Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neuropsychopharmacology ; 46(12): 2148-2157, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34035469

RESUMEN

Clinical and translational studies suggest that prefrontal cortex (PFC) dysregulation is a hallmark feature of several affective disorders. Thus, investigating the mechanisms involved in the regulation of PFC function and synaptic plasticity could aid in developing new medications. In recent years, the mGlu2 and mGlu3 subtypes of metabotropic glutamate (mGlu) receptors have emerged as exciting potential targets for the treatment of affective disorders, as mGlu2/3 antagonists exert antidepressant-like effects across many rodent models. Several recent studies suggest that presynaptic mGlu2 receptors may contribute to these effects by regulating excitatory transmission at synapses from the thalamus to the PFC. Interestingly, we found that mGlu3 receptors also inhibit excitatory drive to the PFC but act by inducing long-term depression (LTD) at amygdala-PFC synapses. It remains unclear, however, whether blockade of presynaptic, postsynaptic, or glial mGlu3 receptors contribute to long-term effects on PFC circuit function and antidepressant-like effects of mGlu2/3 antagonists. To address these outstanding questions, we leveraged transgenic Grm3fl/fl mice and viral-mediated gene transfer to genetically ablate mGlu3 receptors from pyramidal cells in the frontal cortex of adult mice of all sexes. Consistent with a role for mGlu3 in PFC pyramidal cells, mGlu3-dependent amygdala-cortical LTD was eliminated following mGlu3 receptor knockdown. Furthermore, knockdown mice displayed a modest, task-specific anxiolytic phenotype and decreased passive coping behaviors. These studies reveal that postsynaptic mGlu3 receptors are critical for mGlu3-dependent LTD and provide convergent genetic evidence suggesting that modulating cortical mGlu3 receptors may provide a promising new approach for the treatment of mood disorders.


Asunto(s)
Ácido Glutámico , Receptores de Glutamato Metabotrópico/genética , Animales , Ratones , Plasticidad Neuronal , Corteza Prefrontal/metabolismo , Células Piramidales/metabolismo
2.
Neuron ; 105(1): 46-59.e3, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31735403

RESUMEN

Non-selective antagonists of metabotropic glutamate receptor subtypes 2 (mGlu2) and 3 (mGlu3) exert rapid antidepressant-like effects by enhancing prefrontal cortex (PFC) glutamate transmission; however, the receptor subtype contributions and underlying mechanisms remain unclear. Here, we leveraged newly developed negative allosteric modulators (NAMs), transgenic mice, and viral-assisted optogenetics to test the hypothesis that selective inhibition of mGlu2 or mGlu3 potentiates PFC excitatory transmission and confers antidepressant efficacy in preclinical models. We found that systemic treatment with an mGlu2 or mGlu3 NAM rapidly activated biophysically unique PFC pyramidal cell ensembles. Mechanistic studies revealed that mGlu2 and mGlu3 NAMs enhance thalamocortical transmission and inhibit long-term depression by mechanistically distinct presynaptic and postsynaptic actions. Consistent with these actions, systemic treatment with either NAM decreased passive coping and reversed anhedonia in two independent chronic stress models, suggesting that both mGlu2 and mGlu3 NAMs induce antidepressant-like effects through related but divergent mechanisms of action.


Asunto(s)
Regulación Alostérica/fisiología , Corteza Cerebral/fisiología , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Tálamo/fisiología , Adaptación Psicológica/efectos de los fármacos , Anhedonia/efectos de los fármacos , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Corteza Cerebral/metabolismo , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Depresión Sináptica a Largo Plazo/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Vías Nerviosas/fisiología , Plasticidad Neuronal/fisiología , Optogenética , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Células Piramidales/fisiología , Tálamo/metabolismo
3.
Neuropharmacology ; 144: 19-28, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30326237

RESUMEN

Stress can precipitate or worsen symptoms of many psychiatric illnesses. Dysregulation of the prefrontal cortex (PFC) glutamate system may underlie these disruptions and restoring PFC glutamate signaling has emerged as a promising avenue for the treatment of stress disorders. Recently, we demonstrated that activation of metabotropic glutamate receptor subtype 3 (mGlu3) induces a postsynaptic form of long-term depression (LTD) that is dependent on the activity of another subtype, mGlu5. Stress exposure disrupted this plasticity, but the underlying signaling mechanisms and involvement in higher-order cognition have not yet been investigated. Acute stress was applied by 20-min restraint and early reversal learning was evaluated in an operant-based food-seeking task. We employed whole-cell patch-clamp recordings of layer 5 prelimbic (PL)-PFC pyramidal cells to examine mGlu3-LTD and several mechanistically distinct mGlu5-dependent functions. Acute stress impaired both mGlu3-LTD and early reversal learning. Interestingly, potentiating mGlu5 signaling with the mGlu5 positive allosteric modulator (PAM) VU0409551 rescued stress-induced deficits in both mGlu3-LTD and reversal learning. Other aspects of PL-PFC mGlu5 function were not disrupted following stress; however, signaling downstream of mGlu5-Homer interactions, phosphoinositide-3-kinase (PI3K), Akt, and glycogen synthase kinase 3ß was implicated in these phenomena. These findings demonstrate that acute stress disrupts early reversal learning and PL-PFC-dependent synaptic plasticity and that potentiating mGlu5 function can restore these impairments. These findings provide a framework through which modulating coordinated mGlu3/mGlu5 signaling may confer benefits for the treatment of stress-related psychiatric disorders.


Asunto(s)
Corteza Cerebral/metabolismo , Plasticidad Neuronal/fisiología , Receptor del Glutamato Metabotropico 5/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Aprendizaje Inverso/fisiología , Estrés Psicológico/metabolismo , Animales , Conducta Apetitiva/efectos de los fármacos , Conducta Apetitiva/fisiología , Fármacos del Sistema Nervioso Central/farmacología , Corteza Cerebral/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Discapacidades para el Aprendizaje/etiología , Discapacidades para el Aprendizaje/metabolismo , Masculino , Ratones Endogámicos C57BL , Plasticidad Neuronal/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/metabolismo , Receptor del Glutamato Metabotropico 5/agonistas , Restricción Física , Aprendizaje Inverso/efectos de los fármacos , Estrés Psicológico/psicología , Técnicas de Cultivo de Tejidos
4.
Mol Psychiatry ; 24(6): 916-927, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29269844

RESUMEN

Stress can precipitate or worsen symptoms of many psychiatric disorders by dysregulating glutamatergic function within the prefrontal cortex (PFC). Previous studies suggest that antagonists of group II metabotropic glutamate (mGlu) receptors (mGlu2 and mGlu3) reduce stress-induced anhedonia through actions in the PFC, but the mechanisms by which these receptors act are not known. We now report that activation of mGlu3 induces long-term depression (LTD) of excitatory transmission in the PFC at inputs from the basolateral amygdala. Our data suggest mGlu3-LTD is mediated by postsynaptic AMPAR internalization in PFC pyramidal cells, and we observed a profound impairment in mGlu3-LTD following a single, 20-min restraint stress exposure. Finally, blocking mGlu3 activation in vivo prevented the stress-induced maladaptive changes to amydalo-cortical physiology and motivated behavior. These data demonstrate that mGlu3 mediates stress-induced physiological and behavioral impairments and further support the potential for mGlu3 modulation as a treatment for stress-related psychiatric disorders.


Asunto(s)
Receptores de Glutamato Metabotrópico/metabolismo , Receptores de Glutamato Metabotrópico/fisiología , Aminoácidos , Amígdala del Cerebelo/fisiología , Animales , Antagonistas de Aminoácidos Excitadores/farmacología , Ácido Glutámico/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Células Piramidales/metabolismo , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA