Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Behav Brain Res ; 412: 113405, 2021 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-34097900

RESUMO

Traumatic brain injury (TBI) is associated with increased risk for mental health disorders, impacting post-injury quality of life and societal reintegration. TBI is also associated with deficits in psychosocial processing, defined as the cognitive integration of social and emotional behaviors, however little is known about how these deficits manifest and their contributions to post-TBI mental health. In this pre-clinical investigation using rats, a single mild blast TBI (mbTBI) induced impairment of psychosocial processing in the absence of confounding physical polytrauma, post-injury motor deficits, affective abnormalities, or deficits in non-social behavior. Impairment severity correlated with acute upregulations of a known oxidative stress metabolite, 3-hydroxypropylmercapturic acid (3-HPMA), in urine. Resting state fMRI alterations in the acute post-injury period implicated key brain regions known to regulate psychosocial behavior, including orbitofrontal cortex (OFC), which is congruent with our previous report of elevated acrolein, a marker of neurotrauma and 3-HPMA precursor, in this region following mbTBI. OFC of mbTBI-exposed rats demonstrated elevated mRNA expression of metabotropic glutamate receptors 1 and 5 (mGluR1/5) and injection of mGluR1/5-selective agonist in OFC of uninjured rats approximated mbTBI-induced psychosocial processing impairment, demonstrating a novel role for OFC in this psychosocial behavior. Furthermore, OFC may serve as a hotspot for TBI-induced disruption of psychosocial processing and subsequent mental health disorders.


Assuntos
Concussão Encefálica/psicologia , Córtex Pré-Frontal/fisiopatologia , Funcionamento Psicossocial , Acetilcisteína/análogos & derivados , Acetilcisteína/análise , Acetilcisteína/urina , Acroleína/análise , Acroleína/metabolismo , Animais , Traumatismos por Explosões/psicologia , Encéfalo/fisiopatologia , Concussão Encefálica/fisiopatologia , Lesões Encefálicas/psicologia , Modelos Animais de Doenças , Imageamento por Ressonância Magnética , Masculino , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/metabolismo
2.
Theranostics ; 10(24): 11178-11196, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042277

RESUMO

Selective modulation of metabotropic glutamate receptor 2 (mGlu2) represents a novel therapeutic approach for treating brain disorders, including schizophrenia, depression, Parkinson's disease (PD), Alzheimer's disease (AD), drug abuse and addiction. Imaging mGlu2 using positron emission tomography (PET) would allow for in vivo quantification under physiological and pathological conditions and facilitate drug discovery by enabling target engagement studies. In this paper, we aimed to develop a novel specific radioligand derived from negative allosteric modulators (NAMs) for PET imaging of mGlu2. Methods. A focused small molecule library of mGlu2 NAMs with tetrahydro naphthyridine scaffold was synthesized for pharmacology and physicochemical evaluation. GIRK dose-response assays and CNS panel binding selectivity assays were performed to study the affinity and selectivity of mGlu2 NAMs, among which compounds 14a and 14b were selected as PET ligand candidates. Autoradiography in SD rat brain sections was used to confirm the in vitro binding specificity and selectivity of [11C]14a and [11C]14b towards mGlu2. In vivo binding specificity was then studied by PET imaging. Whole body biodistribution study and radiometabolite analysis were conducted to demonstrate the pharmacokinetic properties of [11C]14b as most promising PET mGlu2 PET ligand. Results. mGlu2 NAMs 14a-14g were synthesized in 14%-20% yields in five steps. NAMs 14a and 14b were selected to be the most promising ligands due to their high affinity in GIRK dose-response assays. [11C]14a and [11C]14b displayed similar heterogeneous distribution by autoradiography, consistent with mGlu2 expression in the brain. While PET imaging study showed good brain permeability for both tracers, compound [11C]14b demonstrated superior binding specificity compared to [11C]14a. Further radiometabolite analysis of [11C]14b showed excellent stability in the brain. Conclusions. Compound 14b exhibited high affinity and excellent subtype selectivity, which was then evaluated by in vitro autoradiography and in vivo PET imaging study after labeling with carbon-11. Ligand [11C]14b, which we named [11C]MG2-1904, demonstrated high brain uptake and excellent in vitro/in vivo specific binding towards mGlu2 with high metabolic stability in the brain. As proof-of-concept, our preliminary work demonstrated a successful example of visualizing mGlu2in vivo derived from NAMs, which represents a promising chemotype for further development and optimization aimed for clinical translation.


Assuntos
Naftiridinas/administração & dosagem , Tomografia por Emissão de Pósitrons/métodos , Ensaio Radioligante/métodos , Compostos Radiofarmacêuticos/administração & dosagem , Receptores de Glutamato Metabotrópico/análise , Regulação Alostérica , Animais , Autorradiografia , Encéfalo/diagnóstico por imagem , Radioisótopos de Carbono , Ligantes , Masculino , Modelos Animais , Naftiridinas/química , Naftiridinas/farmacocinética , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Receptores de Glutamato Metabotrópico/metabolismo , Distribuição Tecidual
3.
J Med Chem ; 63(6): 3381-3389, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-32081008

RESUMO

We have synthesized and characterized [18F]-N-(4-chloro-3-((fluoromethyl-d2)thio)phenyl)-picolinamide ([18F]15) as a potential ligand for the positron emission tomography (PET) imaging of mGluR4 in the brain. Radioligand [18F]15 displays central nervous system drug-like properties, including mGluR4 affinity, potent mGluR4 PAM activity, and selectivity against other mGluRs, as well as sufficient metabolic stability. Radiosynthesis was carried out in two steps. The radiochemical yield of [18F]15 was 11.6 ± 2.9% (n = 7, decay corrected) with a purity of 99% and a molar activity of 84.1 ± 11.8 GBq/µmol. Ex vivo biodistribution studies showed reversible binding of [18F]15 in all investigated tissues including the brain, liver, heart, lungs, and kidneys. PET imaging studies in male Sprague Dawley rats showed that [18F]15 accumulates in the brain regions known to express mGluR4. Pretreatment with the unlabeled mGluR4 PAM compounds 13 (methylthio analogue) and 15 showed significant dose-dependent blocking effects. These results suggest that [18F]15 is a promising radioligand for PET imaging mGluR4 in the brain.


Assuntos
Picolinas/farmacologia , Compostos Radiofarmacêuticos/farmacologia , Receptores de Glutamato Metabotrópico/análise , Animais , Encéfalo/metabolismo , Estabilidade de Medicamentos , Radioisótopos de Flúor/química , Ligantes , Masculino , Microssomos Hepáticos/metabolismo , Picolinas/síntese química , Picolinas/farmacocinética , Tomografia por Emissão de Pósitrons , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/farmacocinética , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo
4.
Brain Struct Funct ; 224(8): 2787-2804, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31422483

RESUMO

Preclinical evidence indicates that mGluR5 is a potential therapeutic target for Parkinson's disease and L-DOPA-induced dyskinesia. However, the mechanisms through which these therapeutic benefits are mediated remain poorly understood. Although the regulatory role of mGluR5 on glutamatergic transmission has been examined in various basal ganglia nuclei, very little is known about the localization and function of mGluR5 in the ventral motor and intralaminar thalamic nuclei, the main targets of basal ganglia output in mammals. Thus, we used immuno-electron microscopy to map the cellular and subcellular localization of group I mGluRs (mGluR1a and mGluR5) in the ventral motor and caudal intralaminar thalamic nuclei in rhesus monkeys. Furthermore, using double immuno-electron microscopy, we examined the subsynaptic localization of mGluR5 in relation to cortical and sub-cortical glutamatergic afferents. Four major conclusions can be drawn from these data. First, mGluR1a and mGluR5 are expressed postsynaptically on the plasma membrane of dendrites of projection neurons and GABAergic interneurons in the basal ganglia- and cerebellar-receiving regions of the ventral motor thalamus and in CM. Second, the plasma membrane-bound mGluR5 immunoreactivity is preferentially expressed perisynaptically at the edges of cortical and sub-cortical glutamatergic afferents. Third, the mGluR5 immunoreactivity is more strongly expressed in the lateral than the medial tiers of CM, suggesting a preferential association with thalamocortical over thalamostriatal neurons in the primate CM. Overall, mGluR5 is located to subserve powerful modulatory role of cortical and subcortical glutamatergic transmission in the primate ventral motor thalamus and CM.


Assuntos
Córtex Cerebral/ultraestrutura , Neurônios/ultraestrutura , Terminações Pré-Sinápticas/ultraestrutura , Receptor de Glutamato Metabotrópico 5/análise , Receptores de Glutamato Metabotrópico/análise , Tálamo/ultraestrutura , Vias Aferentes/ultraestrutura , Animais , Dendritos/ultraestrutura , Feminino , Núcleos Intralaminares do Tálamo/ultraestrutura , Macaca mulatta , Masculino
5.
Bioorg Med Chem ; 27(3): 483-491, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30611634

RESUMO

Metabotropic glutamate receptor 2 (mGluR2) has been suggested as a therapeutic target for treating schizophrenia-like symptoms arising from increased glutamate transmission in the human forebrain. However, no reliable positron emission tomography (PET) radiotracer allowing for in vivo visualization of mGluR2 in the human brain is currently available. In this study, we synthesized 4-(2-fluoro-4-[11C]methoxyphenyl)-5-((2-methylpyridin-4-yl)methoxy)picolinamide ([11C]1) and evaluated its potential as a PET tracer for imaging mGluR2 in the rodent brain. Compound 1, a negative allosteric modulator (NAM) of mGluR2, showed high in vitro binding affinity (IC50: 26 nM) for mGluR2 overexpressed in human cells. [11C]1 was synthesized by O-[11C]methylation of the phenol precursor 2 with [11C]methyl iodide. After the reaction, HPLC purification and formulation, [11C]1 of 7.4 ±â€¯2.8 GBq (n = 8) was obtained from [11C]carbon dioxide of 22.5 ±â€¯4.8 GBq (n = 8) with >99% radiochemical purity and 70 ±â€¯32 GBq/µmol (n = 8) molar activity at the end of synthesis. In vitro autoradiography for rat brains showed that [11C]1 binding was heterogeneously distributed in the cerebral cortex, striatum, hippocampus, and cerebellum. This pattern is consistent with the regional distribution pattern of mGluR2 in the rodent brain. The radioactivity was significantly reduced by self- or MNI-137 (a mGluR2 NAM) blocking. Small-animal PET studies indicated a low in vivo specific binding of [11C]1 in the rat brain. The brain uptake was increased in a P-glycoprotein and breast cancer resistant protein double knockout mouse, when compared to a wild-type mouse. While [11C]1 presented limited potential as an in vivo PET tracer for mGluR2, we suggested that it can be used as a lead compound for developing new radiotracers with improved in vivo brain properties.


Assuntos
Encéfalo/diagnóstico por imagem , Ácidos Picolínicos/química , Tomografia por Emissão de Pósitrons , Receptores de Glutamato Metabotrópico/análise , Animais , Encéfalo/metabolismo , Radioisótopos de Carbono , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Estrutura Molecular , Ácidos Picolínicos/síntese química , Ácidos Picolínicos/farmacocinética , Traçadores Radioativos , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade , Distribuição Tecidual
6.
Sci Signal ; 11(548)2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30228224

RESUMO

G protein-coupled receptors (GPCRs) are major drug targets. Developing a method to measure the activities of GPCRs is essential for pharmacology and drug screening. However, it is difficult to measure the effects of a drug by monitoring the receptor on the cell surface; thus, changes in the concentrations of downstream signaling molecules, which depend on the signaling pathway selectivity of the receptor, are often used as an index of receptor activity. We show that single-molecule imaging analysis provides an alternative method for assessing the effects of ligands on GPCRs. Using total internal reflection fluorescence microscopy (TIRFM), we monitored the dynamics of the diffusion of metabotropic glutamate receptor 3 (mGluR3), a class C GPCR, under various ligand conditions. Our single-molecule tracking analysis demonstrated that increases and decreases in the average diffusion coefficient of mGluR3 quantitatively reflected the ligand-dependent inactivation and activation of receptors, respectively. Through experiments with inhibitors and dual-color single-molecule imaging analysis, we found that the diffusion of receptor molecules was altered by common physiological events associated with GPCRs, including G protein binding, and receptor accumulation in clathrin-coated pits. We also confirmed that agonist also decreased the average diffusion coefficient for class A and B GPCRs, demonstrating that this parameter is a good index for estimating ligand effects on many GPCRs regardless of their phylogenetic groups, the chemical properties of the ligands, or G protein-coupling selectivity.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Microscopia de Fluorescência/métodos , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Aminoácidos/metabolismo , Células HEK293 , Humanos , Ligantes , Toxina Pertussis/metabolismo , Toxina Pertussis/farmacologia , Ligação Proteica/efeitos dos fármacos , Ensaio Radioligante/métodos , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/genética , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo , Xantenos/metabolismo
7.
Sci Rep ; 7(1): 14945, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29097780

RESUMO

Metabotropic glutamate receptor subtype 1 (mGluR1) is a crucial pharmacological target for several central nervous system disorders. In this study, we aimed to monitor in vivo regional changes of mGluR1 related to neuroinflammation in the brains of rats after pilocarpine-induced status epilepticus (PISE) using longitudinal positron emission tomography (PET). PISE was induced in rats by administering lithium chloride, followed by repeated pilocarpine hydrochloride treatments. PET assessments were conducted using N-[4-[6-(isopropylamino)-pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-methyl-4-[11C]methylbenzamide ([11C]ITDM), a selective radioligand for mGluR1, and N-benzyl-N-[11C]methyl-2-(7-methyl-8-oxo-2-phenyl-7,8-dihydro-9H-purin-9-yl)acetamide ([11C]DAC), a selective translocator protein PET ligand for neuroinflammation monitoring. PET scans were conducted on PISE rats at 1 day (acute), 1 week (subacute) and 3 weeks (chronic) after repeated seizures. PET with [11C]ITDM showed significant decreases of mGluR1 availability (BPND) in the thalamus and hippocampus after PISE over the chronic period. Conversely, PET with [11C]DAC exhibited a significant increase of radioactive uptake in the forebrain after the acute period, especially in the thalamus. These conflicting changes in the thalamus indicated negative correlation. In conclusion, PET with [11C]ITDM could successfully visualize hippocampal and thalamic declines of mGluR1 related to neuroinflammation, which would help further understanding for mGluR1 functions in neuroexcitotoxicity.


Assuntos
Encéfalo/metabolismo , Epilepsia/induzido quimicamente , Epilepsia/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Benzamidas/análise , Benzamidas/metabolismo , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Radioisótopos de Carbono/análise , Radioisótopos de Carbono/metabolismo , Epilepsia/diagnóstico por imagem , Epilepsia/patologia , Ligantes , Masculino , Pilocarpina , Tomografia por Emissão de Pósitrons , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/análise
8.
Mol Pain ; 13: 1744806917737934, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29020860

RESUMO

Abstract: The dorsal horn of the spinal cord is a crucial site for pain transmission and modulation. Dorsal horn neurons of the spinal cord express group I metabotropic glutamate receptors (group I mGluRs) that exert a complex role in nociceptive transmission. In particular, group I mGluRs promote the activation of L-type calcium channels, voltage-gated channels involved in short- and long-term sensitization to pain. In this study, we analyzed the role of group I mGluRs in spinal nociceptive transmission and the possible cooperation between these receptors and L-type calcium channels in the pathophysiology of pain transmission in the dorsal horn of the spinal cord. We demonstrate that the activation of group I mGluRs induces allodynia and L-type calcium channel-dependent increase in nociceptive field potentials following sciatic nerve stimulation. Surprisingly, in a model of persistent inflammation induced by complete Freund's adjuvant, the activation of group I mGluRs induced an analgesia and a decrease in nociceptive field potentials. Among the group I mGluRs, mGluR1 promotes the activation of L-type calcium channels and increased nociceptive transmission while mGluR5 induces the opposite through the inhibitory network. These results suggest a functional switch exists in pathological conditions that can change the action of group I mGluR agonists into possible analgesic molecules, thereby suggesting new therapeutic perspectives to treat persistent pain in inflammatory settings.


Assuntos
Hiperalgesia/fisiopatologia , Inflamação/metabolismo , Plasticidade Neuronal/fisiologia , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Masculino , Células do Corno Posterior/metabolismo , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/análise , Medula Espinal/fisiologia , Sinapses/metabolismo
9.
Bioorg Med Chem ; 25(3): 1014-1021, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28049619

RESUMO

Brain metabotropic glutamate receptor 2 (mGluR2) has been proposed as a therapeutic target for the treatment of schizophrenia-like symptoms arising from increased glutamate transmission in the forebrain. However, there does not exist a reliable tool for the study of mGluR2 in human neuroimaging. The purpose of this study was to radiosynthesize 1-(cyclopropylmethyl)-4-(4-[11C]methoxyphenyl)piperidin-1-yl-2-oxo-1,2-dihydropyridine-3-carbonitrile ([11C]CMDC) and evaluate its potential as a positron emission tomography (PET) radiotracer for imaging mGluR2 in the rat brain. CMDC, a positive allosteric modulator of mGluR2, showed potent functional activity (EC50: 98nM) for human mGluR2 in vitro. [11C]CMDC was synthesized by O-[11C]methylation of 1-(cyclopropylmethyl)-4-(4-hydroxyphenyl)piperidin-1-yl-2-oxo-1,2-dihydropyridine-3-carbonitrile (1) with [11C]methyl iodide. [11C]CMDC (2.2±0.9GBq; n=20) was obtained from [11C]CO2 of 14.0-17.8GBq with >98% radiochemical purity and 86-150GBq/µmol specific activity at the end of synthesis. In vitro autoradiography indicated that [11C]CMDC binding was expressed (>50% of total binding) in mGluR2-rich brain regions including the cerebral cortex, striatum and hippocampus. However, small-animal PET showed low in vivo specific binding of [11C]CMDC in the rat brain. While [11C]CMDC has limited potential as a PET tracer for brain mGluR2, it can be used to develop new radiotracers with improved behaviors.


Assuntos
Di-Hidropiridinas/química , Piperidinas/química , Tomografia por Emissão de Pósitrons , Receptores de Glutamato Metabotrópico/análise , Animais , Di-Hidropiridinas/síntese química , Di-Hidropiridinas/farmacocinética , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Estrutura Molecular , Piperidinas/síntese química , Piperidinas/farmacocinética , Traçadores Radioativos , Ratos , Ratos Sprague-Dawley , Distribuição Tecidual
10.
Proteomics ; 16(20): 2698-2705, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27392515

RESUMO

The group 1 metabotropic glutamate receptors 1 and 5 (mGluR1/5) have been implicated in mechanisms of synaptic plasticity and may serve as potential therapeutic targets in autism spectrum disorders. The interactome of group 1 mGluRs has remained largely unresolved. Using a knockout-controlled interaction proteomics strategy we examined the mGluR5 protein complex in two brain regions, hippocampus and cortex, and identified mGluR1 as its major interactor in addition to the well described Homer proteins. We confirmed the presence of mGluR1/5 complex by (i) reverse immunoprecipitation using an mGluR1 antibody to pulldown mGluR5 from hippocampal tissue, (ii) coexpression in HEK293 cells followed by coimmunoprecipitation to reveal the direct interaction of mGluR1 and 5, and (iii) superresolution microscopy imaging of hippocampal primary neurons to show colocalization of the mGluR1/5 in the synapse.


Assuntos
Córtex Cerebral/metabolismo , Hipocampo/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animais , Células HEK293 , Humanos , Imunoprecipitação , Camundongos , Receptor de Glutamato Metabotrópico 5/análise , Receptores de Glutamato Metabotrópico/análise
11.
Bioorg Med Chem Lett ; 26(2): 370-374, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26707390

RESUMO

ADX88178 (1) has been recently developed as a potent positive allosteric modulator for metabotropic glutamate receptor 4 (mGluR4). The aim of this study was to develop [(11)C]1 as a novel positron emission tomography ligand and to evaluate its binding ability for mGluR4. Using stannyl precursor 3, [(11)C]1 was efficiently synthesized by introducing an [(11)C]methyl group into a pyrimidine ring via C-(11)C coupling and deprotection reactions, in 16±6% radiochemical yield (n=10). At the end of synthesis, 0.54-1.10GBq of [(11)C]1 was acquired with >98% radiochemical purity and 90-120GBq/µmol of specific activity. In vitro autoradiography and ex vivo biodistribution study in rat brains showed specific binding of [(11)C]1 in the cerebellum, striatum, thalamus, cerebral cortex, and medulla oblongata, which showed dose-dependent decreases by administration with multi-dose of unlabeled 1.


Assuntos
Encéfalo/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Pirimidinas/metabolismo , Compostos Radiofarmacêuticos/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Tiazóis/metabolismo , Animais , Autorradiografia , Encéfalo/diagnóstico por imagem , Masculino , Pirimidinas/síntese química , Pirimidinas/química , Pirimidinas/farmacocinética , Compostos Radiofarmacêuticos/síntese química , Compostos Radiofarmacêuticos/química , Compostos Radiofarmacêuticos/farmacocinética , Ratos , Ratos Sprague-Dawley , Receptores de Glutamato Metabotrópico/análise , Tiazóis/síntese química , Tiazóis/química , Tiazóis/farmacocinética , Distribuição Tecidual
12.
J Histochem Cytochem ; 63(6): 417-26, 2015 06.
Artigo em Inglês | MEDLINE | ID: mdl-25739438

RESUMO

The inhibitory metabotropic glutamate receptor 3 (mGluR3) plays diverse and complex roles in brain function, including synaptic plasticity and neurotransmission. We recently found that mGluR3 is downregulated in the lateral septum (LS) of postpartum females using microarray and qPCR analysis. In this study, we used double fluorescence immunohistochemical approaches to characterize mGluR3 changes in LS of the postpartum brain. The number of mGluR3-immunoractive cells was significantly reduced in the dorsal (LSD) and intermediate (LSI) but not ventral (LSV) parts of the LS in postpartum versus virgin females. mGluR3 immunoreactivity in the LS was found predominantly in neurons (~70%), with a smaller portion (~20%-30%) in astrocytes. Colocalization analysis revealed a reduced mGluR3 expression in neurons but an increased astrocytic localization in postpartum LSI. This change in the pattern of expression suggests that mGluR3 expression is shifted from neurons to astrocytes in postpartum LS, and the decrease in mGluR3 is neuron-specific. Because mGluR3 is inhibitory and negatively regulates glutamate and GABA release, decreases in neuronal expression would increase glutamate and GABA signaling. Given our recent finding that ~90% of LS neurons are GABAergic, the present data suggest that decreases in mGluR3 are a mechanism for elevated GABA in LS in the postpartum state.


Assuntos
Astrócitos/citologia , Camundongos , Neurônios/citologia , Receptores de Glutamato Metabotrópico/análise , Núcleos Septais/citologia , Animais , Astrócitos/química , Astrócitos/metabolismo , Feminino , Imuno-Histoquímica , Camundongos/fisiologia , Neurônios/química , Neurônios/metabolismo , Período Pós-Parto , Receptores de Glutamato Metabotrópico/metabolismo , Núcleos Septais/química , Núcleos Septais/fisiologia
13.
J Med Chem ; 58(3): 1513-23, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25602363

RESUMO

Metabotropic glutamate 1 (mGlu1) receptor is found not only in the brain but also in melanomas and breast cancers. mGlu1 is a promising target for molecular imaging-based diagnosis and treatment of melanoma because its overexpression induces melanocyte carcinogenesis. Here we developed three PET tracers: 4-halogeno-N-[4-[6-(isopropylamino)pyrimidin-4-yl]-1,3-thiazol- 2-yl]-N-[(11)C]methylbenzamide ([(11)C]4-6), which exhibited high uptake in target tumor and decreased uptake in nontarget brain tissues. In vitro binding assay indicated high to moderate binding affinities of 4-6 (Ki, 22-143 nM) for mGlu1 receptor. In vivo biodistribution studies in mice implanted with B16F10 melanoma cells confirmed high radioactive uptake in tumor and low uptake in blood, skin, and muscles. Inhibition of mGlu1 receptor using an mGlu1-selective ligand led to reduced radioactive uptake in the tumor. [(11)C]6 displayed the highest ratio of uptake between tumor and nontarget tissue and may prove useful as a PET tracer for mGlu1 imaging in melanoma.


Assuntos
Benzamidas , Melanoma/diagnóstico , Melanoma/metabolismo , Imagem Molecular , Neoplasias Experimentais/diagnóstico , Neoplasias Experimentais/metabolismo , Receptores de Glutamato Metabotrópico/análise , Tiazóis , Animais , Benzamidas/síntese química , Benzamidas/química , Relação Dose-Resposta a Droga , Humanos , Masculino , Melanoma/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Receptores de Glutamato Metabotrópico/metabolismo , Relação Estrutura-Atividade , Tiazóis/síntese química , Tiazóis/química , Distribuição Tecidual
14.
Mol Pharmacol ; 86(6): 774-85, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25326002

RESUMO

Although G protein-coupled receptors are primarily known for converting extracellular signals into intracellular responses, some receptors, such as the group 1 metabotropic glutamate receptor, mGlu5, are also localized on intracellular membranes where they can mediate both overlapping and unique signaling effects. Thus, besides "ligand bias," whereby a receptor's signaling modality can shift from G protein dependence to independence, canonical mGlu5 receptor signaling can also be influenced by "location bias" (i.e., the particular membrane and/or cell type from which it signals). Because mGlu5 receptors play important roles in both normal development and in disorders such as Fragile X syndrome, autism, epilepsy, addiction, anxiety, schizophrenia, pain, dyskinesias, and melanoma, a large number of drugs are being developed to allosterically target this receptor. Therefore, it is critical to understand how such drugs might be affecting mGlu5 receptor function on different membranes and in different brain regions. Further elucidation of the site(s) of action of these drugs may determine which signal pathways mediate therapeutic efficacy.


Assuntos
Receptor de Glutamato Metabotrópico 5/fisiologia , Receptores de Glutamato Metabotrópico/fisiologia , Transdução de Sinais/fisiologia , Animais , Arrestinas/fisiologia , Cálcio/metabolismo , Humanos , Fosforilação , Receptor de Glutamato Metabotrópico 5/análise , Receptor de Glutamato Metabotrópico 5/química , Receptor de Glutamato Metabotrópico 5/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , beta-Arrestinas
15.
J Nanobiotechnology ; 12: 27, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25123848

RESUMO

BACKGROUND: Glutamate, the main excitatory neurotransmitter, is involved in learning and memory processes but at higher concentration results excitotoxic causing degeneration and neuronal death. Adenosine is a nucleoside that exhibit neuroprotective effects by modulating of glutamate release. Hypoxic and related oxidative conditions, in which adenosine and metabotropic glutamate receptors are involved, have been demonstrated to contribute to neurodegenerative processes occurring in certain human pathologies. RESULTS: Human neuroblastoma cells (SH-SY5Y) were used to evaluate the long time (24, 48 and 72 hours) effects of a [60]fullerene hydrosoluble derivative (t3ss) as potential inhibitor of hypoxic insult. Low oxygen concentration (5% O2) caused cell death, which was avoided by t3ss exposure in a concentration dependent manner. In addition, gene expression analysis by real time PCR of adenosine A1, A2A and A2B and metabotropic glutamate 1 and 5 receptors revealed that t3ss significantly increased A1 and mGlu1 expression in hypoxic conditions. Moreover, t3ss prevented the hypoxia-induced increase in A2A mRNA expression. CONCLUSIONS: As t3ss causes overexpression of adenosine A1 and metabotropic glutamate receptors which have been shown to be neuroprotective, our results point to a radical scavenger protective effect of t3ss through the enhancement of these neuroprotective receptors expression. Therefore, the utility of these nanoparticles as therapeutic target to avoid degeneration and cell death of neurodegenerative diseases is suggested.


Assuntos
Adenosina/genética , Hipóxia Celular/fisiologia , Fulerenos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Receptores de Glutamato Metabotrópico/genética , Adenosina/análise , Adenosina/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Fulerenos/química , Humanos , Substâncias Protetoras/química , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/metabolismo , Sais de Tetrazólio , Tiazóis
16.
J Neurosci ; 34(30): 9891-904, 2014 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-25057192

RESUMO

Spinocerebellar ataxia type 5 (SCA5), a dominant neurodegenerative disease characterized by profound Purkinje cell loss, is caused by mutations in SPTBN2, a gene that encodes ß-III spectrin. SCA5 is the first neurodegenerative disorder reported to be caused by mutations in a cytoskeletal spectrin gene. We have developed a mouse model to understand the mechanistic basis for this disease and show that expression of mutant but not wild-type ß-III spectrin causes progressive motor deficits and cerebellar degeneration. We show that endogenous ß-III spectrin interacts with the metabotropic glutamate receptor 1α (mGluR1α) and that mice expressing mutant ß-III spectrin have cerebellar dysfunction with altered mGluR1α localization at Purkinje cell dendritic spines, decreased mGluR1-mediated responses, and deficient mGluR1-mediated long-term potentiation. These results indicate that mutant ß-III spectrin causes mislocalization and dysfunction of mGluR1α at dendritic spines and connects SCA5 with other disorders involving glutamatergic dysfunction and synaptic plasticity abnormalities.


Assuntos
Modelos Animais de Doenças , Mutação/genética , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/genética , Espectrina/genética , Ataxias Espinocerebelares/genética , Animais , Cerebelo/química , Cerebelo/patologia , Espinhas Dendríticas/química , Espinhas Dendríticas/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Receptores de Glutamato Metabotrópico/metabolismo , Ataxias Espinocerebelares/fisiopatologia
17.
J Cancer Res Clin Oncol ; 140(3): 419-26, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24399291

RESUMO

PURPOSE: Analyze protein and gene expression of mGluRs (mGluR1, mGluR5, mGluR4) in osteosarcoma tissues and discuss the relation between expression level and clinical characteristics of osteosarcoma, and study the clinical significance. METHODS: Detect protein and mRNA expression level of mGluRs (mGluR1, mGluR5, mGluR4) in 40 osteosarcoma tissues and the corresponding adjacent normal tissues by Western blot and RT-PCR accordingly. Immunohistochemistry was adopted to detect the expression of mGluRs (mGluR1, mGluR5, mGluR4) in 118 paraffin embedded osteosarcoma tissues and eight normal bone tissues. Then, the correlation between the expression and clinical characteristics of patients was analyzed. Furthermore, survival analysis of osteosarcoma was performed to study the relation between expression level of mGluRs and patient prognosis. RESULTS: No correlation of mGluR1 and mGluR5 with clinicopathologic characteristics of osteosarcoma was found. Statistical analysis demonstrated that the expression level of mGluR4 shared no significant correlation with gender, age, histologic type and tumor location of patient, but was related to Enneking stage and tumor metastasis (P < 0.05). High mGluR4 expression is more frequently noted in the osteosarcoma tissues with higher Enneking stage and metastasis. The results of Western blot and RT-PCR indicated a significantly increased expression level of mGluR4 gene and protein in osteosarcoma tissues compared with normal tissues. Though higher gene and protein expression of mGluR5 and mGluR1 were also indicated in osteosarcoma tissues compared with normal tissues, no statistical significance was noted for the difference (P > 0.05). According to the survival analysis of 118 osteosarcoma patients, cases in the mGluR4 high-expression group showed inferior disease-free survival rate and poorer overall survival rate. CONCLUSION: High expression of mGluR4 in osteosarcoma tissues is related to poor prognosis, thus holding certain reference value for estimating prognosis of osteosarcoma patients.


Assuntos
Neoplasias Ósseas/química , Neoplasias Ósseas/patologia , Osteossarcoma/química , Osteossarcoma/patologia , Receptores de Glutamato Metabotrópico/análise , Adulto , Idoso , Western Blotting , Neoplasias Ósseas/mortalidade , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Osteossarcoma/mortalidade , Valor Preditivo dos Testes , Prognóstico , RNA Mensageiro/análise , Receptores de Glutamato Metabotrópico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Pharmacol Ther ; 142(3): 281-305, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24362085

RESUMO

Glutamatergic excitatory transmission is implicated in physiological and pathological conditions like learning, memory, neuronal plasticity and emotions, while glutamatergic abnormalities are reported in numerous neurological and psychiatric disorders, including neurodegenerative diseases, epilepsy, stroke, traumatic brain injury, depression, anxiety, schizophrenia and pain. Also, several lines of evidence have accumulated indicating a pivotal role for glutamatergic neurotransmission in mediating addictive behaviors. Among the proteins regulating glutamatergic transmission, the metabotropic glutamate receptors (mGluR) are being developed as pharmacological targets for treating many neuropsychiatric disorders, including drug addiction. In this review we describe the molecular structure of mGluRs and their distribution, physiology and pharmacology in the central nervous system, as well as their use as targets in preclinical studies of drug addiction.


Assuntos
Receptores de Glutamato Metabotrópico/fisiologia , Transtornos Relacionados ao Uso de Substâncias/etiologia , Animais , Ácido Glutâmico/metabolismo , Humanos , Ligantes , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/genética , Transtornos Relacionados ao Uso de Substâncias/tratamento farmacológico
19.
Eur J Neurosci ; 37(1): 118-29, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23134476

RESUMO

Immunohistochemical studies previously revealed the presence of the peptide transmitter N-acetylaspartylglutamate (NAAG) in spinal motor neurons, axons and presumptive neuromuscular junctions (NMJs). At synapses in the central nervous system, NAAG has been shown to activate the type 3 metabotropic glutamate receptor (mGluR3) and is inactivated by an extracellular peptidase, glutamate carboxypeptidase II. The present study tested the hypothesis that NAAG meets the criteria for classification as a co-transmitter at the vertebrate NMJ. Confocal microscopy confirmed the presence of NAAG immunoreactivity and extended the resolution of the peptide's location in the lizard (Anolis carolinensis) NMJ. NAAG was localised to a presynaptic region immediately adjacent to postsynaptic acetylcholine receptors. NAAG was depleted by potassium-induced depolarisation and by electrical stimulation of motor axons. The NAAG receptor, mGluR3, was localised to the presynaptic terminal consistent with NAAG's demonstrated role as a regulator of synaptic release at central synapses. In contrast, glutamate receptors, type 2 metabotropic glutamate receptor (mGluR2) and N-methyl-d-aspartate, were closely associated with acetylcholine receptors in the postsynaptic membrane. Glutamate carboxypeptidase II, the NAAG-inactivating enzyme, was identified exclusively in perisynaptic glial cells. This localisation was confirmed by the loss of immunoreactivity when these cells were selectively eliminated. Finally, electrophysiological studies showed that exogenous NAAG inhibited evoked neurotransmitter release by activating a group II metabotropic glutamate receptor (mGluR2 or mGluR3). Collectively, these data support the conclusion that NAAG is a co-transmitter at the vertebrate NMJ.


Assuntos
Dipeptídeos/farmacologia , Potenciais Pós-Sinápticos em Miniatura/efeitos dos fármacos , Junção Neuromuscular/química , Neurotransmissores/farmacologia , Animais , Dipeptídeos/análise , Agonistas de Aminoácidos Excitatórios/farmacologia , Glutamato Carboxipeptidase II/análise , Imuno-Histoquímica , Lagartos , Neurônios Motores/química , Neurônios Motores/fisiologia , N-Metilaspartato/farmacologia , Junção Neuromuscular/fisiologia , Potássio/farmacologia , Terminações Pré-Sinápticas/química , Receptores Colinérgicos/análise , Receptores de Glutamato Metabotrópico/análise
20.
Handb Exp Pharmacol ; (213): 297-365, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23027420

RESUMO

Metabotropic glutamate receptors (mGluRs) represent exciting targets for the development of novel therapeutic agents for schizophrenia. Recent studies indicate that selective activation of specific mGluR subtypes may provide potential benefits for not only the positive symptoms, but also the negative symptoms and cognitive impairments observed in individuals with schizophrenia. Although optimization of traditional orthosteric agonists may still offer a feasible approach for the activation of mGluRs, important progress has been made in the discovery of novel subtype-selective allosteric ligands, including positive allosteric modulators (PAMs) of mGluR2 and mGluR5. These allosteric mGluR ligands have improved properties for clinical development and have served as key preclinical tools for a more in-depth understanding of the potential roles of these different mGluR subtypes for the treatment of schizophrenia.


Assuntos
Receptores de Glutamato Metabotrópico/fisiologia , Esquizofrenia/tratamento farmacológico , Regulação Alostérica , Animais , Cognição/efeitos dos fármacos , Humanos , Ligantes , Receptores de Glutamato Metabotrópico/análise , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA