Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Pharmacol ; 104(1): 1-16, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37147110

RESUMEN

Current treatments for Alzheimer's disease (AD) help reduce symptoms for a limited time but do not treat the underlying pathology. To identify potential therapeutic targets for AD, an integrative network analysis was previously carried out using 364 human postmortem control, mild cognitive impairment, and AD brains. This analysis identified proline endopeptidase-like protein (PREPL), an understudied protein, as a downregulated protein in late-onset AD patients. In this study we investigate the role of PREPL. Analyses of data from human postmortem samples and PREPL knockdown (KD) cells suggest that PREPL expression modulates pathways associated with protein trafficking, synaptic activities, and lipid metabolism. Furthermore, PREPL KD impairs cell proliferation and modulates the structure of vesicles, levels of neuropeptide-processing enzymes, and secretion of neuropeptides. In addition, decrease in PREPL levels leads to changes in the levels of a number of synaptic proteins as well as changes in the levels of secreted amyloid beta (Aß) 42 peptide and Tau phosphorylation. Finally, we report that local decrease in PREPL levels in mouse hippocampus attenuates long-term potentiation, suggesting a role in synaptic plasticity. Together, our results indicate that PREPL affects neuronal function by modulating protein trafficking and synaptic function, an important mechanism of AD pathogenesis. SIGNIFICANCE STATEMENT: Integrative network analysis reveals proline endopeptidase-like protein (PREPL) to be downregulated in human sporadic late-onset Alzheimer's disease brains. Down regulation of PREPL leads to increases in amyloid beta secretion, Tau phosphorylation, and decreases in protein trafficking and long-term potentiation.


Asunto(s)
Enfermedad de Alzheimer , Prolil Oligopeptidasas , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , Multiómica , Prolil Oligopeptidasas/metabolismo , Transporte de Proteínas
2.
Mol Pharmacol ; 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35605991

RESUMEN

PEN is an abundant neuropeptide that activates GPR83, a G protein-coupled receptor that is considered a novel therapeutic target due to its roles in regulation of feeding, reward, and anxiety-related behaviors. The major form of PEN in the brain is 22 residues in length. Previous studies have identified shorter forms of PEN in mouse brain and neuroendocrine cells; these shorter forms were named PEN18, PEN19 and PEN20, with the number reflecting the length of the peptide. The C-terminal five residues of PEN20 are identical to the C-terminus of a procholecystokinin (proCCK)-derived peptide, named proCCK56-62, that is present in mouse brain. ProCCK56-62 is highly conserved across species although it has no homology to the bioactive cholecystokinin domain. ProCCK56-62 and a longer form, proCCK56-63 were tested for their ability to engage GPR83. Both peptides bind GPR83 with high affinity, activate second messenger pathways, and induce ligand-mediated receptor endocytosis. Interestingly, the shorter PEN peptides, ProCC56-62, and ProCCK56-63 differentially activate signal transduction pathways. Whereas PEN22 and PEN20 facilitate receptor coupling to Gai, PEN18, PEN19 and ProCCK peptides facilitate coupling to Gas. Furthermore, the ProCCK peptides exhibit dose dependent Ga subtype selectivity in that they faciliate coupling to Gas at low concentrations and Gai at high concentrations. These data demonstrate that peptides derived from two distinct peptide precursors can differentially activate GPR83, and that GPR83 exhibits Ga subtype preference depending on the nature and concentration of the peptide. These results are consistent with the emerging idea that endogenous neuropeptides function as biased ligands. Significance Statement We found that peptides derived from proCCK bind and activate GPR83, a G protein-coupled receptor that is known to bind peptides derived from proSAAS. Different forms of the proCCK- and proSAAS-derived peptides show biased agonism, activating Gas or Gai depending on the length of the peptide and/or its concentration.

3.
J Neurosci ; 38(11): 2780-2795, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29459374

RESUMEN

A homozygous nonsense mutation in the cereblon (CRBN) gene results in autosomal recessive, nonsyndromic intellectual disability that is devoid of other phenotypic features, suggesting a critical role of CRBN in mediating learning and memory. In this study, we demonstrate that adult male Crbn knock-out (CrbnKO) mice exhibit deficits in hippocampal-dependent learning and memory tasks that are recapitulated by focal knock-out of Crbn in the adult dorsal hippocampus, with no changes in social or repetitive behavior. Cellular studies identify deficits in long-term potentiation at Schaffer collateral CA1 synapses. We further show that Crbn is robustly expressed in the mouse hippocampus and CrbnKO mice exhibit hyperphosphorylated levels of AMPKα (Thr172). Examination of processes downstream of AMP-activated protein kinase (AMPK) finds that CrbnKO mice have a selective impairment in mediators of the mTORC1 translation initiation pathway in parallel with lower protein levels of postsynaptic density glutamatergic proteins and higher levels of excitatory presynaptic markers in the hippocampus with no change in markers of the unfolded protein response or autophagy pathways. Acute pharmacological inhibition of AMPK activity in adult CrbnKO mice rescues learning and memory deficits and normalizes hippocampal mTORC1 activity and postsynaptic glutamatergic proteins without altering excitatory presynaptic markers. Thus, this study identifies that loss of Crbn results in learning, memory, and synaptic defects as a consequence of exaggerated AMPK activity, inhibition of mTORC1 signaling, and decreased glutamatergic synaptic proteins. Thus, CrbnKO mice serve as an ideal model of intellectual disability to further explore molecular mechanisms of learning and memory.SIGNIFICANCE STATEMENT Intellectual disability (ID) is one of the most common neurodevelopmental disorders. The cereblon (CRBN) gene has been linked to autosomal recessive, nonsyndromic ID, characterized by an intelligence quotient between 50 and 70 but devoid of other phenotypic features, making cereblon an ideal protein for the study of the fundamental aspects of learning and memory. Here, using the cereblon knock-out mouse model, we show that cereblon deficiency disrupts learning, memory, and synaptic function via AMP-activated protein kinase hyperactivity, downregulation of mTORC1, and dysregulation of excitatory synapses, with no changes in social or repetitive behaviors, consistent with findings in the human population. This establishes the cereblon knock-out mouse as a model of pure ID without the confounding behavioral phenotypes associated with other current models of ID.


Asunto(s)
Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Discapacidades para el Aprendizaje/genética , Discapacidades para el Aprendizaje/fisiopatología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Proteínas del Tejido Nervioso/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Región CA1 Hipocampal/fisiopatología , Potenciales Postsinápticos Excitadores/genética , Hipocampo/metabolismo , Hipocampo/fisiopatología , Discapacidad Intelectual/tratamiento farmacológico , Discapacidades para el Aprendizaje/tratamiento farmacológico , Potenciación a Largo Plazo/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/biosíntesis , Trastornos de la Memoria/tratamiento farmacológico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Inhibidores de Proteínas Quinasas/uso terapéutico , Conducta Social
4.
Proc Natl Acad Sci U S A ; 113(21): 6041-6, 2016 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-27162327

RESUMEN

Among the opioid receptors, the κ-opioid receptor (κOR) has been gaining considerable attention as a potential therapeutic target for the treatment of complex CNS disorders including depression, visceral pain, and cocaine addiction. With an interest in discovering novel ligands targeting κOR, we searched natural products for unusual scaffolds and identified collybolide (Colly), a nonnitrogenous sesquiterpene from the mushroom Collybia maculata. This compound has a furyl-δ-lactone core similar to that of Salvinorin A (Sal A), another natural product from the plant Salvia divinorum Characterization of the molecular pharmacological properties reveals that Colly, like Sal A, is a highly potent and selective κOR agonist. However, the two compounds differ in certain signaling and behavioral properties. Colly exhibits 10- to 50-fold higher potency in activating the mitogen-activated protein kinase pathway compared with Sal A. Taken with the fact that the two compounds are equipotent for inhibiting adenylyl cyclase activity, these results suggest that Colly behaves as a biased agonist of κOR. Behavioral studies also support the biased agonistic activity of Colly in that it exhibits ∼10-fold higher potency in blocking non-histamine-mediated itch compared with Sal A, and this difference is not seen in pain attenuation by these two compounds. These results represent a rare example of functional selectivity by two natural products that act on the same receptor. The biased agonistic activity, along with an easily modifiable structure compared with Sal A, makes Colly an ideal candidate for the development of novel therapeutics targeting κOR with reduced side effects.


Asunto(s)
Agaricales/química , Antipruriginosos/farmacología , Diterpenos de Tipo Clerodano/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Receptores Opioides kappa/agonistas , Sistemas de Mensajero Secundario/efectos de los fármacos , Animales , Antipruriginosos/química , Diterpenos de Tipo Clerodano/química , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo
5.
J Neurosci ; 34(2): 527-38, 2014 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-24403152

RESUMEN

Learned associations between environmental cues and morphine use play an important role in the maintenance and/or relapse of opioid addiction. Although previous studies suggest that context-dependent morphine treatment alters glutamatergic transmission and synaptic plasticity in the hippocampus, their role in morphine conditioned place preference (CPP) and reinstatement remains unknown. We investigated changes in synaptic plasticity and NMDAR expression in the hippocampus after the expression, extinction, and reinstatement of morphine CPP. Here we report that morphine CPP is associated with increased basal synaptic transmission, impaired hippocampal long-term potentiation (LTP), and increased synaptic expression of the NR1 and NR2b NMDAR subunits. Changes in synaptic plasticity, synaptic NR1 and NR2b expression, and morphine CPP were absent when morphine was not paired with a specific context. Furthermore, hippocampal LTP was impaired and synaptic NR2b expression was increased after extinction of morphine CPP, indicating that these alterations in plasticity may be involved in the mechanisms underlying the learning of drug-environment associations. After extinction of morphine CPP, a priming dose of morphine was sufficient to reinstate morphine CPP and was associated with LTP that was indistinguishable from saline control groups. In contrast, morphine CPP extinguished mice that received a saline priming dose did not show CPP and had disrupted hippocampal LTP. Finally, we found that reinstatement of morphine CPP was prevented by the selective blockade of the NR2b subunit in the hippocampus. Together, these data suggest that alterations in synaptic plasticity and glutamatergic transmission play an important role in the reinstatement of morphine CPP.


Asunto(s)
Extinción Psicológica/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Dependencia de Morfina/fisiopatología , Transmisión Sináptica/fisiología , Animales , Condicionamiento Clásico , Señales (Psicología) , Electrofisiología , Potenciales Postsinápticos Excitadores/fisiología , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de N-Metil-D-Aspartato/metabolismo
6.
Mol Cell Neurosci ; 51(1-2): 22-31, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22789621

RESUMEN

Purkinje cell (PC) dysfunction or death has been implicated in a number of disorders including ataxia, autism and multiple sclerosis. Plasma membrane calcium ATPase 2 (PMCA2), an important calcium (Ca(2+)) extrusion pump that interacts with synaptic signaling complexes, is most abundantly expressed in PCs compared to other neurons. Using the PMCA2 heterozygous mouse as a model, we investigated whether a reduction in PMCA2 levels affects PC function. We focused on Ca(2+) signaling and the expression of glutamate receptors which play a key role in PC function including synaptic plasticity. We found that the amplitude of depolarization and 2-amino-3-(5-methyl-3-oxo-1,2-oxazol-4-yl)propanoic acid receptor (AMPAR)-mediated Ca(2+) transients are significantly higher in cultured PMCA2(+/-) PCs than in PMCA2(+/+) PCs. This is due to increased Ca(2+) influx, since P/Q type voltage-gated Ca(2+) channel (VGCC) expression was more pronounced in PCs and cerebella of PMCA2(+/-) mice and VGCC blockade prevented the elevation in amplitude. Neuronal nitric oxide synthase (nNOS) activity was higher in PMCA2(+/-) cerebella and inhibition of nNOS or the soluble guanylate cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway, which mediates nitric oxide (NO) signaling, reduced the amplitude of Ca(2+) transients in PMCA2(+/-) PCs, in vitro. In addition, there was an age-dependent decrease in metabotropic glutamate receptor 1 (mGluR1) and AMPA receptor subunit GluR2/3 transcript and protein levels at 8 weeks of age. These changes were followed by PC loss in the 20-week-old PMCA2(+/-) mice. Our studies highlight the importance of PMCA2 in Ca(2+) signaling, glutamate receptor expression and survival of Purkinje cells.


Asunto(s)
ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Células de Purkinje/metabolismo , Factores de Edad , Animales , Calcio/metabolismo , Canales de Calcio/metabolismo , Señalización del Calcio , Muerte Celular , Células Cultivadas , Regulación de la Expresión Génica , Heterocigoto , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Células de Purkinje/citología , Receptores AMPA/genética , Receptores AMPA/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Transcripción Genética
7.
bioRxiv ; 2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37205394

RESUMEN

Hyperexcitability in the orbitofrontal cortex (OFC) is a key clinical feature of anhedonic domains of Major Depressive Disorder (MDD). However, the cellular and molecular substrates underlying this dysfunction remain unknown. Here, cell-population-specific chromatin accessibility profiling in human OFC unexpectedly mapped genetic risk for MDD exclusively to non-neuronal cells, and transcriptomic analyses revealed significant glial dysregulation in this region. Characterization of MDD-specific cis-regulatory elements identified ZBTB7A - a transcriptional regulator of astrocyte reactivity - as an important mediator of MDD-specific chromatin accessibility and gene expression. Genetic manipulations in mouse OFC demonstrated that astrocytic Zbtb7a is both necessary and sufficient to promote behavioral deficits, cell-type-specific transcriptional and chromatin profiles, and OFC neuronal hyperexcitability induced by chronic stress - a major risk factor for MDD. These data thus highlight a critical role for OFC astrocytes in stress vulnerability and pinpoint ZBTB7A as a key dysregulated factor in MDD that mediates maladaptive astrocytic functions driving OFC hyperexcitability.

8.
J Neurosci ; 31(45): 16279-91, 2011 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-22072679

RESUMEN

Glutamatergic systems, including AMPA receptors (AMPARs), are involved in opiate-induced neuronal and behavioral plasticity, although the mechanisms underlying these effects are not fully understood. In the present study, we investigated the effects of repeated morphine administration on AMPAR expression, synaptic plasticity, and context-dependent behavioral sensitization to morphine. We found that morphine treatment produced changes of synaptic AMPAR expression in the hippocampus, a brain area that is critically involved in learning and memory. These changes could be observed 1 week after the treatment, but only when mice developed context-dependent behavioral sensitization to morphine in which morphine treatment was associated with drug administration environment. Context-dependent behavioral sensitization to morphine was also associated with increased basal synaptic transmission and disrupted hippocampal long-term potentiation (LTP), whereas these effects were less robust when morphine administration was not paired with the drug administration environment. Interestingly, some effects may be related to the prior history of morphine exposure in the drug-associated environment, since alterations of AMPAR expression, basal synaptic transmission, and LTP were observed in mice that received a saline challenge 1 week after discontinuation of morphine treatment. Furthermore, we demonstrated that phosphorylation of GluA1 AMPAR subunit plays a critical role in the acquisition and expression of context-dependent behavioral sensitization, as this behavior is blocked by a viral vector that disrupts GluA1 phosphorylation. These data provide evidence that glutamatergic signaling in the hippocampus plays an important role in context-dependent sensitization to morphine and supports further investigation of glutamate-based strategies for treating opiate addiction.


Asunto(s)
Hipocampo/efectos de los fármacos , Dependencia de Morfina/metabolismo , Morfina/farmacología , Actividad Motora/efectos de los fármacos , Narcóticos/farmacología , Receptores AMPA/metabolismo , Alanina/genética , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Guanilato-Quinasas/metabolismo , Etiquetado Corte-Fin in Situ/métodos , Potenciación a Largo Plazo/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Proteínas Nucleares/metabolismo , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Distribución Aleatoria , Receptores AMPA/genética , Serina/genética , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo
9.
Brain Res ; 1789: 147951, 2022 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-35618016

RESUMEN

Carboxypeptidase E (CPE) is an essential enzyme that contributes to the biosynthesis of the vast majority of neuropeptides and peptide hormones. There are several reports claiming that small decreases in CPE activity cause physiological changes in animals and/or cultured cells, but these studies did not provide evidence that neuropeptide levels were affected by decreased CPE activity. In the present study, we tested if CPE is a rate-limiting enzyme in neuropeptide production using CpeNeo mice, which contain a neomycin cassette within the Cpe gene that eliminates enzyme expression. Homozygous CpeNeo/Neo mice show defects found in Cpefat/fat and/or Cpe global knockout (KO) mice, including greatly decreased levels of most neuropeptides, severely impaired fertility, depressive-like behavior, adult-onset obesity, and anxiety-like behavior. Removal of the neomycin cassette with Flp recombinase under a germline promoter restored expression of CPE activity and resulted in normal behavioral and physiological properties, including levels of neuropeptides. Mice heterozygous for the CpeNeo allele have greatly reduced levels of Cpe mRNA and CPE-like enzymatic activity. Despite the decreased levels of Cpe expression, heterozygous CpeNeo mice are behaviorally and physiologically identical to wild-type mice, with normal levels of most neuropeptides. These results indicate that CPE is not a rate-limiting enzyme in the production of most neuropeptides, casting doubt upon studies claiming small decreases in CPE activity contribute to obesity or other physiological effects.


Asunto(s)
Carboxipeptidasa H , Mutación con Pérdida de Función , Neuropéptidos , ARN Mensajero , Animales , Conducta Animal/efectos de los fármacos , Carboxipeptidasa H/genética , Carboxipeptidasa H/metabolismo , Ratones , Ratones Noqueados , Neomicina/farmacología , Neuropéptidos/metabolismo , Obesidad/genética , Obesidad/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
Front Neurosci ; 15: 675769, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512237

RESUMEN

Anxiety disorders are prevalent across the United States and result in a large personal and societal burden. Currently, numerous therapeutic and pharmaceutical treatment options exist. However, drugs to classical receptor targets have shown limited efficacy and often come with unpleasant side effects, highlighting the need to identify novel targets involved in the etiology and treatment of anxiety disorders. GPR83, a recently deorphanized receptor activated by the abundant neuropeptide PEN, has also been identified as a glucocorticoid regulated receptor (and named GIR) suggesting that this receptor may be involved in stress-responses that underlie anxiety. Consistent with this, GPR83 null mice have been found to be resistant to stress-induced anxiety. However, studies examining the role of GPR83 within specific brain regions or potential sex differences have been lacking. In this study, we investigate anxiety-related behaviors in male and female mice with global knockout and following local GPR83 knockdown in female mice. We find that a global knockdown of GPR83 has minimal impact on anxiety-like behaviors in female mice and a decrease in anxiety-related behaviors in male mice. In contrast, a local GPR83 knockdown in the basolateral amygdala leads to more anxiety-related behaviors in female mice. Local GPR83 knockdown in the central amygdala or nucleus accumbens (NAc) showed no significant effect on anxiety-related behaviors. Finally, dexamethasone administration leads to a significant decrease in receptor expression in the amygdala and NAc of female mice. Together, our studies uncover a significant, but divergent role for GPR83 in different brain regions in the regulation of anxiety-related behaviors, which is furthermore dependent on sex.

11.
Cell Chem Biol ; 28(1): 105-112.e4, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33217339

RESUMEN

Neuropeptides and peptide hormones are important cell-cell signaling molecules that mediate many physiological processes. Unlike classic neurotransmitters, peptides undergo cell-type-specific post-translational modifications that affect their biological activity. To enable the identification of the peptide repertoire of a genetically defined cell type, we generated mice with a conditional disruption of the gene for carboxypeptidase E (Cpe), an essential neuropeptide-processing enzyme. The loss of Cpe leads to accumulation of neuropeptide precursors containing C-terminal basic residues, which serve as tags for affinity purification. The purified peptides are subsequently identified using quantitative peptidomics, thereby revealing the specific forms of neuropeptides in cells with the disrupted Cpe gene. To validate the method, we used mice expressing Cre recombinase under the proopiomelanocortin (Pomc) promoter and analyzed hypothalamic and pituitary extracts, detecting peptides derived from proopiomelanocortin (as expected) and also proSAAS in POMC neurons. This technique enables the analyses of specific forms of peptides in any Cre-expressing cell type.


Asunto(s)
Encéfalo/metabolismo , Carboxipeptidasa H/genética , Neuropéptidos/análisis , Hipófisis/metabolismo , Animales , Encéfalo/citología , Carboxipeptidasa H/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuropéptidos/metabolismo , Hipófisis/citología
12.
Nat Commun ; 11(1): 3942, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32770063

RESUMEN

Though discovered over 100 years ago, the molecular foundation of sporadic Alzheimer's disease (AD) remains elusive. To better characterize the complex nature of AD, we constructed multiscale causal networks on a large human AD multi-omics dataset, integrating clinical features of AD, DNA variation, and gene- and protein-expression. These probabilistic causal models enabled detection, prioritization and replication of high-confidence master regulators of AD-associated networks, including the top predicted regulator, VGF. Overexpression of neuropeptide precursor VGF in 5xFAD mice partially rescued beta-amyloid-mediated memory impairment and neuropathology. Molecular validation of network predictions downstream of VGF was also achieved in this AD model, with significant enrichment for homologous genes identified as differentially expressed in 5xFAD brains overexpressing VGF. Our findings support a causal role for VGF in protecting against AD pathogenesis and progression.


Asunto(s)
Enfermedad de Alzheimer/etiología , Encéfalo/patología , Factores de Crecimiento Nervioso/metabolismo , Mapas de Interacción de Proteínas , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Ratones Transgénicos , Factores de Crecimiento Nervioso/genética , Mapeo de Interacción de Proteínas , Proteómica
13.
Neuropharmacology ; 157: 107666, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31199956

RESUMEN

GPR83, the receptor for the neuropeptide PEN, exhibits high expression in the nucleus accumbens of the human and rodent brain, suggesting that it plays a role in modulating the mesolimbic reward pathway. However, the cell-type specific expression of GPR83, its functional impact in the reward pathway, and in drug reward-learning has not been fully explored. Using GPR83/eGFP mice, we show high GPR83 expression on cholinergic interneurons in the nucleus accumbens and moderate expression on ventral tegmental area dopamine neurons. In GPR83 knockout mice, baseline dopamine release in the nucleus accumbens is enhanced which disrupts the ratio of tonic vs phasic release. Additionally, GPR83 knockout leads to changes in the expression of dopamine-related genes. Using the morphine conditioned place preference model, we identify sex differences in morphine reward-learning, show that GPR83 is upregulated in the nucleus accumbens following morphine conditioned place preference, and show that shRNA-mediated knockdown of GPR83 in the nucleus accumbens leads to attenuation morphine reward. Together, these findings detect GPR83 expression in the reward-pathway, and show its involvement in dopamine release and morphine reward-learning.


Asunto(s)
Aprendizaje/fisiología , Receptores Acoplados a Proteínas G/fisiología , Recompensa , Caracteres Sexuales , Animales , Neuronas Colinérgicas/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Interneuronas/metabolismo , Aprendizaje/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Morfina/farmacología , Núcleo Accumbens/metabolismo , ARN Interferente Pequeño/farmacología , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Regulación hacia Arriba/efectos de los fármacos , Área Tegmental Ventral/metabolismo
14.
Prog Mol Biol Transl Sci ; 159: 1-25, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30340784

RESUMEN

G-protein coupled receptors (GPCRs) are a superfamily of receptors responsible for initiation of a myriad of intracellular signaling cascades. Currently, GPCRs represent approximately 34% of marketed pharmaceuticals, a large portion of which have no known endogenous ligand. These orphan GPCRs represent a large pool of novel targets for drug development. Very recently, the neuropeptide PEN, derived from the proteolytic processing of the precursor proSAAS, has been identified as a selective, high-affinity endogenous ligand for the orphan receptor, GPR83. GPR83 is highly expressed in the brain, spleen and thymus, indicating that this receptor may be a target to treat neurological and immune disorders. In the brain GPR83 is expressed in regions involved in the reward pathway, stress/anxiety responses, learning and memory and metabolism. However, the cell type specific expression of GPR83 in these regions has only recently begun to be characterized. In the immune system, GPR83 expression is regulated by Foxp3 in T-regulatory cells that are involved in autoimmune responses. Moreover, in the brain this receptor is regulated by interactions with other GPCRs, such as the recently deorphanized receptor, GPR171, and other hypothalamic receptors such as MC4R and GHSR. The following review will summarize the properties of GPR83 and highlight its known and potential significance in health and disease, as well as its promise as a novel target for drug development.


Asunto(s)
Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/inmunología , Terapia Molecular Dirigida , Sistemas Neurosecretores/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Transducción de Señal
15.
Front Neurosci ; 12: 886, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30542261

RESUMEN

Opioids remain among the most effective pain-relieving therapeutics. However, their long-term use is limited due to the development of tolerance and potential for addiction. For many years, researchers have explored the underlying mechanisms that lead to this decreased effectiveness of opioids after repeated use, and numerous theories have been proposed to explain these changes. The most widely studied theories involve alterations in receptor trafficking and intracellular signaling. Other possible mechanisms include the recruitment of new structural neuronal and microglia networks. While many of these theories have been developed using molecular and cellular techniques, more recent behavioral data also supports these findings. In this review, we focus on the mechanisms that underlie tolerance within the descending pain modulatory pathway, including alterations in intracellular signaling, neural-glial interactions, and neurotransmission following opioid exposure. Developing a better understanding of the relationship between these various mechanisms, within different parts of this pathway, is vital for the identification of more efficacious, novel therapeutics to treat chronic pain.

16.
Neuropsychopharmacology ; 41(11): 2668-78, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27170097

RESUMEN

In people with a prior history of opioid misuse, cues associated with previous drug intake can trigger relapse even after years of abstinence. Examining the processes that lead to the formation and maintenance of the memories between cues/context and the opioid may help to discover new therapeutic candidates to treat drug-seeking behavior. The hippocampus is a brain region essential for learning and memory, which has been involved in the mechanisms underlying opioid cravings. The formation of memories and associations are thought to be dependent on synaptic strengthening associated with structural plasticity of dendritic spines. Here, we assess how dendritic spines in the CA1 region of the hippocampus are affected by morphine-conditioning training. Our results show that morphine pairing with environmental cues (ie, the conditioned place preference (CPP) apparatus) triggers a significant decrease in the number of thin dendritic spines in the hippocampus. Interestingly, this effect was observed regardless of the expression of a conditioned response when mice were trained using an unpaired morphine CPP design and was absent when morphine was administered in the home cage. To investigate the mechanism underlying this structural plasticity, we examined the role of Rho GTPase in dendritic spine remodeling. We found that synaptic expression of RhoA increased with morphine conditioning and blocking RhoA signaling prevented the expression of morphine-induced CPP. Our findings uncover novel mechanisms in response to morphine-associated environmental cues and the underlying alterations in spine plasticity.


Asunto(s)
Región CA1 Hipocampal/citología , Condicionamiento Operante/efectos de los fármacos , Señales (Psicología) , Morfina/farmacología , Narcóticos/farmacología , Plasticidad Neuronal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Densidad Postsináptica/efectos de los fármacos , Densidad Postsináptica/metabolismo , Densidad Postsináptica/ultraestructura , Células Piramidales/ultraestructura , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Sinaptosomas/ultraestructura , Factores de Tiempo , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
17.
Sci Signal ; 9(425): ra43, 2016 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27117253

RESUMEN

PEN is an abundant peptide in the brain that has been implicated in the regulation of feeding. We identified a receptor for PEN in mouse hypothalamus and Neuro2A cells. PEN bound to and activated GPR83, a G protein (heterotrimeric guanine nucleotide)-binding protein)-coupled receptor (GPCR). Reduction of GPR83 expression in mouse brain and Neuro2A cells reduced PEN binding and signaling, consistent with GPR83 functioning as the major receptor for PEN. In some brain regions, GPR83 colocalized with GPR171, a GPCR that binds the neuropeptide bigLEN, another neuropeptide that is involved in feeding and is generated from the same precursor protein as is PEN. Coexpression of these two receptors in cell lines altered the signaling properties of each receptor, suggesting a functional interaction. Our data established PEN as a neuropeptide that binds GPR83 and suggested that these two ligand-receptor systems-PEN-GPR83 and bigLEN-GPR171-may be functionally coupled in the regulation of feeding.


Asunto(s)
Hipotálamo/metabolismo , Neuropéptido Y/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Regulación del Apetito/fisiología , Western Blotting , Células CHO , Membrana Celular/metabolismo , Células Cultivadas , Cricetulus , Células HEK293 , Humanos , Masculino , Ratones , Fosforilación , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética
18.
Sci Signal ; 9(410): ra5, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26758213

RESUMEN

Heterotrimeric guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs) can form multiprotein complexes (heteromers), which can alter the pharmacology and functions of the constituent receptors. Previous findings demonstrated that the Gq/11-coupled serotonin 5-HT2A receptor and the Gi/o-coupled metabotropic glutamate 2 (mGlu2) receptor-GPCRs that are involved in signaling alterations associated with psychosis-assemble into a heteromeric complex in the mammalian brain. In single-cell experiments with various mutant versions of the mGlu2 receptor, we showed that stimulation of cells expressing mGlu2-5-HT2A heteromers with an mGlu2 agonist led to activation of Gq/11 proteins by the 5-HT2A receptors. For this crosstalk to occur, one of the mGlu2 subunits had to couple to Gi/o proteins, and we determined the relative location of the Gi/o-contacting subunit within the mGlu2 homodimer of the heteromeric complex. Additionally, mGlu2-dependent activation of Gq/11, but not Gi/o, was reduced in the frontal cortex of 5-HT2A knockout mice and was reduced in the frontal cortex of postmortem brains from schizophrenic patients. These findings offer structural insights into this important target in molecular psychiatry.


Asunto(s)
Multimerización de Proteína , Receptor de Serotonina 5-HT2A/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Esquizofrenia/metabolismo , Transducción de Señal , Regulación Alostérica , Animales , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Receptor de Serotonina 5-HT2A/genética , Receptores de Glutamato Metabotrópico/genética , Esquizofrenia/genética
20.
Pain ; 156(5): 849-858, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25734988

RESUMEN

N-methyl-D-aspartate receptor (NMDAR) antagonists have been shown to reduce mechanical hypersensitivity in animal models of inflammatory pain. However, their clinical use is associated with significant dose-limiting side effects. Small-conductance Ca-activated K channels (SK) have been shown to modulate NMDAR activity in the brain. We demonstrate that in vivo activation of SK channels in the spinal cord can alleviate mechanical hypersensitivity in a rat model of inflammatory pain. Intrathecal (i.t.) administration of the SK channel activator, 6,7-dichloro-1H-indole-2,3-dione 3-oxime (NS309), attenuates complete Freund adjuvant (CFA)-induced mechanical hypersensitivity in a dose-dependent manner. Postsynaptic expression of the SK channel subunit, SK3, and apamin-sensitive SK channel-mediated currents recorded from superficial laminae are significantly reduced in the dorsal horn (DH) after CFA. Complete Freund adjuvant-induced decrease in SK-mediated currents can be reversed in vitro by bath application of NS309. In addition, immunostaining for the SK3 subunit indicates that SK3-containing channels within DH neurons can have both somatic and dendritic localization. Double immunostaining shows coexpression of SK3 and NMDAR subunit, NR1, compatible with functional interaction. Moreover, we demonstrate that i.t. coadministration of NS309 with an NMDAR antagonist reduces the dose of NMDAR antagonist, DL-2-amino-5-phosphonopentanoic acid (DL-AP5), required to produce antinociceptive effects in the CFA model. This reduction could attenuate the unwanted side effects associated with NMDAR antagonists, giving this combination potential clinical implications.


Asunto(s)
Indoles/farmacología , Inflamación/inducido químicamente , Oximas/farmacología , Dolor/tratamiento farmacológico , Células del Asta Posterior/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Animales , Modelos Animales de Enfermedad , Adyuvante de Freund/toxicidad , Indoles/administración & dosificación , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/fisiopatología , Inyecciones Espinales , Masculino , Oximas/administración & dosificación , Dolor/inducido químicamente , Dolor/metabolismo , Dolor/fisiopatología , Umbral del Dolor , Células del Asta Posterior/metabolismo , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA