Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Mol Psychiatry ; 29(4): 1205-1215, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38418578

RESUMEN

The ionotropic glutamate delta receptor GluD1, encoded by the GRID1 gene, is involved in synapse formation, function, and plasticity. GluD1 does not bind glutamate, but instead cerebellin and D-serine, which allow the formation of trans-synaptic bridges, and trigger transmembrane signaling. Despite wide expression in the nervous system, pathogenic GRID1 variants have not been characterized in humans so far. We report homozygous missense GRID1 variants in five individuals from two unrelated consanguineous families presenting with intellectual disability and spastic paraplegia, without (p.Thr752Met) or with (p.Arg161His) diagnosis of glaucoma, a threefold phenotypic association whose genetic bases had not been elucidated previously. Molecular modeling and electrophysiological recordings indicated that Arg161His and Thr752Met mutations alter the hinge between GluD1 cerebellin and D-serine binding domains and the function of this latter domain, respectively. Expression, trafficking, physical interaction with metabotropic glutamate receptor mGlu1, and cerebellin binding of GluD1 mutants were not conspicuously altered. Conversely, upon expression in neurons of dissociated or organotypic slice cultures, we found that both GluD1 mutants hampered metabotropic glutamate receptor mGlu1/5 signaling via Ca2+ and the ERK pathway and impaired dendrite morphology and excitatory synapse density. These results show that the clinical phenotypes are distinct entities segregating in the families as an autosomal recessive trait, and caused by pathophysiological effects of GluD1 mutants involving metabotropic glutamate receptor signaling and neuronal connectivity. Our findings unravel the importance of GluD1 receptor signaling in sensory, cognitive and motor functions of the human nervous system.


Asunto(s)
Discapacidad Intelectual , Receptores de Glutamato Metabotrópico , Transducción de Señal , Sinapsis , Humanos , Discapacidad Intelectual/genética , Masculino , Sinapsis/metabolismo , Sinapsis/genética , Femenino , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Transducción de Señal/genética , Homocigoto , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Linaje , Adulto , Paraplejía/genética , Paraplejía/metabolismo , Animales , Niño , Neuronas/metabolismo , Adolescente , Células HEK293 , Mutación/genética
2.
Cells ; 12(15)2023 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-37566031

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by progressive degeneration of motor neurons (MNs). Astrocytes display a toxic phenotype in ALS, which results in MN damage. Glutamate (Glu)-mediated excitotoxicity and group I metabotropic glutamate receptors (mGluRs) play a pathological role in the disease progression. We previously demonstrated that in vivo genetic ablation or pharmacological modulation of mGluR5 reduced astrocyte activation and MN death, prolonged survival and ameliorated the clinical progression in the SOD1G93A mouse model of ALS. This study aimed to investigate in vitro the effects of mGluR5 downregulation on the reactive spinal cord astrocytes cultured from adult late symptomatic SOD1G93A mice. We observed that mGluR5 downregulation in SOD1G93A astrocytes diminished the cytosolic Ca2+ overload under resting conditions and after mGluR5 simulation and reduced the expression of the reactive glial markers GFAP, S100ß and vimentin. In vitro exposure to an anti-mGluR5 antisense oligonucleotide or to the negative allosteric modulator CTEP also ameliorated the altered reactive astrocyte phenotype. Downregulating mGluR5 in SOD1G93A mice reduced the synthesis and release of the pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α and ameliorated the cellular bioenergetic profile by improving the diminished oxygen consumption and ATP synthesis and by lowering the excessive lactate dehydrogenase activity. Most relevantly, mGluR5 downregulation hampered the neurotoxicity of SOD1G93A astrocytes co-cultured with spinal cord MNs. We conclude that selective reduction in mGluR5 expression in SOD1G93A astrocytes positively modulates the astrocyte reactive phenotype and neurotoxicity towards MNs, further supporting mGluR5 as a promising therapeutic target in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Enfermedades Neurodegenerativas , Receptor del Glutamato Metabotropico 5 , Animales , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Astrocitos/metabolismo , Regulación hacia Abajo/genética , Ácido Glutámico/metabolismo , Ratones Transgénicos , Enfermedades Neurodegenerativas/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Receptor del Glutamato Metabotropico 5/genética
3.
Arch Biochem Biophys ; 744: 109698, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37487948

RESUMEN

Numerous epidemiological studies suggest a link between Parkinson's disease (PD) and cancer, indicating that PD-associated proteins may mediate the development of cancer. Here, we investigated a potential role of PD-associated protein α-synuclein in regulating liver cancer progression in vivo and in vitro. We found the negative correlation of α-synuclein with metabotropic glutamate receptor 5 (mGluR5) and γ-synuclein by analyzing the data from The Cancer Genome Atlas database, liver cancer patients and hepatoma cells with overexpressed α-synuclein. Moreover, upregulated α-synuclein suppressed the growth, migration, and invasion. α-synuclein was found to associate with mGluR5 and γ-synuclein, and the truncated N-terminal of α-synuclein was essential for the interaction. Furthermore, overexpressed α-synuclein exerted the inhibitory effect on hepatoma cells through the degradation of mGluR5 and γ-synuclein via α-synuclein-dependent autophagy-lysosomal pathway (ALP). Consistently, in vivo experiments with rotenone-induced rat model of PD also confirmed that, upregulated α-synuclein in liver cancer tissues through targeting on mGluR5/α-synuclein/γ-synuclein complex inhibited tumorigenesis involving in ALP-dependent degradation of mGluR5 and γ-synuclein. These findings give an insight into an important role of PD-associated protein α-synuclein accompanied by the complex of mGluR5/α-synuclein/γ-synuclein in distant communications between PD and liver cancer, and provide a new strategy in therapeutics for the treatment of liver cancer.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad de Parkinson , Animales , Ratas , alfa-Sinucleína/metabolismo , Autofagia/fisiología , Carcinogénesis , Transformación Celular Neoplásica , gamma-Sinucleína/genética , gamma-Sinucleína/metabolismo , Enfermedad de Parkinson/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Regulación hacia Arriba , Humanos
4.
Psychopharmacology (Berl) ; 240(4): 837-851, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36725696

RESUMEN

RATIONALE AND OBJECTIVE: Post-traumatic stress disorder (PTSD) is a prevalent and debilitating psychiatric disorder. However, its specific etiological mechanism remains unclear. Previous studies have shown that traumatic stress changes metabotropic glutamate receptor 5 (mGluR5) expression in the hippocampus (HIP) and prefrontal cortex (PFC). More importantly, mGluR5 expression is often accompanied by alterations in brain-derived neurotrophic factor (BDNF). Furthermore, BDNF/tropomyosin-associated kinase B (TrkB) signaling plays multiple roles, including roles in neuroplasticity and antidepressant activity, by regulating glutamate transporter-1 (GLT-1) expression. This study aims to explore the effects of inhibiting mGluR5 on PTSD-like behaviors and BDNF, TrkB, and GLT-1 expression in the HIP and PFC of inevitable foot shock (IFS)-treated rats. METHODS: Seven-day IFS was used to establish a PTSD rat model, and 2-methyl-6-(phenylethynyl)-pyridine (MPEP) (10 mg/kg, intraperitoneal injection) was used to inhibit the activity of mGluR5 during IFS in rats. After modeling, behavioral changes and mGluR5, BDNF, TrkB, and GLT-1 expression in the PFC and HIP were examined. RESULTS: First, the IFS procedure induced PTSD-like behavior. Second, IFS increased the expression of mGluR5 and decreased BDNF, TrkB, and GLT-1 expression in the PFC and HIP. Third, the mGluR5 antagonist blocked the above behavioral and molecular alterations. CONCLUSIONS: mGluR5 was involved in IFS-induced PTSD-like behavior by changing BDNF, TrkB, and GLT-1 expression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Transportador 2 de Aminoácidos Excitadores , Receptor del Glutamato Metabotropico 5 , Receptor trkB , Trastornos por Estrés Postraumático , Animales , Ratas , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/metabolismo , Corteza Prefrontal/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Receptor trkB/genética , Receptor trkB/metabolismo , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/metabolismo , Transportador 2 de Aminoácidos Excitadores/genética , Transportador 2 de Aminoácidos Excitadores/metabolismo
5.
PLoS One ; 17(11): e0275937, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36383609

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in communication, and social skills, as well as repetitive and/or restrictive interests and behaviors. The severity of ASD varies from mild to severe, drastically interfering with the quality of life of affected individuals. The current occurrence of ASD in the United States is about 1 in 44 children. The precise pathophysiology of ASD is still unknown, but it is believed that ASD is heterogeneous and can arise due to genetic etiology. Although various genes have been implicated in predisposition to ASD, metabotropic glutamate receptor 5 (mGluR5) is one of the most common downstream targets, which may be involved in autism. mGluR5 signaling has been shown to play a crucial role in neurodevelopment and neural transmission making it a very attractive target for understanding the pathogenesis of ASD. In the present study, we determined the effect of genetic ablation of mGluR5 (Grm5) on an ASD-like phenotype using a rat model to better understand the role of mGluR5 signaling in behavior patterns and clinical manifestations of ASD. We observed that mGluR5 Ko rats exhibited exaggerated self-grooming and increased marble burying, as well as deficits in social novelty. Our results suggest that mGluR5 Ko rats demonstrate an ASD-like phenotype, specifically impaired social interaction as well as repetitive and anxiety-like behavior, which are correlates of behavior symptoms observed in individuals with ASD. The mGluR5 Ko rat model characterized in this study may be explored to understand the molecular mechanisms underlying ASD and for developing effective therapeutic modalities.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Ratas , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Modelos Animales de Enfermedad , Fenotipo , Calidad de Vida , Receptor del Glutamato Metabotropico 5/genética
6.
Sci Adv ; 8(33): eabn7357, 2022 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-35984881

RESUMEN

Schizophrenia is a polygenetic disease, the heterogeneity of which is likely complicated by epigenetic modifications yet to be elucidated. Here, we performed transcriptomic analysis of peripheral blood RNA from monozygotic twins discordant for schizophrenia and identified a schizophrenia-associated down-regulated microRNA, miR-501-3p. We showed that the loss of miR-501-3p in germline knockout (KO) male mice resulted in dendritic structure defects, glutamatergic transmission enhancement, and sociability, memory, and sensorimotor gating disruptions, which were attenuated when miR-501 expression was conditionally restored in the nervous system. Combining the results of proteomic analyses with the known genes linked to schizophrenia revealed that metabotropic glutamate receptor 5 (mGluR5) was one of the miR-501-3p targets and was elevated in vivo upon loss of miR-501. Treatment with the mGluR5 negative allosteric modulator 3-2((-methyl-4-thiazolyl) ethynyl) pyridine or the N-methyl-d-aspartate receptor antagonist 2-amino-5-phosphonopentanoic acid ameliorated the deficits observed in Mir501-KO mice. The epigenetic and pathophysiological mechanism that links miR-501-3p to the modulation of glutamatergic transmission provides etiological implications for schizophrenia.


Asunto(s)
MicroARNs , Receptor del Glutamato Metabotropico 5 , Esquizofrenia , Animales , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Proteómica , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Esquizofrenia/genética
7.
Zhonghua Yi Xue Za Zhi ; 102(27): 2108-2114, 2022 Jul 19.
Artículo en Chino | MEDLINE | ID: mdl-35844113

RESUMEN

Objectives: To investigate the association of single nucleotide polymorphisms (SNP) of glutamate receptor metabotropic 5 (GRM5) gene with schizophrenia susceptibility(SZ) in a Chinese Han population. Methods: Twenty-two SNPs located in GRM5 gene in 528 paranoid SZ patients and 528 control subjects recruited from northern Henanwere analyzed. The clinical features of 267 first-episode SZ patients were assessed with the Positive and Negative Syndrome Scale (PANSS). Results: The SZ group included 264 males and 264 females, aged (27±8) years; the healthy control group had 264 males and 264 females, aged (28±8) years.The differences in the genotypic and allelic frequencies of two SNPs (rs567990 and rs12421343) were statistically significant between the SZ patients and control groups (all P<0.05). The allele frequency of rs504183 was also statistically different between the two groups (P=0.030). When the subjects were stratified by sex, the genotypic and allelic frequencies of rs12421343 in female subjects were statistically different between the SZ patients and control groups. The allele frequencies of SNPs (rs12422021, rs567990, and rs7101540) were also statisticallydifferent between the two groups (all P<0.05). Meanwhile, rs567990 AG+GG carriers had a higher risk for SZ than AA carriers in female subjects(OR=1.946, 95%CI: 1.264-2.995). In addition, the patients with different genotypes (GG, AA+AG) of rs12422021 showed statistically significant differences in PANSS total score(84.8±24.4 vs 75.3±18.6), positive (16.2±4.3 vs 14.4±4.2), excitement (12.4±5.1 vs 10.2±4.1) and cognitive impairment factor scores (15.2±6.8 vs 13.3±3.9) (all P<0.05). The patients with AC and the other two genotypes (AA and CC) of rs504183 showed statistically significant differences in PANSS negative factor score(27.4±9.9 vs 24.7±8.4 and 23.4±8.1, both P<0.05). Conclusion: The current study provides further evidence that GRM5 is associated with SZ, and suggests a putative sex difference.


Asunto(s)
Receptor del Glutamato Metabotropico 5 , Esquizofrenia , Estudios de Casos y Controles , China , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Receptor del Glutamato Metabotropico 5/genética , Receptores de Glutamato/genética , Esquizofrenia/genética
8.
Sci Rep ; 12(1): 5621, 2022 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-35379852

RESUMEN

Obesity represents a global health problem and is characterized by metabolic dysfunctions and a low-grade chronic inflammatory state, which can increase the risk of comorbidities, such as atherosclerosis, diabetes and insulin resistance. Here we tested the hypothesis that the genetic deletion of metabotropic glutamate receptor 5 (mGluR5) may rescue metabolic and inflammatory features present in BACHD mice, a mouse model of Huntington's disease (HD) with an obese phenotype. For that, we crossed BACHD and mGluR5 knockout mice (mGluR5-/-) in order to obtain the following groups: Wild type (WT), mGluR5-/-, BACHD and BACHD/mGluR5-/- (double mutant mice). Our results showed that the double mutant mice present decreased body weight as compared to BACHD mice in all tested ages and reduced visceral adiposity as compared to BACHD at 6 months of age. Additionally, 12-month-old double mutant mice present increased adipose tissue levels of adiponectin, decreased leptin levels, and increased IL-10/TNF ratio as compared to BACHD mice. Taken together, our preliminary data propose that the absence of mGluR5 reduce weight gain and visceral adiposity in BACHD mice, along with a decrease in the inflammatory state in the visceral adipose tissue (VAT), which may indicate that mGluR5 may play a role in adiposity modulation.


Asunto(s)
Enfermedad de Huntington , Animales , Enfermedad de Huntington/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Obesidad/complicaciones , Obesidad/genética , Obesidad/metabolismo , Fenotipo , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo
9.
Cancer Gene Ther ; 29(10): 1487-1501, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35396501

RESUMEN

DNA damaging agents are used as chemotherapeutics in many cancers, including hepatocellular carcinoma (HCC). However, they are associated with problems such as low sensitivity to chemotherapy and the induction of liver injury, underscoring the need to identify new therapies. Here, we investigated the differential regulatory effect of metabotropic glutamate receptor 5 (mGlu5) on chemosensitivity in HCC and chemotoxicity to the normal liver. The expression of mGlu5 was higher in HCC than in the normal liver, and correlated with poor prognosis according to The Cancer Genome Atlas database and Integrative Molecular Database of Hepatocellular Carcinoma. Cisplatin, oxaliplatin or methyl methanesulfonate (MMS) caused cell death by decreasing mGlu5 expression in HCC cells and increased mGlu5 expression in hepatic cells. In HCC cells, inhibition of mGlu5 aggravated MMS-induced DNA damage by increasing intracellular Ca2+ overload and mitogen-activated protein kinase (MAPK) activation, thereby promoting cell death, and activation of mGlu5 rescued the effect of MMS. However, in hepatic cells, mGlu5 inhibition alleviated MMS-induced DNA damage by downregulating Ca2+-derived MAPK pathways to advance hepatic cell survival. The opposite effects of mGlu5 overexpression or knockdown on MMS-induced DNA damage supported that cell death is a result of the differential regulation of mGlu5 expression. Inhibition of mGlu5 increased chemosensitivity and decreased chemotoxicity in a rat tumor model. This study suggests that mGlu5 inhibition could act synergistically with HCC chemotherapeutics with minimal side effects, which may improve the treatment of patients with HCC in the future.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Cisplatino , Daño del ADN , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Metilmetanosulfonato , Proteínas Quinasas Activadas por Mitógenos/genética , Oxaliplatino , Ratas , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/metabolismo
10.
J Neurosci ; 42(5): 731-748, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-34907024

RESUMEN

Group I mGluRs have diverse functions in some fundamental neuronal processes, including modulation of synaptic plasticity; and dysregulation of these receptors could lead to various neuropsychiatric disorders. Trafficking of Group I mGluRs plays critical roles in controlling the precise spatiotemporal localization and activity of these receptors, both of which contribute to proper downstream signaling. Using "molecular replacement" approach in hippocampal neurons derived from mice of both sexes, we demonstrate a critical role for the postsynaptic density protein Norbin in regulating the ligand-induced internalization of Group I mGluRs. We show that Norbin associates with protein kinase A (PKA) through its N-terminus and anchors mGluR5 through its C-terminus, both of which are necessary for the ligand-mediated endocytosis of mGluR5, a member of the Group I mGluR family. A point mutation (A687G) at the C-terminus of Norbin inhibits the binding of Norbin to mGluR5 and blocks mGluR5 endocytosis. Finally, we demonstrate an important mechanism by which Norbin regulates mGluR-mediated AMPAR endocytosis in hippocampal neurons, a cellular correlate for mGluR-dependent synaptic plasticity. Norbin, through its PKA-binding regions, recruits PKA to AMPARs on activation of mGluRs; and deletion of the PKA-binding regions of Norbin inhibits mGluR-triggered AMPAR endocytosis. We further report that Norbin is important specifically for the mGluR-mediated AMPAR endocytosis, but not for NMDAR-dependent AMPAR endocytosis. Thus, this study unravels a novel role for Norbin in the internalization of mGluRs and mGluR-mediated AMPAR endocytosis that can have clinical relevance to the function of Group I mGluRs in pathologic processes.SIGNIFICANCE STATEMENT The postsynaptic protein Norbin interacts with mGluR5, and both of them have been implicated in disorders, such as schizophrenia. However, the mechanistic basis underlying the regulation of mGluRs by Norbin remains elusive. We have identified Norbin as an essential mediator of ligand-mediated endocytosis of Group I mGluRs. Mechanistically, Norbin N-terminus associates with protein kinase-A (PKA) and C-terminus binds to mGluR5 to coordinate receptor internalization. A point mutation NorA687G inhibits endocytosis by disrupting this interaction. Additionally, Norbin is critical for the recruitment of PKA to AMPARs on activation of Group I mGluRs that assists in mGluR-mediated AMPAR endocytosis. Thus, Norbin has a dual function in the hippocampus: regulation of mGluR internalization and PKA-dependent modulation of mGluR-mediated AMPAR endocytosis, a prerequisite for mGluR-mediated synaptic plasticity.


Asunto(s)
Endocitosis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Plasticidad Neuronal/fisiología , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Receptor del Glutamato Metabotropico 5/genética , Receptores AMPA/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsis/genética
11.
Sci Rep ; 11(1): 15897, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34354107

RESUMEN

Fragile X Syndrome (FXS) is a neurodevelopmental disorder caused by silencing of the Fragile X Mental Retardation (FMR1) gene. The resulting loss of Fragile X Mental Retardation Protein (FMRP) leads to excessive glutamate signaling via metabotropic glutamate subtype 5 receptors (mGluR5) which has been implicated in the pathogenesis of the disorder. In the present study we used the radioligand 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB) in simultaneous PET-MR imaging of males with FXS and age- and gender-matched controls to assess the availability of mGlu5 receptors in relevant brain areas. Patients with FXS showed lower [18F]FPEB binding potential (p < 0.01), reflecting reduced mGluR5 availability, than the healthy controls throughout the brain, with significant group differences in insula, anterior cingulate, parahippocampal, inferior temporal and olfactory cortices, regions associated with deficits in inhibition, memory, and visuospatial processes characteristic of the disorder. The results are among the first to provide in vivo evidence of decreased availability of mGluR5 in the brain in individuals with FXS than in healthy controls. The consistent results across the subjects, despite the tremendous challenges with neuroimaging this population, highlight the robustness of the protocol and support for its use in drug occupancy studies; extending our radiotracer development and application efforts from mice to humans.


Asunto(s)
Síndrome del Cromosoma X Frágil/diagnóstico por imagen , Síndrome del Cromosoma X Frágil/genética , Receptor del Glutamato Metabotropico 5/genética , Adulto , Biomarcadores/metabolismo , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Radioisótopos de Flúor , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Humanos , Masculino , Persona de Mediana Edad , Tomografía de Emisión de Positrones/métodos , Receptor del Glutamato Metabotropico 5/metabolismo
12.
Pharmacol Rep ; 73(4): 1096-1108, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34426901

RESUMEN

The role of adenosine A2A receptor (A2AR) and striatal-enriched protein tyrosine phosphatase (STEP) interactions in the striatal-pallidal GABA neurons was recently discussed in relation to A2AR overexpression and cocaine-induced increases of brain adenosine levels. As to phosphorylation, combined activation of A2AR and metabotropic glutamate receptor 5 (mGluR5) in the striatal-pallidal GABA neurons appears necessary for phosphorylation of the GluA1 unit of the AMPA receptor to take place. Robert Yasuda (J Neurochem 152: 270-272, 2020) focused on finding a general mechanism by which STEP activation is enhanced by increased A2AR transmission in striatal-pallidal GABA neurons expressing A2AR and dopamine D2 receptor. In his Editorial, he summarized in a clear way the significant effects of A2AR activation on STEP in the dorsal striatal-pallidal GABA neurons which involves a rise of intracellular levels of calcium causing STEP activation through its dephosphorylation. However, the presence of the A2AR in an A2AR-fibroblast growth factor receptor 1 (FGFR1) heteroreceptor complex can be required in the dorsal striatal-pallidal GABA neurons for the STEP activation. Furthermore, Won et al. (Proc Natl Acad Sci USA 116: 8028-8037, 2019) found in mass spectrometry experiments that the STEP splice variant STEP61 can bind to mGluR5 and inactivate it. In addition, A2AR overexpression can lead to increased formation of A2AR-mGluR5 heterocomplexes in ventral striatal-pallidal GABA neurons. It involves enhanced facilitatory allosteric interactions leading to increased Gq-mediated mGluR5 signaling activating STEP. The involvement of both A2AR and STEP in the actions of cocaine on synaptic downregulation was also demonstrated. The enhancement of mGluR5 protomer activity by the A2AR protomer in A2AR-mGluR5 heterocomplexes in the nucleus accumbens shell appears to have a novel significant role in STEP mechanisms by both enhancing the activation of STEP and being a target for STEP61.


Asunto(s)
Neuronas GABAérgicas/fisiología , Fosforilación/genética , Fosforilación/fisiología , Células del Asta Posterior/fisiología , Receptor de Adenosina A2A/metabolismo , Animales , Cocaína/farmacología , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/patología , Neuronas GABAérgicas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Células del Asta Posterior/efectos de los fármacos , Subunidades de Proteína/efectos de los fármacos , Proteínas Tirosina Fosfatasas/genética , Receptor de Adenosina A2A/genética , Receptor del Glutamato Metabotropico 5/genética
13.
Cells ; 10(6)2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34199502

RESUMEN

The metabotropic glutamate receptor 5 (mGluR5) is an essential modulator of synaptic plasticity, learning and memory; whereas in pathological conditions, it is an acknowledged therapeutic target that has been implicated in multiple brain disorders. Despite robust pre-clinical data, mGluR5 antagonists failed in several clinical trials, highlighting the need for a better understanding of the mechanisms underlying mGluR5 function. In this study, we dissected the molecular synaptic modulation mediated by mGluR5 using genetic and pharmacological mouse models to chronically and acutely reduce mGluR5 activity. We found that next to dysregulation of synaptic proteins, the major regulation in protein expression in both models concerned specific processes in mitochondria, such as oxidative phosphorylation. Second, we observed morphological alterations in shape and area of specifically postsynaptic mitochondria in mGluR5 KO synapses using electron microscopy. Third, computational and biochemical assays suggested an increase of mitochondrial function in neurons, with increased level of NADP/H and oxidative damage in mGluR5 KO. Altogether, our observations provide diverse lines of evidence of the modulation of synaptic mitochondrial function by mGluR5. This connection suggests a role for mGluR5 as a mediator between synaptic activity and mitochondrial function, a finding which might be relevant for the improvement of the clinical potential of mGluR5.


Asunto(s)
Mitocondrias/metabolismo , NADP/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Mitocondrias/genética , NADP/genética , Oxidación-Reducción , Receptor del Glutamato Metabotropico 5/genética , Sinapsis/genética
14.
Int J Mol Sci ; 22(11)2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-34070808

RESUMEN

Metabotropic glutamate receptor subtype 5 (mGlu5) is implicated in the pathophysiology of Alzheimer´s disease (AD). However, its alteration at the subcellular level in neurons is still unexplored. Here, we provide a quantitative description on the expression and localisation patterns of mGlu5 in the APP/PS1 model of AD at 12 months of age, combining immunoblots, histoblots and high-resolution immunoelectron microscopic approaches. Immunoblots revealed that the total amount of mGlu5 protein in the hippocampus, in addition to downstream molecules, i.e., Gq/11 and PLCß1, was similar in both APP/PS1 mice and age-matched wild type mice. Histoblots revealed that mGlu5 expression in the brain and its laminar expression in the hippocampus was also unaltered. However, the ultrastructural techniques of SDS-FRL and pre-embedding immunogold demonstrated that the subcellular localisation of mGlu5 was significantly reduced along the neuronal surface of hippocampal principal cells, including CA1 pyramidal cells and DG granule cells, in APP/PS1 mice at 12 months of age. The decrease in the surface localisation of mGlu5 was accompanied by an increase in its frequency at intracellular sites in the two neuronal populations. Together, these data demonstrate, for the first time, a loss of mGlu5 at the plasma membrane and accumulation at intracellular sites in different principal cells of the hippocampus in APP/PS1 mice, suggesting an alteration of the excitability and synaptic transmission that could contribute to the cognitive dysfunctions in this AD animal model. Further studies are required to elucidate the specificity of mGlu5-associated molecules and downstream signalling pathways in the progression of the pathology.


Asunto(s)
Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Membrana Celular/metabolismo , Hipocampo/metabolismo , Células Piramidales/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Membrana Celular/patología , Modelos Animales de Enfermedad , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Regulación de la Expresión Génica , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Presenilina-1/genética , Presenilina-1/metabolismo , Células Piramidales/patología , Receptor del Glutamato Metabotropico 5/metabolismo , Transmisión Sináptica
15.
Behav Brain Res ; 411: 113378, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34029630

RESUMEN

Autism is a neurodevelopmental disorder characterized by impaired social interactions and restricted and repetitive behaviors. Although group 1 metabotropic glutamate receptors (mGluRs), and in particular mGluR5, have been extensively proposed as potential targets for intervention in autism and other neurodevelopmental disorders, there has not been a comprehensive analysis of the effect of mGluR5 loss on behaviors typically assessed in autism mouse models thought to be correlates of behavioral symptoms of human disorders. Here we present a behavioral characterization of mice with complete or partial loss of mGluR5 (homozygous or heterozygous null mutations in Grm5 gene). We tested several autism related behaviors including social interaction, repetitive grooming, digging and locomotor behaviors. We found that digging and marble burying behaviors were almost completely abolished in mGluR5 ko mice, although self-grooming was not altered. Social interaction was impaired in ko but not in heterozygote (het) mice. In tests of locomotor activity and anxiety related behaviors, mGluR5 ko mice exhibited hyperactivity and reduced anxiety in the open field test but unexpectedly, showed hypoactivity in the elevated zero-maze test. There was no impairment in motor learning in the accelerating rotarod in both ko and het mutant. Together these results provide support for the importance of mGluR5 in motor and social behaviors that are specifically affected in autism disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Actividad Motora/genética , Receptor del Glutamato Metabotropico 5/metabolismo , Animales , Ansiedad/genética , Ansiedad/fisiopatología , Trastorno del Espectro Autista/metabolismo , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/fisiología , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Conducta Social , Conducta Estereotipada
16.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799851

RESUMEN

Multiple lines of evidence suggest that dysfunction of the metabotropic glutamate receptor subtype 5 (mGluR5) plays a role in the pathogenesis of autism spectrum disorder (ASD). Yet animal and human investigations of mGluR5 expression provide conflicting findings about the nature of dysregulation of cerebral mGluR5 pathways in subtypes of ASD. The demonstration of reduced mGluR5 expression throughout the living brains of men with fragile X syndrome (FXS), the most common known single-gene cause of ASD, provides a clue to examine mGluR5 expression in ASD. We aimed to (A) compare and contrast mGluR5 expression in idiopathic autism spectrum disorder (IASD), FXS, and typical development (TD) and (B) show the value of positron emission tomography (PET) for the application of precision medicine for the diagnosis and treatment of individuals with IASD, FXS, and related conditions. Two teams of investigators independently administered 3-[18F]fluoro-5-(2-pyridinylethynyl)benzonitrile ([18F]FPEB), a novel, specific mGluR5 PET ligand to quantitatively measure the density and the distribution of mGluR5s in the brain regions, to participants of both sexes with IASD and TD and men with FXS. In contrast to participants with TD, mGluR5 expression was significantly increased in the cortical regions of participants with IASD and significantly reduced in all regions of men with FXS. These results suggest the feasibility of this protocol as a valuable tool to measure mGluR5 expression in clinical trials of individuals with IASD and FXS and related conditions.


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Corteza Cerebral/metabolismo , Síndrome del Cromosoma X Frágil/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Adolescente , Adulto , Animales , Trastorno del Espectro Autista/diagnóstico por imagen , Trastorno del Espectro Autista/genética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Corteza Cerebral/diagnóstico por imagen , Femenino , Síndrome del Cromosoma X Frágil/diagnóstico por imagen , Síndrome del Cromosoma X Frágil/genética , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Tomografía de Emisión de Positrones/métodos , Receptor del Glutamato Metabotropico 5/genética , Adulto Joven
17.
Cell Rep ; 35(2): 108952, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852851

RESUMEN

The mechanisms controlling the post-natal maturation of astrocytes play a crucial role in ensuring correct synaptogenesis. We show that mitochondrial biogenesis in developing astrocytes is necessary for coordinating post-natal astrocyte maturation and synaptogenesis. The astrocytic mitochondrial biogenesis depends on the transient upregulation of metabolic regulator peroxisome proliferator-activated receptor gamma (PPARγ) co-activator 1α (PGC-1α), which is controlled by metabotropic glutamate receptor 5 (mGluR5). At tissue level, the loss or downregulation of astrocytic PGC-1α sustains astrocyte proliferation, dampens astrocyte morphogenesis, and impairs the formation and function of neighboring synapses, whereas its genetic re-expression is sufficient to restore the mitochondria compartment and correct astroglial and synaptic defects. Our findings show that the developmental enhancement of mitochondrial biogenesis in astrocytes is a critical mechanism controlling astrocyte maturation and supporting synaptogenesis, thus suggesting that astrocytic mitochondria may be a therapeutic target in the case of neurodevelopmental and psychiatric disorders characterized by impaired synaptogenesis.


Asunto(s)
Astrocitos/metabolismo , Mitocondrias/genética , Neurogénesis/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Receptor del Glutamato Metabotropico 5/genética , Sinapsis/metabolismo , Transmisión Sináptica/genética , Animales , Animales Recién Nacidos , Astrocitos/citología , Encéfalo/citología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Diferenciación Celular , Proliferación Celular , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Biogénesis de Organelos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Cultivo Primario de Células , Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/genética , Sinapsis/ultraestructura
18.
Neurobiol Dis ; 154: 105338, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33775821

RESUMEN

Fragile X syndrome (FXS) is a common form of intellectual disability and autism caused by the lack of Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein involved in RNA transport and protein synthesis. Upon cellular stress, global protein synthesis is blocked and mRNAs are recruited into stress granules (SGs), together with RNA-binding proteins including FMRP. Activation of group-I metabotropic glutamate (mGlu) receptors stimulates FMRP-mediated mRNA transport and protein synthesis, but their role in SGs formation is unexplored. To this aim, we pre-treated wild type (WT) and Fmr1 knockout (KO) cultured astrocytes with the group-I-mGlu receptor agonist (S)-3,5-Dihydroxyphenylglycine (DHPG) and exposed them to sodium arsenite (NaAsO2), a widely used inducer of SGs formation. In WT cultures the activation of group-I mGlu receptors reduced SGs formation and recruitment of FMRP into SGs, and also attenuated phosphorylation of eIF2α, a key event crucially involved in SGs formation and inhibition of protein synthesis. In contrast, Fmr1 KO astrocytes, which exhibited a lower number of SGs than WT astrocytes, did not respond to agonist stimulation. Interestingly, the mGlu5 receptor negative allosteric modulator (NAM) 2-methyl-6-(phenylethynyl)pyridine (MPEP) antagonized DHPG-mediated SGs reduction in WT and reversed SGs formation in Fmr1 KO cultures. Our findings reveal a novel function of mGlu5 receptor as modulator of SGs formation and open new perspectives for understanding cellular response to stress in FXS pathophysiology.


Asunto(s)
Astrocitos/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Gránulos de Estrés/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/patología , Células Cultivadas , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/antagonistas & inhibidores , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Ratones , Ratones Noqueados , Estrés Oxidativo/fisiología , Receptor del Glutamato Metabotropico 5/genética , Gránulos de Estrés/patología
19.
Mol Biol Rep ; 48(1): 595-600, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33394235

RESUMEN

We previously reported that Numb, a protein localized to clathrin-coated vesicles, regulates the membrane expression of metabotropic glutamate receptor 5 (mGluR5) and is critical to social behaviors. However, the distinct actions of Numb isoforms on mGluR5 have not been investigated. Here, we showed that the expression patterns of Numb-p72 and Numb-p65, two important isoforms of Numb, were distinct in HEK293T cells. Numb-p72, but not Numb-p65, bound to mGluR5α, and enhanced mGluR5 membrane expression by inhibiting its internalization. Our results suggest that a complete structure is required for Numb to bind to mGluR5 and to modulate mGluR5 trafficking.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Transporte de Proteínas/genética , Receptor del Glutamato Metabotropico 5/genética , Movimiento Celular/genética , Células HEK293 , Hipocampo/metabolismo , Humanos , Neuronas/metabolismo , Unión Proteica/genética , Isoformas de Proteínas/genética
20.
Neurobiol Aging ; 98: 225-230, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33341653

RESUMEN

Impaired mRNA translation (protein synthesis) is linked to Alzheimer's disease (AD) pathophysiology. Recent studies revealed the role of increased phosphorylation of eukaryotic elongation factor 2 (eEF2) in AD-associated cognitive deficits. Phosphorylation of eEF2 (at the Thr56 site) by its only known kinase eEF2K leads to inhibition of general protein synthesis. AD is considered as a disease of "synaptic failure" characterized by impairments of synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD). Deficiency of metabotropic glutamate receptor 5-dependent LTD (mGluR-LTD) is indicated in cognitive syndromes associated with various neurological disorders, including AD, but the molecular signaling mechanisms underlying the mGluR-LTD dysregulation in AD remain unclear. In this brief communication, we report genetic repression of eEF2K in aged APP/PS1 AD model mice prevented AD-associated hippocampal mGluR-LTD deficits. Using a pharmacological approach, we further observed that impairments of mGluR-LTD in APP/PS1 mice were rescued by treating hippocampal slices with a small molecule eEF2K antagonist NH125. Our findings, taken together, suggest a critical role of abnormal protein synthesis dysregulation at the elongation phase in AD-associated mGluR-LTD failure, thus providing insights into a mechanistic understanding of synaptic impairments in AD and other related dementia syndromes.


Asunto(s)
Enfermedad de Alzheimer/etiología , Potenciación a Largo Plazo/genética , Potenciación a Largo Plazo/fisiología , Factor 2 de Elongación Peptídica/genética , Factor 2 de Elongación Peptídica/metabolismo , Receptor del Glutamato Metabotropico 5/genética , Receptor del Glutamato Metabotropico 5/fisiología , Enfermedad de Alzheimer/genética , Animales , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Imidazoles/farmacología , Ratones Transgénicos , Plasticidad Neuronal/genética , Factor 2 de Elongación Peptídica/antagonistas & inhibidores , Fosforilación , Biosíntesis de Proteínas , Receptor del Glutamato Metabotropico 5/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA