Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Bioconjug Chem ; 35(2): 164-173, 2024 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-38113481

RESUMEN

Opioid use disorder (OUD) has become a public health crisis, with recent significant increases in the number of deaths due to overdose. Vaccination can provide an attractive complementary strategy to combat OUD. A key for high vaccine efficacy is the induction of high levels of antibodies specific to the drug of abuse. Herein, a powerful immunogenic carrier, virus-like particle mutant bacteriophage Qß (mQß), has been investigated as a carrier of a small molecule hapten 6-AmHap mimicking heroin. The mQß-6-AmHap conjugate was able to induce significantly higher levels of IgG antibodies against 6-AmHap than mice immunized with the corresponding tetanus toxoid-6-AmHap conjugate in head-to-head comparison studies in multiple strains of mice. The IgG antibody responses were persistent with high anti-6-AmHap titers 600 days after being immunized with mQß-6-AmHap. The antibodies induced exhibited strong binding toward multiple heroin/morphine derivatives that have the potential to be abused, while binding weakly to medications used for OUD treatment and pain relief. Furthermore, vaccination effectively reduced the impacts of morphine on mice in both ambulation and antinociception assays, highlighting the translational potential of the mQß-6-AmHap conjugate to mitigate the harmful effects of drugs of abuse.


Asunto(s)
Analgésicos Opioides , Heroína , Ratones , Animales , Analgésicos Opioides/farmacología , Heroína/química , Heroína/farmacología , Morfina , Derivados de la Morfina , Inmunoglobulina G
2.
Nature ; 516(7529): 51-5, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25383518

RESUMEN

ß-catenin is a multi-functional protein that has an important role in the mature central nervous system; its dysfunction has been implicated in several neuropsychiatric disorders, including depression. Here we show that in mice ß-catenin mediates pro-resilient and anxiolytic effects in the nucleus accumbens, a key brain reward region, an effect mediated by D2-type medium spiny neurons. Using genome-wide ß-catenin enrichment mapping, we identify Dicer1-important in small RNA (for example, microRNA) biogenesis--as a ß-catenin target gene that mediates resilience. Small RNA profiling after excising ß-catenin from nucleus accumbens in the context of chronic stress reveals ß-catenin-dependent microRNA regulation associated with resilience. Together, these findings establish ß-catenin as a critical regulator in the development of behavioural resilience, activating a network that includes Dicer1 and downstream microRNAs. We thus present a foundation for the development of novel therapeutic targets to promote stress resilience.


Asunto(s)
ARN Helicasas DEAD-box/genética , Regulación de la Expresión Génica , MicroARNs/genética , Resiliencia Psicológica , Ribonucleasa III/genética , Estrés Fisiológico/genética , beta Catenina/metabolismo , Adaptación Fisiológica/genética , Animales , ARN Helicasas DEAD-box/metabolismo , Depresión/fisiopatología , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Neuronas/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal , beta Catenina/genética
3.
Nature ; 493(7433): 532-6, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23235832

RESUMEN

Ventral tegmental area (VTA) dopamine neurons in the brain's reward circuit have a crucial role in mediating stress responses, including determining susceptibility versus resilience to social-stress-induced behavioural abnormalities. VTA dopamine neurons show two in vivo patterns of firing: low frequency tonic firing and high frequency phasic firing. Phasic firing of the neurons, which is well known to encode reward signals, is upregulated by repeated social-defeat stress, a highly validated mouse model of depression. Surprisingly, this pathophysiological effect is seen in susceptible mice only, with no apparent change in firing rate in resilient individuals. However, direct evidence--in real time--linking dopamine neuron phasic firing in promoting the susceptible (depression-like) phenotype is lacking. Here we took advantage of the temporal precision and cell-type and projection-pathway specificity of optogenetics to show that enhanced phasic firing of these neurons mediates susceptibility to social-defeat stress in freely behaving mice. We show that optogenetic induction of phasic, but not tonic, firing in VTA dopamine neurons of mice undergoing a subthreshold social-defeat paradigm rapidly induced a susceptible phenotype as measured by social avoidance and decreased sucrose preference. Optogenetic phasic stimulation of these neurons also quickly induced a susceptible phenotype in previously resilient mice that had been subjected to repeated social-defeat stress. Furthermore, we show differences in projection-pathway specificity in promoting stress susceptibility: phasic activation of VTA neurons projecting to the nucleus accumbens (NAc), but not to the medial prefrontal cortex (mPFC), induced susceptibility to social-defeat stress. Conversely, optogenetic inhibition of the VTA-NAc projection induced resilience, whereas inhibition of the VTA-mPFC projection promoted susceptibility. Overall, these studies reveal novel firing-pattern- and neural-circuit-specific mechanisms of depression.


Asunto(s)
Depresión/fisiopatología , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/citología , Conducta Social , Estrés Psicológico/fisiopatología , Animales , Depresión/etiología , Preferencias Alimentarias , Masculino , Ratones , Vías Nerviosas , Núcleo Accumbens/fisiología , Optogenética , Fenotipo , Corteza Prefrontal/fisiología , Estrés Psicológico/complicaciones , Sacarosa/administración & dosificación , Factores de Tiempo , Área Tegmental Ventral/fisiología
4.
J Neurosci ; 35(40): 13773-83, 2015 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-26446228

RESUMEN

The hippocampus (HPC) is known to play an important role in learning, a process dependent on synaptic plasticity; however, the molecular mechanisms underlying this are poorly understood. ΔFosB is a transcription factor that is induced throughout the brain by chronic exposure to drugs, stress, and variety of other stimuli and regulates synaptic plasticity and behavior in other brain regions, including the nucleus accumbens. We show here that ΔFosB is also induced in HPC CA1 and DG subfields by spatial learning and novel environmental exposure. The goal of the current study was to examine the role of ΔFosB in hippocampal-dependent learning and memory and the structural plasticity of HPC synapses. Using viral-mediated gene transfer to silence ΔFosB transcriptional activity by expressing ΔJunD (a negative modulator of ΔFosB transcriptional function) or to overexpress ΔFosB, we demonstrate that HPC ΔFosB regulates learning and memory. Specifically, ΔJunD expression in HPC impaired learning and memory on a battery of hippocampal-dependent tasks in mice. Similarly, general ΔFosB overexpression also impaired learning. ΔJunD expression in HPC did not affect anxiety or natural reward, but ΔFosB overexpression induced anxiogenic behaviors, suggesting that ΔFosB may mediate attentional gating in addition to learning. Finally, we found that overexpression of ΔFosB increases immature dendritic spines on CA1 pyramidal cells, whereas ΔJunD reduced the number of immature and mature spine types, indicating that ΔFosB may exert its behavioral effects through modulation of HPC synaptic function. Together, these results suggest collectively that ΔFosB plays a significant role in HPC cellular morphology and HPC-dependent learning and memory. SIGNIFICANCE STATEMENT: Consolidation of our explicit memories occurs within the hippocampus, and it is in this brain region that the molecular and cellular processes of learning have been most closely studied. We know that connections between hippocampal neurons are formed, eliminated, enhanced, and weakened during learning, and we know that some stages of this process involve alterations in the transcription of specific genes. However, the specific transcription factors involved in this process are not fully understood. Here, we demonstrate that the transcription factor ΔFosB is induced in the hippocampus by learning, regulates the shape of hippocampal synapses, and is required for memory formation, opening up a host of new possibilities for hippocampal transcriptional regulation.


Asunto(s)
Hipocampo/metabolismo , Aprendizaje/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Reacción de Prevención , Condicionamiento Psicológico/fisiología , Espinas Dendríticas/metabolismo , Dependovirus/genética , Ambiente , Conducta Exploratoria/fisiología , Miedo/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Conducta Espacial
5.
J Neurosci ; 34(11): 3878-87, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24623766

RESUMEN

Decreased medial prefrontal cortex (mPFC) neuronal activity is associated with social defeat-induced depression- and anxiety-like behaviors in mice. However, the molecular mechanisms underlying the decreased mPFC activity and its prodepressant role remain unknown. We show here that induction of the transcription factor ΔFosB in mPFC, specifically in the prelimbic (PrL) area, mediates susceptibility to stress. ΔFosB induction in PrL occurred selectively in susceptible mice after chronic social defeat stress, and overexpression of ΔFosB in this region, but not in the nearby infralimbic (IL) area, enhanced stress susceptibility. ΔFosB produced these effects partly through induction of the cholecystokinin (CCK)-B receptor: CCKB blockade in mPFC induces a resilient phenotype, whereas CCK administration into mPFC mimics the anxiogenic- and depressant-like effects of social stress. We previously found that optogenetic stimulation of mPFC neurons in susceptible mice reverses several behavioral abnormalities seen after chronic social defeat stress. Therefore, we hypothesized that optogenetic stimulation of cortical projections would rescue the pathological effects of CCK in mPFC. After CCK infusion in mPFC, we optogenetically stimulated mPFC projections to basolateral amygdala or nucleus accumbens, two subcortical structures involved in mood regulation. Stimulation of corticoamygdala projections blocked the anxiogenic effect of CCK, although no effect was observed on other symptoms of social defeat. Conversely, stimulation of corticoaccumbens projections reversed CCK-induced social avoidance and sucrose preference deficits but not anxiogenic-like effects. Together, these results indicate that social stress-induced behavioral deficits are mediated partly by molecular adaptations in mPFC involving ΔFosB and CCK through cortical projections to distinct subcortical targets.


Asunto(s)
Trastornos de Ansiedad/fisiopatología , Colecistoquinina/fisiología , Trastorno Depresivo/fisiopatología , Corteza Prefrontal/fisiología , Proteínas Proto-Oncogénicas c-fos/fisiología , Receptor de Colecistoquinina B/fisiología , Animales , Ansiolíticos/farmacología , Trastornos de Ansiedad/patología , Mapeo Encefálico , Enfermedad Crónica , Trastorno Depresivo/patología , Indoles/farmacología , Sistema Límbico/citología , Sistema Límbico/efectos de los fármacos , Sistema Límbico/fisiología , Masculino , Meglumina/análogos & derivados , Meglumina/farmacología , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/fisiología , Corteza Prefrontal/citología , Corteza Prefrontal/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/genética , Receptor de Colecistoquinina B/antagonistas & inhibidores , Receptor de Colecistoquinina B/genética , Predominio Social , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología
6.
J Neurochem ; 132(2): 243-53, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25099208

RESUMEN

Drugs of abuse modulate the function and activity of the mesolimbic dopamine circuit. To identify novel mediators of drug-induced neuroadaptations in the ventral tegmental area (VTA), we performed RNA sequencing analysis on VTA samples from mice administered repeated saline, morphine, or cocaine injections. One gene that was similarly up-regulated by both drugs was serum- and glucocorticoid-inducible kinase 1 (SGK1). SGK1 activity, as measured by phosphorylation of its substrate N-myc downstream regulated gene (NDRG), was also increased robustly by chronic drug treatment. Increased NDRG phosphorylation was evident 1 but not 24 h after the last drug injection. SGK1 phosphorylation itself was similarly modulated. To determine the role of increased SGK1 activity on drug-related behaviors, we over-expressed constitutively active (CA) SGK1 in the VTA. SGK1-CA expression reduced locomotor sensitization elicited by repeated cocaine, but surprisingly had the opposite effect and promoted locomotor sensitization to morphine, without affecting the initial locomotor responses to either drug. SGK1-CA expression did not significantly affect morphine or cocaine conditioned place preference, although there was a trend toward increased conditioned place preference with both drugs. Further characterizing the role of this kinase in drug-induced changes in VTA may lead to improved understanding of neuroadaptations critical to drug dependence and addiction. We find that repeated, but not acute, morphine or cocaine administration induces an increase in serum- and glucocorticoid-inducible kinase (SGK1) gene expression and activity in the ventral tegmental area (VTA). This increase in SGK1 activity may play a role in drug-dependent behaviors and suggests a novel signaling cascade for potential intervention in drug dependence and addiction.


Asunto(s)
Cocaína/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Inmediatas-Precoces/biosíntesis , Morfina/farmacología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Área Tegmental Ventral/efectos de los fármacos , Animales , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Clásico/fisiología , Inducción Enzimática/efectos de los fármacos , Genes Reporteros , Vectores Genéticos , Proteínas Inmediatas-Precoces/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Proteínas del Tejido Nervioso/genética , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos , Área Tegmental Ventral/enzimología
7.
J Neurosci ; 33(10): 4295-307, 2013 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-23467346

RESUMEN

The transcription factor ΔFosB and the brain-enriched calcium/calmodulin-dependent protein kinase II (CaMKIIα) are induced in the nucleus accumbens (NAc) by chronic exposure to cocaine or other psychostimulant drugs of abuse, in which the two proteins mediate sensitized drug responses. Although ΔFosB and CaMKIIα both regulate AMPA glutamate receptor expression and function in NAc, dendritic spine formation on NAc medium spiny neurons (MSNs), and locomotor sensitization to cocaine, no direct link between these molecules has to date been explored. Here, we demonstrate that ΔFosB is phosphorylated by CaMKIIα at the protein-stabilizing Ser27 and that CaMKII is required for the cocaine-mediated accumulation of ΔFosB in rat NAc. Conversely, we show that ΔFosB is both necessary and sufficient for cocaine induction of CaMKIIα gene expression in vivo, an effect selective for D1-type MSNs in the NAc shell subregion. Furthermore, induction of dendritic spines on NAc MSNs and increased behavioral responsiveness to cocaine after NAc overexpression of ΔFosB are both CaMKII dependent. Importantly, we demonstrate for the first time induction of ΔFosB and CaMKII in the NAc of human cocaine addicts, suggesting possible targets for future therapeutic intervention. These data establish that ΔFosB and CaMKII engage in a cell-type- and brain-region-specific positive feedforward loop as a key mechanism for regulating the reward circuitry of the brain in response to chronic cocaine.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Trastornos Relacionados con Cocaína/patología , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Núcleo Accumbens/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Adolescente , Adulto , Anciano , Análisis de Varianza , Animales , Benzazepinas/farmacología , Calcio/metabolismo , Inmunoprecipitación de Cromatina , Trastornos Relacionados con Cocaína/metabolismo , Estudios de Cohortes , Antagonistas de Dopamina/farmacología , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/fisiología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Núcleo Accumbens/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Salicilamidas/farmacología , Serina/metabolismo , Adulto Joven
8.
J Biol Chem ; 288(1): 20-32, 2013 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-23161550

RESUMEN

As an approach to elucidating dopamine transporter (DAT) phosphorylation characteristics, we examined in vitro phosphorylation of a recombinant rat DAT N-terminal peptide (NDAT) using purified protein kinases. We found that NDAT becomes phosphorylated at single distinct sites by protein kinase A (Ser-7) and calcium-calmodulin-dependent protein kinase II (Ser-13) and at multiple sites (Ser-4, Ser-7, and Ser-13) by protein kinase C (PKC), implicating these residues as potential sites of DAT phosphorylation by these kinases. Mapping of rat striatal DAT phosphopeptides by two-dimensional thin layer chromatography revealed basal and PKC-stimulated phosphorylation of the same peptide fragments and comigration of PKC-stimulated phosphopeptide fragments with NDAT Ser-7 phosphopeptide markers. We further confirmed by site-directed mutagenesis and mass spectrometry that Ser-7 is a site for PKC-stimulated phosphorylation in heterologously expressed rat and human DATs. Mutation of Ser-7 and nearby residues strongly reduced the affinity of rat DAT for the cocaine analog (-)-2ß-carbomethoxy-3ß-(4-fluorophenyl) tropane (CFT), whereas in rat striatal tissue, conditions that promote DAT phosphorylation caused increased CFT affinity. Ser-7 mutation also affected zinc modulation of CFT binding, with Ala and Asp substitutions inducing opposing effects. These results identify Ser-7 as a major site for basal and PKC-stimulated phosphorylation of native and expressed DAT and suggest that Ser-7 phosphorylation modulates transporter conformational equilibria, shifting the transporter between high and low affinity cocaine binding states.


Asunto(s)
Cocaína/química , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/química , Serina/química , Animales , Sitios de Unión , Cromatografía en Capa Delgada/métodos , Cocaína/análogos & derivados , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Inhibidores de Captación de Dopamina/química , Células HEK293 , Humanos , Cinética , Masculino , Espectrometría de Masas/métodos , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Unión Proteica , Conformación Proteica , Ratas , Ratas Sprague-Dawley
9.
J Neurosci ; 32(22): 7577-84, 2012 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-22649236

RESUMEN

The molecular mechanism underlying induction by cocaine of ΔFosB, a transcription factor important for addiction, remains unknown. Here, we demonstrate a necessary role for two transcription factors, cAMP response element binding protein (CREB) and serum response factor (SRF), in mediating this induction within the mouse nucleus accumbens (NAc), a key brain reward region. CREB and SRF are both activated in NAc by cocaine and bind to the fosB gene promoter. Using viral-mediated Cre recombinase expression in the NAc of single- or double-floxed mice, we show that deletion of both transcription factors from this brain region completely blocks cocaine induction of ΔFosB in NAc, whereas deletion of either factor alone has no effect. Furthermore, deletion of both SRF and CREB from NAc renders animals less sensitive to the rewarding effects of moderate doses of cocaine when tested in the conditioned place preference (CPP) procedure and also blocks locomotor sensitization to higher doses of cocaine. Deletion of CREB alone has the opposite effect and enhances both cocaine CPP and locomotor sensitization. In contrast to ΔFosB induction by cocaine, ΔFosB induction in NAc by chronic social stress, which we have shown previously requires activation of SRF, is unaffected by the deletion of CREB alone. These surprising findings demonstrate the involvement of distinct transcriptional mechanisms in mediating ΔFosB induction within this same brain region by cocaine versus stress. Our results also establish a complex mode of regulation of ΔFosB induction in response to cocaine, which requires the concerted activities of both SRF and CREB.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Cocaína/farmacología , Inhibidores de Captación de Dopamina/farmacología , Núcleo Accumbens/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factor de Respuesta Sérica/metabolismo , Análisis de Varianza , Animales , Proteína de Unión a CREB/deficiencia , Inmunoprecipitación de Cromatina , Condicionamiento Operante/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Núcleo Accumbens/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-fos/genética , ARN Mensajero/metabolismo , Factor de Respuesta Sérica/deficiencia , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología , Transducción Genética
10.
J Neurosci ; 32(16): 5385-97, 2012 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-22514303

RESUMEN

Attention deficit/hyperactivity disorder (ADHD) is the most commonly diagnosed disorder of school-age children. Although genetic and brain-imaging studies suggest a contribution of altered dopamine (DA) signaling in ADHD, evidence of signaling perturbations contributing to risk is largely circumstantial. The presynaptic, cocaine- and amphetamine (AMPH)-sensitive DA transporter (DAT) constrains DA availability at presynaptic and postsynaptic receptors following vesicular release and is targeted by the most commonly prescribed ADHD therapeutics. Using polymorphism discovery approaches with an ADHD cohort, we identified a hDAT (human DAT) coding variant, R615C, located in the distal C terminus of the transporter, a region previously implicated in constitutive and regulated transporter trafficking. Here, we demonstrate that, whereas wild-type DAT proteins traffic in a highly regulated manner, DAT 615C proteins recycle constitutively and demonstrate insensitivity to the endocytic effects of AMPH and PKC (protein kinase C) activation. The disrupted regulation of DAT 615C parallels a redistribution of the transporter variant away from GM1 ganglioside- and flotillin1-enriched membranes, and is accompanied by altered CaMKII (calcium/calmodulin-dependent protein kinase II) and flotillin-1 interactions. Using C-terminal peptides derived from wild-type DAT and the R615C variant, we establish that the DAT 615C C terminus can act dominantly to preclude AMPH regulation of wild-type DAT. Mutagenesis of DAT C-terminal sequences suggests that phosphorylation of T613 may be important in sorting DAT between constitutive and regulated pathways. Together, our studies support a coupling of DAT microdomain localization with transporter regulation and provide evidence of perturbed DAT activity and DA signaling as a risk determinant for ADHD.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Microdominios de Membrana/genética , Polimorfismo de Nucleótido Simple/genética , Adolescente , Anfetamina/farmacología , Análisis de Varianza , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Bencilaminas/farmacología , Biotinilación , Calcio/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular Transformada , Niño , Preescolar , Toxina del Cólera/metabolismo , Estudios de Cohortes , Dopamina/metabolismo , Dopamina/farmacología , Inhibidores de Captación de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Electroquímica , Femenino , Humanos , Inmunoprecipitación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Microdominios de Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Sulfonamidas/farmacología , Transfección/métodos , Tritio/metabolismo
11.
Proc Natl Acad Sci U S A ; 107(3): 1217-22, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20080553

RESUMEN

The prairie vole (Microtus ochrogaster) is a socially monogamous rodent species that forms pair bonds after mating, a behavior in which central dopamine (DA) has been implicated. Here, we used male prairie voles to examine the effects of drug exposure on pair bonding and related neural circuitry. In our first experiment, amphetamine (AMPH) motivated behavior was examined using a conditioned place preference (CPP) paradigm and was shown to be mediated by activation of D1-like DA receptors. Next, we examined the effects of repeated AMPH exposure on pair bonding. Intact and saline pretreated control males displayed mating-induced partner preferences, whereas males pretreated with AMPH at the doses effective to induce CPP failed to show mating-induced partner preferences. Such AMPH treatment also enhanced D1, but not D2, DA receptor expression in the nucleus accumbens (NAcc). Furthermore, pharmacological blockade of D1-like DA receptors in the NAcc rescued mating-induced partner preferences in AMPH-treated males. Together, our data indicate that repeated AMPH exposure may narrow the behavioral repertoire of male prairie voles via a DA receptor-specific mechanism in the NAcc, resulting in the impairment of pair bond formation.


Asunto(s)
Anfetamina/farmacología , Arvicolinae/fisiología , Dopamina/metabolismo , Núcleo Accumbens/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Conducta Social , Animales , Western Blotting , Femenino , Masculino
12.
J Neurosci ; 31(25): 9084-92, 2011 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-21697359

RESUMEN

Based on earlier gene expression and chromatin array data, we identified the protein, dishevelled (DVL)-2, as being regulated in the nucleus accumbens (NAc), a key brain reward region, in the mouse social defeat model of depression. Here, we validate these findings by showing that DVL2 mRNA and protein levels are downregulated in NAc of mice susceptible to social defeat stress, effects not seen in resilient mice. Other DVL isoforms, DVL1 and DVL3, show similar patterns of regulation. Downregulation of DVL was also demonstrated in the NAc of depressed humans examined postmortem. Interestingly, several members of the WNT (Wingless)-DVL signaling cascade, including phospho-GSK3ß (glycogen synthase kinase-3ß), also show significant downregulation in the NAc of susceptible, but not resilient, mice, demonstrating concerted regulation of this pathway in the NAc due to social defeat stress. By using viral-mediated gene transfer to overexpress a dominant-negative mutant of DVL in NAc, or by using a pharmacological inhibitor of DVL administered into this brain region, we show that blockade of DVL function renders mice more susceptible to social defeat stress and promotes depression-like behavior in other assays. Similar prodepression-like effects were induced upon overexpressing GSK3ß in the NAc, while overexpressing a dominant-negative mutant of GSK3ß promoted resilience to social defeat stress. These findings are consistent with the knowledge that downregulation of DVL and phospho-GSK3ß reflects an increase in GSK3ß activity. These studies reveal a novel role for the DVL-GSK3ß signaling pathway, acting within the brain's reward circuitry, in regulating susceptibility to chronic stress.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Depresión/metabolismo , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Núcleo Accumbens/metabolismo , Fosfoproteínas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Depresión/etiología , Proteínas Dishevelled , Dominación-Subordinación , Glucógeno Sintasa Quinasa 3 beta , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo
13.
Artículo en Inglés | MEDLINE | ID: mdl-34404738

RESUMEN

Depression and related mood disorders constitute an enormous burden on health, quality of life, and the global economy, and women have roughly twice the lifetime risk of men for experiencing depression. Here, we review sex differences in human brain physiology that may be connected to the increased susceptibility of women to major depressive disorder (MDD). Moreover, we summarize decades of preclinical research using animal models for the study of mood dysfunction that uncover some of the potential molecular, cellular, and circuit-level mechanisms that may underlie sex differences and disease etiology. We place particular emphasis on a series of recent studies demonstrating the central contribution of the circuit projecting from ventral hippocampus to nucleus accumbens and how inherent sex differences in the excitability of this circuit may predict and drive depression-related behaviors. The findings covered in this review underscore the continued need for studies using preclinical models and circuit-specific strategies for uncovering molecular and physiological mechanisms that could lead to potential sex-specific diagnosis, prognosis, prevention, and/or treatments for MDD and other mood disorders.


Asunto(s)
Trastorno Depresivo Mayor , Animales , Depresión , Femenino , Humanos , Masculino , Modelos Animales , Calidad de Vida , Caracteres Sexuales
14.
Mucosal Immunol ; 15(5): 964-976, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35869148

RESUMEN

Intestinal epithelial barrier function is compromised in inflammatory bowel disease and barrier dysfunction contributes to disease progression. Extracellular nucleotides/nucleosides generated in gut inflammation may regulate barrier function through actions on diverse cell types. Enteric glia modulate extracellular purinergic signaling and exert pathophysiological effects on mucosal permeability. These glia may regulate inflammation with paracrine responses, theoretically mediated via adenosine 2B receptor (A2BR) signaling. As the cell-specific roles of A2BRs in models of colitis and barrier dysfunction are unclear, we studied glial A2BRs in acute dextran sodium sulfate (DSS) colitis. We performed and validated conditional ablation of glial A2BRs in Sox10CreERT2+/-;Adora2bf/f mice. Overt intestinal disease activity indices in DSS-colitis were comparable between Sox10CreERT2+/-;Adora2bf/f mice and littermate controls. However, ablating glial A2BRs protected against barrier dysfunction following acute DSS-colitis. These benefits were associated with the normalization of tight junction protein expression and localization including claudin-1, claudin-8, and occludin. Glial A2BR signaling increased levels of proinflammatory mediators in the colon and cell-intrinsic regulation of genes including Csf3, Cxcl1, Cxcl10, and Il6. Our studies show that glial A2BR signaling exacerbates immune responses during DSS-colitis and that this adenosinergic cell-specific mechanism contributes to persistent gut epithelial barrier dysfunction.


Asunto(s)
Colitis , Mucosa Intestinal , Adenosina/metabolismo , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colon/metabolismo , Sulfato de Dextran , Modelos Animales de Enfermedad , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Neuroglía/metabolismo
15.
Physiol Behav ; 247: 113707, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35063424

RESUMEN

The lateral hypothalamic area (LHA) is essential for ingestive behavior but has primarily been studied in modulating feeding, with comparatively scant attention on drinking. This is partly because most LHA neurons simultaneously promote feeding and drinking, suggesting that ingestive behaviors track together. A notable exception are LHA neurons expressing neurotensin (LHANts neurons): activating these neurons promotes water intake but modestly restrains feeding. Here we investigated the connectivity of LHANts neurons, their necessity and sufficiency for drinking and feeding, and how timing and resource availability influence their modulation of these behaviors. LHANts neurons project broadly throughout the brain, including to the lateral preoptic area (LPO), a brain region implicated in modulating drinking behavior. LHANts neurons also receive inputs from brain regions implicated in sensing hydration and energy status. While activation of LHANts neurons is not required to maintain homeostatic water or food intake, it selectively promotes drinking during the light cycle, when ingestive drive is low. Activating LHANts neurons during this period also increases willingness to work for water or palatable fluids, regardless of their caloric content. By contrast, LHANts neuronal activation during the dark cycle does not promote drinking, but suppresses feeding during this time. Finally, we demonstrate that the activation of the LHANts â†’ LPO projection is sufficient to mediate drinking behavior, but does not suppress feeding as observed after generally activating all LHANts neurons. Overall, our work suggests how and when LHANts neurons oppositely modulate ingestive behaviors.


Asunto(s)
Área Hipotalámica Lateral , Neurotensina , Alimentos , Área Hipotalámica Lateral/metabolismo , Neuronas/metabolismo , Neurotensina/metabolismo , Agua
16.
Artículo en Inglés | MEDLINE | ID: mdl-31964652

RESUMEN

Opioid drugs are highly valued as potent analgesics; however, there are significant risks associated with long-term use because of their abuse liability. Opioids cause changes in ventral tegmental area (VTA) gene expression and cell activity that have been linked to addiction-related behaviors in rodent models. Here, we focus on VTA dopamine (DA) neurons and review the cellular, structural, and synaptic plasticity changes induced by acute and chronic opioid exposure. We also discuss many avenues for future research including determination of whether opioid neuroadaptations are specific for subpopulations of VTA DA neurons. A better understanding of the molecular adaptations within the cells and circuits that drive opioid abuse is crucial for the development of better treatments for substance use disorders and to create novel, safer pain-relieving therapeutics.


Asunto(s)
Analgésicos Opioides/farmacología , Plasticidad Neuronal/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Neuronas Dopaminérgicas/efectos de los fármacos , Humanos
17.
Neuropsychopharmacology ; 46(9): 1574-1583, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34007042

RESUMEN

Drugs of abuse regulate the activity of the mesolimbic dopamine (DA) system, and drug-induced changes in ventral tegmental area (VTA) cellular activity and gene regulation are linked to behavioral outputs associated with addiction. Previous work from our lab determined that VTA serum- and glucocorticoid-inducible kinase 1 (SGK1) transcription and catalytic activity were increased by repeated cocaine administration; however, it was unknown if these biochemical changes contributed to cocaine-elicited behaviors. Using transgenic and viral-mediated manipulations, we investigated the role of VTA SGK1 catalytic activity in regulating cocaine conditioned place preference and self-administration. We showed intra-VTA infusion of a catalytically inactive SGK1 mutant (K127Q) significantly decreased cocaine conditioned place preference (CPP). Further, we found that K127Q expression in VTA DA neurons significantly decreased cocaine CPP, while this same manipulation in VTA GABA neurons had no effect. However, blunted VTA DA SGK1 catalytic activity did not alter cocaine self-administration. Altogether, these studies identify the specific VTA cells critical for SGK1-mediated effects on cocaine CPP but not self-administration.


Asunto(s)
Cocaína , Área Tegmental Ventral , Cocaína/farmacología , Condicionamiento Clásico , Neuronas Dopaminérgicas , Glucocorticoides
18.
Neuroscience ; 461: 11-22, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33689861

RESUMEN

Despite the high prevalence of major depressive disorder (MDD), understanding of the biological underpinnings remains limited. Rodent models suggest that changes in activity and output of dopamine (DA) neurons in the ventral tegmental area (VTA) are important for depressive-like phenotypes. Additionally, brain inflammatory processes are thought to contribute to MDD pathology and inflammation in the VTA has been linked to changes in VTA DA neuronal activity. Thus, we sought to determine whether there is increased inflammatory signaling in the VTA following forms of chronic stress that induce depressive-like symptoms. First, we subjected male mice to either physical or vicarious chronic social defeat stress (CSDS), paradigms known to induce long-term depressive-like behavior and changes in VTA signaling. Second, we subjected male and female mice to subchronic variable stress (SCVS), a paradigm that induces depressive-like behavior only in female mice. We then isolated mRNA from the VTA and assessed proinflammatory gene regulation via RT-PCR. Our results show that physical, but not vicarious, CSDS increases interleukin 1ß (IL-1ß) mRNA expression and this inversely correlates with social interaction score. In contrast, IL-1ß expression was unchanged in male or female mice following SCVS. No significant increases in VTA ionized calcium binding adapter molecule 1 (Iba1) and glial fibrillary acidic protein (GFAP) immunochemistry were detected following CSDS that would be indicative of a robust inflammatory response. In conclusion, we show that chronic stressors distinctively alter expression of proinflammatory genes in the VTA and changes may depend on the severity and time-course of the stress exposure.


Asunto(s)
Trastorno Depresivo Mayor , Área Tegmental Ventral , Animales , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Femenino , Masculino , Ratones , Estrés Psicológico
19.
J Neurosci ; 29(11): 3529-37, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19295158

RESUMEN

Although chronic cocaine-induced changes in dendritic spines on nucleus accumbens (NAc) neurons have been correlated with behavioral sensitization, the molecular pathways governing these structural changes, and their resulting behavioral effects, are poorly understood. The transcription factor, nuclear factor kappa B (NFkappaB), is rapidly activated by diverse stimuli and regulates expression of many genes known to maintain cell structure. Therefore, we evaluated the role of NFkappaB in regulating cocaine-induced dendritic spine changes on medium spiny neurons of the NAc and the rewarding effects of cocaine. We show that chronic cocaine induces NFkappaB-dependent transcription in the NAc of NFkappaB-Lac transgenic mice. This induction of NFkappaB activity is accompanied by increased expression of several NFkappaB genes, the promoters of which show chromatin modifications after chronic cocaine exposure consistent with their transcriptional activation. To study the functional significance of this induction, we used viral-mediated gene transfer to express either a constitutively active or dominant-negative mutant of Inhibitor of kappa B kinase (IKKca or IKKdn), which normally activates NFkappaB signaling, in the NAc. We found that activation of NFkappaB by IKKca increases the number of dendritic spines on NAc neurons, whereas inhibition of NFkappaB by IKKdn decreases basal dendritic spine number and blocks the increase in dendritic spines after chronic cocaine. Moreover, inhibition of NFkappaB blocks the rewarding effects of cocaine and the ability of previous cocaine exposure to increase an animal's preference for cocaine. Together, these studies establish a direct role for NFkappaB pathways in the NAc to regulate structural and behavioral plasticity to cocaine.


Asunto(s)
Cocaína/administración & dosificación , FN-kappa B/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Recompensa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Animales , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/ultraestructura , Núcleo Accumbens/citología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/fisiología , Células PC12 , Ratas
20.
Neurosci Lett ; 725: 134909, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32169587

RESUMEN

The hippocampus has a well-known role in mediating learning and memory, and its function can be directly regulated by both stress and glucocorticoid receptor activation. Hippocampal contributions to learning are thought to be dependent on changes in the plasticity of synapses within specific subregions, and these functional changes are accompanied by morphological changes in the number and shape of dendritic spines, the physical correlates of these glutamatergic synapses. Serum- and glucocorticoid-inducible kinase 1 (SGK1) regulates dendritic spine morphology in the prefrontal cortex, and modulation of SGK1 expression in mouse hippocampus regulates learning. However, the role of SGK1 in dendritic spine morphology within the CA1 and dentate gyrus regions of the hippocampus are unknown. Thus, herpes simplex viral vectors expressing GFP and various SGK1 constructs, including wild type SGK1, a catalytically inactive version of SGK1 (K127Q), and a phospho-defective version of SGK1 (S78A), were infused into the hippocampus of adult mice and confocal fluorescent microscopy was used to visualize dendritic spines. We show that increasing expression of SGK1 in the dentate gyrus increased the total number of spines, driven primarily by an increase in mushroom spines, while decreasing SGK1 activity (K127Q) in the CA1 region increased the total number of dendritic spines, driven by a significant increase in mushroom and stubby spines. The differential effects of SGK1 in these regions may be mediated by the interactions of SGK1 with multiple pathways required for spine formation and stability. As the formation of mature synapses is a crucial component of learning and memory, this indicates that SGK1 is a potential target in the pathway underlying stress-associated changes in cognition and memory.


Asunto(s)
Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Proteínas Inmediatas-Precoces/biosíntesis , Proteínas Serina-Treonina Quinasas/biosíntesis , Animales , Espinas Dendríticas/química , Activación Enzimática/fisiología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/análisis , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/análisis , Hipocampo/química , Proteínas Inmediatas-Precoces/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA