Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Mol Cell ; 81(7): 1425-1438.e10, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33662272

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Hipocampo/metabolismo , Depresión Sináptica a Largo Plazo , Extensión de la Cadena Peptídica de Translación , Factor 2 de Elongación Peptídica/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Animales , Proteínas de Unión al ADN/genética , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Noqueados , Factor 2 de Elongación Peptídica/genética , Fosforilación , Receptores de Glutamato Metabotrópico/genética
2.
J Neurosci ; 41(11): 2344-2359, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33500274

RESUMEN

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.


Asunto(s)
Axones/patología , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Mutación Puntual/genética , Terminales Presinápticos , Receptores de Glutamato Metabotrópico/genética , Animales , Axones/efectos de los fármacos , Recuento de Células , Supervivencia Celular , Proteínas Quinasas Dependientes de AMP Cíclico/biosíntesis , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Femenino , Regulación de la Expresión Génica , Masculino , Neuronas/metabolismo , Neuronas/patología , Embarazo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Transducción de Señal/genética , Sinapsis/patología , Secuenciación del Exoma
3.
J Psychiatry Neurosci ; 43(2): 102-110, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29481317

RESUMEN

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.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Núcleo Accumbens/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de N-Metil-D-Aspartato/biosíntesis , Esquizofrenia/metabolismo , Estudios de Casos y Controles , Femenino , Proteínas de Andamiaje Homer/metabolismo , Humanos , Masculino , Persona de Mediana Edad
4.
Neurochem Res ; 42(9): 2468-2478, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28365868

RESUMEN

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.


Asunto(s)
Isquemia/metabolismo , Isquemia/prevención & control , Oligodendroglía/metabolismo , Nervio Óptico/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Animales , Animales Recién Nacidos , AMP Cíclico/metabolismo , Glucosa/farmacología , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Nervio Óptico/efectos de los fármacos , Nervio Óptico/patología , Técnicas de Cultivo de Órganos
5.
Proc Natl Acad Sci U S A ; 110(42): 16963-8, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082084

RESUMEN

Identification of genes influencing complex traits is hampered by genetic heterogeneity, the modest effect size of many alleles, and the likely involvement of rare and uncommon alleles. Etiologic complexity can be simplified in model organisms. By genomic sequencing, linkage analysis, and functional validation, we identified that genetic variation of Grm2, which encodes metabotropic glutamate receptor 2 (mGluR2), alters alcohol preference in animal models. Selectively bred alcohol-preferring (P) rats are homozygous for a Grm2 stop codon (Grm2 *407) that leads to largely uncompensated loss of mGluR2. mGluR2 receptor expression was absent, synaptic glutamate transmission was impaired, and expression of genes involved in synaptic function was altered. Grm2 *407 was linked to increased alcohol consumption and preference in F2 rats generated by intercrossing inbred P and nonpreferring rats. Pharmacologic blockade of mGluR2 escalated alcohol self-administration in Wistar rats, the parental strain of P and nonpreferring rats. The causal role of mGluR2 in altered alcohol preference was further supported by elevated alcohol consumption in Grm2 (-/-) mice. Together, these data point to mGluR2 as an origin of alcohol preference and a potential therapeutic target.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Codón de Terminación , Receptores de Glutamato Metabotrópico , Transmisión Sináptica/genética , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/patología , Animales , Cruzamientos Genéticos , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ácido Glutámico/metabolismo , Ratones , Ratones Noqueados , Ratas , Ratas Wistar , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de Glutamato Metabotrópico/genética , Sinapsis/genética , Sinapsis/metabolismo , Transmisión Sináptica/efectos de los fármacos
6.
Proc Natl Acad Sci U S A ; 110(12): 4804-9, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23382250

RESUMEN

Epigenetic mechanisms are involved in the pathophysiology of depressive disorders and are unique potential targets for therapeutic intervention. The acetylating agent L-acetylcarnitine (LAC), a well-tolerated drug, behaves as an antidepressant by the epigenetic regulation of type 2 metabotropic glutamate (mGlu2) receptors. It caused a rapid and long-lasting antidepressant effect in Flinders Sensitive Line rats and in mice exposed to chronic unpredictable stress, which, respectively, model genetic and environmentally induced depression. In both models, LAC increased levels of acetylated H3K27 bound to the Grm2 promoter and also increased acetylation of NF-ĸB-p65 subunit, thereby enhancing the transcription of Grm2 gene encoding for the mGlu2 receptor in hippocampus and prefrontal cortex. Importantly, LAC reduced the immobility time in the forced swim test and increased sucrose preference as early as 3 d of treatment, whereas 14 d of treatment were needed for the antidepressant effect of chlorimipramine. Moreover, there was no tolerance to the action of LAC, and the antidepressant effect was still seen 2 wk after drug withdrawal. Conversely, NF-ĸB inhibition prevented the increase in mGlu2 expression induced by LAC, whereas the use of a histone deacetylase inhibitor supported the epigenetic control of mGlu2 expression. Finally, LAC had no effect on mGlu2 knockout mice exposed to chronic unpredictable stress, and a single injection of the mGlu2/3 receptor antagonist LY341495 partially blocked LAC action. The rapid and long-lasting antidepressant action of LAC strongly suggests a unique approach to examine the epigenetic hypothesis of depressive disorders in humans, paving the way for more efficient antidepressants with faster onset of action.


Asunto(s)
Acetilcarnitina/farmacología , Antidepresivos/farmacología , Epigénesis Genética/efectos de los fármacos , Hipocampo/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Corteza Prefrontal/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Acetilación/efectos de los fármacos , Aminoácidos , Animales , Clomipramina/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Hipocampo/patología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/genética , Nootrópicos/farmacología , Corteza Prefrontal/patología , Ratas , Receptores de Glutamato Metabotrópico/genética , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/patología , Factores de Tiempo , Xantenos
7.
Breast Cancer Res Treat ; 151(1): 57-73, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25859923

RESUMEN

Metabotropic glutamate receptor 1 (mGluR1/Grm1) is a member of the G-protein-coupled receptor superfamily, which was once thought to only participate in synaptic transmission and neuronal excitability, but has more recently been implicated in non-neuronal tissue functions. We previously described the oncogenic properties of Grm1 in cultured melanocytes in vitro and in spontaneous melanoma development with 100 % penetrance in vivo. Aberrant mGluR1 expression was detected in 60-80 % of human melanoma cell lines and biopsy samples. As most human cancers are of epithelial origin, we utilized immortalized mouse mammary epithelial cells (iMMECs) as a model system to study the transformative properties of Grm1. We introduced Grm1 into iMMECs and isolated several stable mGluR1-expressing clones. Phenotypic alterations in mammary acinar architecture were assessed using three-dimensional morphogenesis assays. We found that mGluR1-expressing iMMECs exhibited delayed lumen formation in association with decreased central acinar cell death, disrupted cell polarity, and a dramatic increase in the activation of the mitogen-activated protein kinase pathway. Orthotopic implantation of mGluR1-expressing iMMEC clones into mammary fat pads of immunodeficient nude mice resulted in mammary tumor formation in vivo. Persistent mGluR1 expression was required for the maintenance of the tumorigenic phenotypes in vitro and in vivo, as demonstrated by an inducible Grm1-silencing RNA system. Furthermore, mGluR1 was found be expressed in human breast cancer cell lines and breast tumor biopsies. Elevated levels of extracellular glutamate were observed in mGluR1-expressing breast cancer cell lines and concurrent treatment of MCF7 xenografts with glutamate release inhibitor, riluzole, and an AKT inhibitor led to suppression of tumor progression. Our results are likely relevant to human breast cancer, highlighting a putative role of mGluR1 in the pathophysiology of breast cancer and the potential of mGluR1 as a novel therapeutic target.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Receptores de Glutamato Metabotrópico/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Humanos , Células MCF-7 , Glándulas Mamarias Animales/efectos de los fármacos , Glándulas Mamarias Animales/patología , Ratones , Receptores de Glutamato Metabotrópico/biosíntesis , Riluzol/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Neurochem Res ; 40(5): 1074-82, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25894678

RESUMEN

Substantial evidence reveals that prenatal stress is closely linked with abnormal behavior in offspring, but the mechanism remains unclear. In this study, our aim was to observe the alterations of behaviors, metabotropic glutamate receptor-1/5 (mGluR1/5) and brain-derived neurotrophic factor (BDNF) in various brain regions of prenatally stressed offspring rats. The forced swimming test (FST) and the open field test (OFT) were carried on 1-month-old offspring rats. The expression levels of mGluR1, mGluR5, and BDNF mRNA were measured in various brain regions of the offspring rats. Our results showed that the immobile time in the FST was significantly increased in the late prenatal stress (LPS) group compared with that in the control group, especially in the female rats. Similarly, in the OFT, the rats in both the mid prenatal stress (MPS) and LPS groups demonstrated anxiety-like behavior, especially the male rats in the LPS group. The expression of mGluR1 protein was increased in the hippocampus and prefrontal cortex of rats from the LPS group, as well as in the prefrontal cortex of rats from the MPS group. Meanwhile, the expression of mGluR5 protein was facilitated in the hippocampus and prefrontal cortex of rats in the LPS group. The expression of mGluR5 protein was increased in the striatum of the female rats in both MPS and LPS groups, and only in the male rats from the LPS group. In addition, the reduced BDNF mRNA level was detected in the hippocampus and prefrontal cortex in the LPS rats, and in the striatum of the female rats in LPS group. These results indicate that alterations of the mGluR1, mGluR5 and BDNF mRNA may contribute to the depression-like and anxiety-like behaviors of prenatally stressed offspring rats.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptor del Glutamato Metabotropico 5/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Estrés Psicológico/metabolismo , Animales , Ansiedad/metabolismo , Ansiedad/psicología , Encéfalo/metabolismo , Depresión/metabolismo , Depresión/psicología , Femenino , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/psicología , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/psicología
9.
Neurochem Res ; 40(11): 2200-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26318863

RESUMEN

The study assessed involvement of Ca(2+) signaling mediated by the metabotropic glutamate receptors mGluR1/5 in brain tolerance induced by hypoxic preconditioning. Acute slices of rat piriform cortex were tested 1 day after exposure of adult rats to mild hypobaric hypoxia for 2 h at a pressure of 480 hPa once a day for three consecutive days. We detected 44.1 ± 11.6 % suppression of in vitro anoxia-induced increases of intracellular Ca(2+) levels and a fivefold increase in Ca(2+) transients evoked by selective mGluR1/5 agonist, DHPG. Western blot analysis of cortical homogenates demonstrated a 11 ± 4 % decrease in mGluR1 immunoreactivity (IR), and in the nuclei-enriched fraction a 12 ± 3 % increase in IR of phospholipase Cß1 (PLCß1), which is a major mediator of mGluR1/5 signaling. Immunocytochemical analysis of the cortex revealed increase in the mGluR1/5 and PLCß1 IR in perikarya, and a decrease in IR of the neuronal inositol trisphosphate receptors (IP3Rs). We suggest that enhanced expression of mGluR5 and PLCß1 and potentiation of Ca(2+) signaling may represent pro-survival upregulation of Ca(2+)-dependent genomic processes, while decrease in mGluR1 and IP3R IR may be attributed to a feedback mechanism preventing excessive intracellular Ca(2+) release.


Asunto(s)
Presión del Aire , Corteza Cerebral/metabolismo , Hipoxia/metabolismo , Receptor del Glutamato Metabotropico 5/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Transducción de Señal/genética , Animales , Señalización del Calcio/genética , Receptores de Inositol 1,4,5-Trifosfato/biosíntesis , Receptores de Inositol 1,4,5-Trifosfato/genética , Masculino , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Fosfolipasa C beta/biosíntesis , Fosfolipasa C beta/genética , Corteza Piriforme/metabolismo , Ratas , Ratas Wistar , Receptor del Glutamato Metabotropico 5/agonistas , Receptor del Glutamato Metabotropico 5/genética , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/genética , Regulación hacia Arriba
10.
J Neurosci ; 33(48): 18951-65, 2013 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-24285900

RESUMEN

Hyperactivity of the glutamatergic system is involved in the development of central sensitization in the pain neuraxis, associated with allodynia and hyperalgesia observed in patients with chronic pain. Herein we study the ability of type 4 metabotropic glutamate receptors (mGlu4) to regulate spinal glutamate signaling and alleviate chronic pain. We show that mGlu4 are located both on unmyelinated C-fibers and spinal neurons terminals in the inner lamina II of the spinal cord where they inhibit glutamatergic transmission through coupling to Cav2.2 channels. Genetic deletion of mGlu4 in mice alters sensitivity to strong noxious mechanical compression and accelerates the onset of the nociceptive behavior in the inflammatory phase of the formalin test. However, responses to punctate mechanical stimulation and nocifensive responses to thermal noxious stimuli are not modified. Accordingly, pharmacological activation of mGlu4 inhibits mechanical hypersensitivity in animal models of inflammatory or neuropathic pain while leaving acute mechanical perception unchanged in naive animals. Together, these results reveal that mGlu4 is a promising new target for the treatment of chronic pain.


Asunto(s)
Agonistas de Aminoácidos Excitadores/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/agonistas , Animales , Western Blotting , Carragenina , Enfermedad Crónica , Constricción Patológica/patología , Fenómenos Electrofisiológicos/fisiología , Técnica del Anticuerpo Fluorescente , Inmersión/fisiopatología , Interneuronas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/fisiología , Dimensión del Dolor/efectos de los fármacos , Técnicas de Placa-Clamp , Ácidos Fosfínicos/administración & dosificación , Ácidos Fosfínicos/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de Glutamato Metabotrópico/genética , Rizotomía , Células Receptoras Sensoriales/fisiología , Médula Espinal/citología , Médula Espinal/fisiología , Transmisión Sináptica/fisiología
11.
J Neurosci ; 33(2): 495-506a, 2013 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-23303930

RESUMEN

Anomalies in prefrontal cortex (PFC) function are posited to underpin difficulties in learning to suppress drug-seeking behavior during abstinence. Because group 1 metabotropic glutamate receptors (mGluRs) regulate drug-related learning, we assayed the consequences of extended access to intravenous cocaine (6 h/d; 0.25 mg/infusion for 10 d) on the PFC expression of group 1 mGluRs and the relevance of observed changes for cocaine seeking. After protracted withdrawal, cocaine-experienced animals exhibited a time-dependent intensification of cue-induced cocaine-seeking behavior and an impaired extinction of this behavior. These behavioral phenomena were associated with a time-dependent reduction in mGluR1/5 expression within ventromedial PFC (vmPFC) of cocaine-experienced animals exposed to extinction testing but not in untested ones. Interestingly, pharmacological manipulations of vmPFC mGluR1/5 produced no immediate effects on cue-induced cocaine-seeking behavior but produced residual effects on a subsequent test for cocaine seeking. At 3 d withdrawal, cocaine-experienced rats infused intra-vmPFC with mGluR1/5 antagonists, either before or after an initial test for cocaine seeking, persisted in their cocaine seeking akin to cocaine-experienced rats in protracted withdrawal. Conversely, cocaine-experienced rats infused with an mGluR1/5 agonist before the initial test for cocaine-seeking at 30 d withdrawal exhibited a facilitation of extinction learning. These data indicate that cue-elicited deficits in vmPFC group 1 mGluR function mediate resistance to extinction during protracted withdrawal from a history of extensive cocaine self-administration and pose pharmacological stimulation of these receptors as a potential approach to facilitate learned suppression of drug-seeking behavior that may aid drug abstinence.


Asunto(s)
Trastornos Relacionados con Cocaína/fisiopatología , Extinción Psicológica/fisiología , Corteza Prefrontal/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Western Blotting , Trastornos Relacionados con Cocaína/psicología , Condicionamiento Operante , Señales (Psicología) , Antagonistas de Aminoácidos Excitadores/farmacología , Extinción Psicológica/efectos de los fármacos , Inyecciones , Masculino , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Corteza Prefrontal/efectos de los fármacos , Piridinas/administración & dosificación , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Recurrencia , Autoadministración , Síndrome de Abstinencia a Sustancias/psicología
12.
J Recept Signal Transduct Res ; 34(4): 261-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24495291

RESUMEN

CONTEXT/OBJECTIVE: Previous studies have demonstrated that various subtypes of the metabotropic glutamate receptors (mGluRs) are expressed in the dorsal root ganglion (DRG) of the peripheral nervous system (PNS), implicating that glutamate potentially contributes to sensory transmission through these receptors. While mGluR expression has been investigated largely in the DRG, the present study focused on mGluR expression on neurons and satellite glial cells (SGCs) of the trigeminal ganglion (TG). MATERIALS AND METHODS: To address the presence of mGluRs in rat TG neurons and their corresponding SGCs, the trigeminal ganglia from six adult male Wistar rats were isolated and immunohistochemistry and immunocytochemistry were performed. The expression of mGluR1α-, mGluR2/3- and mGluR8 on TG neurons and SGCs was investigated in tissue slices and isolated cells. RESULTS: 35.1 ± 6.0% of the TG neurons were positive for mGluR1α, whereas 39.9 ± 7.7% and 55.5 ± 6.3% were positive for mGluR2/3 and mGluR8, respectively. Immunoreactive neurons expressing mGluRs were mainly medium- to large sized, with a smaller population of small-sized neurons showing immunoreactivity. The SGCs showed immunoreactivity toward mGluR1α and mGluR8, but not mGluR2/3, both in the tissue and in isolated cells. CONCLUSIONS: Findings from the present study showed that trigeminal neurons express mGluR1α, mGluR2/3 and mGluR8, while SGCs only express mGluR1α and mGluR8. This novel evidence may advance investigations on a possible role of mGluRs in relation to trigeminal pain transmission within the craniofacial region.


Asunto(s)
Dolor Facial/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Células Satélites Perineuronales/metabolismo , Animales , Dolor Facial/patología , Regulación de la Expresión Génica/genética , Humanos , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Especificidad de Órganos , Ratas , Receptores de Glutamato Metabotrópico/metabolismo , Células Satélites Perineuronales/patología , Ganglio del Trigémino/metabolismo , Ganglio del Trigémino/patología
13.
J Neurosci ; 32(13): 4632-44, 2012 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-22457509

RESUMEN

Inhibitory synapses display a great diversity through varying combinations of presynaptic GABA and glycine release and postsynaptic expression of GABA and glycine receptor subtypes. We hypothesized that increased flexibility offered by this dual transmitter system might serve to tune the inhibitory phenotype to the properties of afferent excitatory synaptic inputs in individual cells. Vestibulocerebellar unipolar brush cells (UBC) receive a single glutamatergic synapse from a mossy fiber (MF), which makes them an ideal model to study excitatory-inhibitory interactions. We examined the functional phenotypes of mixed inhibitory synapses formed by Golgi interneurons onto UBCs in rat slices. We show that glycinergic IPSCs are present in all cells. An additional GABAergic component of large amplitude is only detected in a subpopulation of UBCs. This GABAergic phenotype is strictly anti-correlated with the expression of type II, but not type I, metabotropic glutamate receptors (mGluRs) at the MF synapse. Immunohistochemical stainings and agonist applications show that global UBC expression of glycine and GABA(A) receptors matches the pharmacological profile of IPSCs. Paired recordings of Golgi cells and UBCs confirm the postsynaptic origin of the inhibitory phenotype, including the slow kinetics of glycinergic components. These results strongly suggest the presence of a functional coregulation of excitatory and inhibitory phenotypes at the single-cell level. We propose that slow glycinergic IPSCs may provide an inhibitory tone, setting the gain of the MF to UBC relay, whereas large and fast GABAergic IPSCs may in addition control spike timing in mGluRII-negative UBCs.


Asunto(s)
Cerebelo/fisiología , Ácido Glutámico/fisiología , Inhibición Neural/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transmisión Sináptica/fisiología , Animales , Cerebelo/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Agonistas del GABA/farmacología , Agonistas del GABA/fisiología , Antagonistas del GABA/farmacología , Glicina/fisiología , Glicinérgicos/farmacología , Técnicas In Vitro , Potenciales Postsinápticos Inhibidores/efectos de los fármacos , Potenciales Postsinápticos Inhibidores/fisiología , Interneuronas/fisiología , Ácido Kaínico/farmacología , Masculino , Fibras Nerviosas/fisiología , Inhibición Neural/efectos de los fármacos , Neuronas/fisiología , Ratas , Ratas Wistar , Receptores de Glicina/antagonistas & inhibidores , Receptores de Glicina/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/fisiología
14.
Biochim Biophys Acta ; 1819(11-12): 1123-31, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22771868

RESUMEN

We demonstrated that ectopic expression of metabotropic glutamate receptor 1 (mGluR1/Grm1) in mouse melanocytes was sufficient to induce melanoma development in vivo with 100% penetrance. We also showed that about 60% of human melanoma biopsies and cell lines, but not benign nevi or normal human melanocytes expressed mGluR1, suggesting that GRM1 may be involved in melanomagenesis. mGluR1 is expressed primarily in neurons. In various non-neuronal cells, mGluR1 expression is regulated via binding of Neuron-Restrictive-Silencer-Factor (NRSF) to a Neuron-Restrictive-Silencer-Element (NRSE). Here, we report on the possibility that aberrant mGluR1 expression in melanoma is due to alterations in NRSF and/or NRSE. We show that in human melanocytes, binding of NRSF to NRSE in the GRM1 promoter region is necessary for the suppression of mGluR1 expression. We also show that inhibiting the expression of the transcription factor Sp1 or interference with its ability to bind DNA can result in increased mGluR1 expression perhaps via its function as a negative regulator. In addition, we also provide evidence that demethylation within the promoter region of GRM1 may also be a mechanism for the derepression of mGluR1 expression in melanocytes that progress to cell transformation and tumor formation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanocitos/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Metilación de ADN/genética , Humanos , Melanocitos/patología , Melanoma/genética , Melanoma/patología , Proteínas de Neoplasias/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Elementos de Respuesta , Elementos Silenciadores Transcripcionales , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo
15.
Neurobiol Dis ; 56: 1-5, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23578490

RESUMEN

Fragile X Syndrome (FXS) is a heritable form of mental retardation caused by a non-coding trinucleotide expansion of the FMR1 gene leading to loss of expression of this RNA binding protein. Mutations in this gene are strongly linked to enhanced Group I metabotropic glutamate receptor (mGluR) signaling. A recent report found that mGluR5-dependent endogenous cannabinoid signaling is enhanced in hippocampal slices from fmr1 knockout mice, suggesting a link between FXS and cannabinoid signaling. Alterations in cannabinoid signaling have an impact on learning and memory and may therefore be linked to some aspects of the FXS phenotype. We have used autaptic hippocampal neurons cultured from fmr1 knockout mice to further explore the interaction between endocannabinoid signaling and FMRP. These neurons express several robust forms of retrograde endocannabinoid signaling including depolarization induced suppression of excitation (DSE) and a metabotropic form (MSE) that results from Group I mGluR activation. We now report that young fmr1 neurons exhibit considerably enhanced DSE, likely via increased production of 2-AG, rather than enhanced mGluR-MSE. We find that depolarizations as brief as 50ms, which do not ordinarily produce DSE, routinely inhibited glutamate release. Furthermore, as neuronal cultures mature, CB1-receptor signaling strongly desensitizes. Our results suggest that loss of FMRP broadly affects the endocannabinoid signaling system, possibly through local 2-AG over production. Furthermore, the net effect of the loss of FMRP may actually be diminished cannabinoid signaling due to receptor desensitization as an adaptation to 2-AG overproduction.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Hipocampo/fisiopatología , Neuronas/fisiología , Receptor Cannabinoide CB1/fisiología , Sinapsis/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Baclofeno/farmacología , Interpretación Estadística de Datos , Fenómenos Electrofisiológicos , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/fisiología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/fisiología , Síndrome del Cromosoma X Frágil/fisiopatología , Agonistas del GABA/farmacología , Metoxihidroxifenilglicol/análogos & derivados , Metoxihidroxifenilglicol/farmacología , Ratones , Ratones Noqueados , Receptor Cannabinoide CB1/genética , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de Glutamato Metabotrópico/genética
16.
Int J Mol Sci ; 14(11): 22857-75, 2013 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-24264036

RESUMEN

It is known that the medial vestibular nucleus (MVN) and the cerebellar flocculus are the key areas, which contribute to the behavioral recovery ("vestibular compensation") after unilateral labyrinthectomy (UL). In these areas, how the genetic activities of the metabotropic glutamate receptors mGluR2 and mGluR7 performance after UL is unknown. With the means of quantitative real-time PCR, Western blotting, and immunohistochemistry, we analyzed the expression of mGluR2 and mGluR7 in the bilateral MVN and the flocculus of rats in different stages after UL (the 1st, 3rd, and 7th day). Our results show that in the MVN, the mRNA, and protein expressions of mGluR7 were ipsilaterally decreased at the 1st day following UL. However, in the MVN, no change was observed in the mRNA and protein expressions of mGluR2. On the other hand, the mRNA and protein expression of mGluR2 were enhanced in the ipsilateral flocculus at the 1st day following UL, while in the flocculus no change was shown in mGluR7 mRNA and protein expressions. Our results suggest that mGluR2 and mGluR7 may contribute to the early rebalancing of spontaneous resting activity in the MVN.


Asunto(s)
Receptores de Glutamato Metabotrópico/biosíntesis , Vestíbulo del Laberinto/cirugía , Animales , Regulación de la Expresión Génica , ARN Mensajero/biosíntesis , Ratas , Núcleos Vestibulares/metabolismo
17.
J Neurosci ; 31(40): 14223-34, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976507

RESUMEN

Despite the pronounced neurological deficits associated with mental retardation and autism, the degree to which neocortical circuit function is altered remains unknown. Here, we study changes in neocortical network function in the form of persistent activity states in the mouse model of fragile X syndrome--the Fmr1 knock-out (KO). Persistent activity states, or UP states, in the neocortex underlie the slow oscillation which occurs predominantly during slow-wave sleep, but may also play a role during awake states. We show that spontaneously occurring UP states in the primary somatosensory cortex are 38-67% longer in Fmr1 KO slices. In vivo, UP states reoccur with a clear rhythmic component consistent with that of the slow oscillation and are similarly longer in the Fmr1 KO. Changes in neocortical excitatory circuitry likely play the major role in this alteration as supported by three findings: (1) longer UP states occur in slices of isolated neocortex, (2) pharmacologically isolated excitatory circuits in Fmr1 KO neocortical slices display prolonged bursting states, and (3) selective deletion of Fmr1 in cortical excitatory neurons is sufficient to cause prolonged UP states whereas deletion in inhibitory neurons has no effect. Excess signaling mediated by the group 1 glutamate metabotropic receptor, mGluR5, contributes to the longer UP states. Genetic reduction or pharmacological blockade of mGluR5 rescues the prolonged UP state phenotype. Our results reveal an alteration in network function in a mouse model of intellectual disability and autism which may impact both slow-wave sleep and information processing during waking states.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Neocórtex/fisiología , Red Nerviosa/fisiología , Receptores de Glutamato Metabotrópico/fisiología , Transducción de Señal/fisiología , Animales , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/biosíntesis , Receptores de Glutamato Metabotrópico/genética
18.
J Neurosci Res ; 90(2): 447-60, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22034224

RESUMEN

Hypoxia/ischemia induces proliferation of neural progenitor cells (NPCs) in rodent and human brain; however, the mechanisms remain unknown. We investigated the effects of metabotropic glutamate receptor 5 (mGluR5) on NPC proliferation under hypoxia, the expression of cyclin D1, and the activation of the mitogen-activated protein kinases (MAPKs) signaling pathway in cell culture. The results showed that hypoxia induced mGluR5 expression on NPCs in vitro. Under hypoxia, the mGluR5 agonists DHPG and CHPG significantly increased NPC proliferation in cell activity, diameter of neurospheres, bromodeoxyuridine (BrdU) incorporation and cell division, and expression of cyclin D1, with decreasing cell death. The mGluR5 siRNA and antagonist MPEP decreased the NPC proliferation and expression of cyclin D1, with increasing cell death. Phosphorylated JNK and ERK increased with the proliferation of NPCs after DHPG and CHPG treatment under hypoxia, while p-p38 level decreased. These results demonstrate that the expression of mGluR5 was upregulated during the proliferation of rat NPCs stimulated by hypoxia in vitro. The activation of the ERK and JNK signaling pathway and the expression of cyclin D1 were increased in this process. These finding suggest the involvement of mGluR5 in rat NPC proliferation and provide a target molecule in neural repair after ischemia/hypoxia injury of CNS.


Asunto(s)
Proliferación Celular , Sistema de Señalización de MAP Quinasas/fisiología , Células-Madre Neurales/metabolismo , Receptores de Glutamato Metabotrópico/biosíntesis , Animales , Hipoxia de la Célula/genética , Células Cultivadas , Activación Enzimática/fisiología , Femenino , Células-Madre Neurales/patología , Embarazo , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/genética , Células Madre/metabolismo
19.
J Pharmacol Exp Ther ; 343(1): 167-77, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22787118

RESUMEN

Positive allosteric modulators (PAMs) of metabotropic glutamate receptor 4 (mGluR4) have been proposed as a novel therapeutic approach for the treatment of Parkinson's disease. However, evaluation of this proposal has been limited by the availability of appropriate pharmacological tools to interrogate the target. In this study, we describe the properties of a novel mGluR4 PAM. 5-Methyl-N-(4-methylpyrimidin-2-yl)-4-(1H-pyrazol-4-yl)thiazol-2-amine (ADX88178) enhances glutamate-mediated activation of human and rat mGluR4 with EC(50) values of 4 and 9 nM, respectively. The compound is highly selective for mGluR4 with minimal activities at other mGluRs. Oral administration of ADX88178 in rats is associated with high bioavailability and results in cerebrospinal fluid exposure of >50-fold the in vitro EC(50) value. ADX88178 reverses haloperidol-induced catalepsy in rats at 3 and 10 mg/kg. It is noteworthy that this compound alone has no impact on forelimb akinesia resulting from a bilateral 6-hydroxydopamine lesion in rats. However, coadministration of a low dose of L-DOPA (6 mg/kg) enabled a robust, dose-dependent reversal of the forelimb akinesia deficit. ADX88178 also increased the effects of quinpirole in lesioned rats and enhanced the effects of L-DOPA in MitoPark mice. It is noteworthy that the enhancement of the actions of L-DOPA was not associated with an exacerbation of L-DOPA-induced dyskinesias in rats. ADX88178 is a novel, potent, and selective mGluR4 PAM that is a valuable tool for exploring the therapeutic potential of mGluR4 modulation. The use of this novel tool molecule supports the proposal that activation of mGluR4 may be therapeutically useful in Parkinson's disease.


Asunto(s)
Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/fisiología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Agonistas de Aminoácidos Excitadores/farmacología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedad de Parkinson/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/biosíntesis
20.
Neuropathol Appl Neurobiol ; 37(5): 484-99, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21114681

RESUMEN

AIM: Neuropathological changes classically associated with sheep scrapie do not always correlate with clinical disease. We aimed to determine if selected neuromodulatory responses were altered during the course of the infection as it has been described in Creutzfeldt-Jakob disease and experimental bovine spongiform encephalopathy. METHODS: Hemi-brains from healthy sheep and natural scrapie cases at two stages of infection were examined for biochemical alterations related to the expression of type I metabotropic glutamatergic receptors (mGluR(1) ) and type I adenosine receptors I (A(1) R), and of selected downstream intermediate signalling targets. Immunohistochemistry for different scrapie-related neuropathological changes was performed in the contralateral hemi-brains. RESULTS: PrP(d) deposition, spongiform change, astrocytosis and parvalbumin expression were significantly altered in brains from clinically affected sheep compared with preclinical cases and negative controls; the latter also showed significantly higher immunoreactivity for synaptophysin than clinical cases. Between clinically affected and healthy sheep, no differences were found in the protein levels of mGluR(1) , while phospholipase Cß1 expression in terminally ill sheep was increased in some brain areas but decreased in others. Adenyl cyclase 1 and A(1) R levels were significantly lower in various brain areas of affected sheep. No abnormal biochemical expression levels of these markers were found in preclinically infected sheep. CONCLUSIONS: These findings point towards an involvement of mGluR(1) and A(1) R downstream pathways in natural scrapie. While classical prion disease lesions and neuromodulatory responses converge in some affected regions, they do not do so in others suggesting that there are independent regulatory factors for distinct degenerative and neuroprotective responses.


Asunto(s)
Receptor de Adenosina A1/biosíntesis , Receptores de Glutamato Metabotrópico/biosíntesis , Scrapie/metabolismo , Scrapie/patología , Animales , Western Blotting , Encéfalo/metabolismo , Encéfalo/patología , Inmunohistoquímica , Ovinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA