting injury of oligodendrocytes in developing and young adult white matter, which may be useful for protecting oligodendrocytes in neuropathologies involving excitoxicity and ischemia/hypoxia.


Assuntos
Isquemia/metabolismo , Isquemia/prevenção & controle , Oligodendroglia/metabolismo , Nervo Óptico/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Animais , Animais Recém-Nascidos , AMP Cíclico/metabolismo , Glucose/farmacologia , Isquemia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/patologia , Nervo Óptico/efeitos dos fármacos , Nervo Óptico/patologia , Técnicas de Cultura de Órgãos
17.
Gene Expr Patterns ; 22(2): 46-53, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27818290

RESUMO

The glutamatergic system directs central nervous system (CNS) neuronal activity and may underlie various neuropsychiatric disorders. Glutamate transmits its effects through multiple receptor classes. Class II metabotropic glutamate receptors, mGlu2 and mGlu3, play an important role in regulating synaptic release of different neurotransmitter systems and consequently modulate signaling across several neuronal subtypes. Drugs targeting mGlu2 and mGlu3 are seen as potential therapeutics for various psychiatric and neurological disorders, and defining their expression through development can aid in understanding their distinct function. Here, non-radioactive in situ hybridization was used to detect mGlu2 and mGlu3 mRNA in the CNS of 129SvEv mice at PN1, PN8, PN25, PN40, and PN100. At PN1, mGlu2 and mGlu3 are strongly expressed cortically, most notably in layer III and V. Subcortically, mGlu2 is detected in thalamic nuclei; mGlu3 is highly expressed in the striatum. By PN8, the most notable changes are in hippocampus and cortex, with mGlu2 densely expressed in the dentate gyrus, and showing increased cortical levels especially in medial cortex. At PN8, mGlu3 is observed in cortex and striatum, with highest levels detected in reticular thalamic nucleus. At PN25 patterns of expression approximated those observed across adulthood (PN40 & PN100): mGlu2 expression was high in cortex and dentate gyrus while mGlu3 showed expression in the reticular thalamic nucleus, cortex, and striatum. These studies provide a foundation for future research seeking to parse out the roles of mGlu2 from mGlu3, paving the way for better understanding of how these receptors regulate activity in the brain.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Hipocampo/metabolismo , Receptores de Glutamato Metabotrópico/genética , Animais , Sistema Nervoso Central/metabolismo , Córtex Cerebelar/metabolismo , Giro Denteado/metabolismo , Camundongos , Camundongos Knockout , Receptores de Glutamato Metabotrópico/biossíntese
18.
Sci Rep ; 6: 34391, 2016 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-27721389

RESUMO

Group 1 metabotropic glutamate receptors (mGluR1/mGluR5) play an integral role in neurodevelopment and are implicated in psychiatric disorders, such as schizophrenia. mGluR1 and mGluR5 are expressed as homodimers, which is important for their functionality and pharmacology. We examined the protein expression of dimeric and monomeric mGluR1α and mGluR5 in the prefrontal cortex (PFC) and hippocampus throughout development (juvenile/adolescence/adulthood) and in the perinatal phencyclidine (PCP) model of schizophrenia. Under control conditions, mGluR1α dimer expression increased between juvenile and adolescence (209-328%), while monomeric levels remained consistent. Dimeric mGluR5 was steadily expressed across all time points; monomeric mGluR5 was present in juveniles, dramatically declining at adolescence and adulthood (-97-99%). The mGluR regulators, Homer 1b/c and Norbin, significantly increased with age in the PFC and hippocampus. Perinatal PCP treatment significantly increased juvenile dimeric mGluR5 levels in the PFC and hippocampus (37-50%) but decreased hippocampal mGluR1α (-50-56%). Perinatal PCP treatment also reduced mGluR1α dimer levels in the PFC at adulthood (-31%). These results suggest that Group 1 mGluRs have distinct dimeric and monomeric neurodevelopmental patterns, which may impact their pharmacological profiles at specific ages. Perinatal PCP treatment disrupted the early expression of Group 1 mGluRs which may underlie neurodevelopmental alterations observed in this model.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hipocampo/embriologia , Neurogênese , Fenciclidina/efeitos adversos , Córtex Pré-Frontal/embriologia , Multimerização Proteica , Receptor de Glutamato Metabotrópico 5/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Esquizofrenia , Animais , Feminino , Masculino , Fenciclidina/farmacologia , Ratos , Ratos Sprague-Dawley , Esquizofrenia/induzido quimicamente , Esquizofrenia/metabolismo
19.
J Comp Neurol ; 524(12): 2363-78, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27121676

RESUMO

Metabotropic glutamate receptors (mGluRs) are mainly known for regulating excitability of neurons. However, mGluR6 at the photoreceptor-ON bipolar cell synapse mediates sign inversion through glutamatergic inhibition. Although this is currently the only confirmed function of mGluR6, other functions have been suggested. Here we present Tg(mglur6b:EGFP)zh1, a new transgenic zebrafish line recapitulating endogenous expression of one of the two mglur6 paralogs in zebrafish. Investigating transgene as well as endogenous mglur6b expression within the zebrafish retina indicates that EGFP and mglur6b mRNA are not only expressed in bipolar cells, but also in a subset of ganglion and amacrine cells. The amacrine cells labeled in Tg(mglur6b:EGFP)zh1 constitute a novel cholinergic, non-GABAergic, non-starburst amacrine cell type described for the first time in teleost fishes. Apart from the retina, we found transgene expression in subsets of periventricular neurons of the hypothalamus, Purkinje cells of the cerebellum, various cell types of the optic tectum, and mitral/ruffed cells of the olfactory bulb. These findings suggest novel functions of mGluR6 besides sign inversion at ON bipolar cell dendrites, opening up the possibility that inhibitory glutamatergic signaling may be more prevalent than currently thought. J. Comp. Neurol. 524:2363-2378, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Fluorescência Verde/biossíntese , Receptores de Glutamato Metabotrópico/biossíntese , Retina/metabolismo , Transdução de Sinais/fisiologia , Animais , Animais Geneticamente Modificados , Encéfalo/embriologia , Proteínas de Fluorescência Verde/genética , Receptores de Glutamato Metabotrópico/genética , Retina/embriologia , Peixe-Zebra
20.
J Nucl Med ; 57(2): 242-7, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26514176

RESUMO

UNLABELLED: A recent study from our laboratory found that (18)F-FIMX is an excellent PET radioligand for quantifying metabotropic glutamate receptor 1 (mGluR1) in monkey brain. This study evaluated the ability of (18)F-FIMX to quantify mGluR1 in humans. A second goal was to use the relative density of mGluR1 gene transcripts in brain regions to estimate specific uptake and nondisplaceable uptake (VND) in each brain region. METHODS: After injection of 189 ± 3 MBq of (18)F-FIMX, 12 healthy volunteers underwent a dynamic PET scan over 120 min. For 6 volunteers, images were acquired until 210 min. A metabolite-corrected arterial input function was measured from the radial artery. Four other subjects underwent whole-body scanning to estimate radiation exposure. RESULTS: (18)F-FIMX uptake into the human brain was high (SUV = 4-6 in the cerebellum), peaked at about 10 min, and washed out rapidly. An unconstrained 2-tissue-compartment model fitted the data well, and distribution volume (VT) (mL⋅cm(-3)) values ranged from 1.5 in the caudate to 11 in the cerebellum. A 120-min scan provided stable VT values in all regions except the cerebellum, for which an acquisition time of at least 170 min was necessary. VT values in brain regions correlated well with mGluR1 transcript density, and the correlation suggested that VND of (18)F-FIMX was quite low (0.5 mL⋅cm(-3)). This measure of VND in humans was similar to that from a receptor blocking study in monkeys, after correcting for differences in plasma protein binding. Similar to other (18)F-labeled ligands, the effective dose was about 23 µSv/MBq. CONCLUSION: (18)F-FIMX can quantify mGluR1 in the human brain with a 120- to 170-min scan. Correlation of brain uptake with the relative density of mGluR1 transcript allows specific receptor binding of a radioligand to be quantified without injecting pharmacologic doses of a blocking agent.


Assuntos
Benzamidas , Química Encefálica/genética , Compostos Radiofarmacêuticos , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Tiazóis , Adulto , Benzamidas/farmacocinética , Encéfalo/diagnóstico por imagem , Feminino , Regulação da Expressão Gênica/genética , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Tomografia por Emissão de Pósitrons/métodos , Ligação Proteica , Artéria Radial/diagnóstico por imagem , Radiometria , Compostos Radiofarmacêuticos/farmacocinética , Receptores de Glutamato Metabotrópico/biossíntese , Tiazóis/farmacocinética , Contagem Corporal Total , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...