Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 112(19): 6188-93, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25918363

RESUMEN

Current treatments for major depressive disorder (MDD) have a time lag and are ineffective for a large number of patients. Development of novel pharmacological therapies requires a comprehensive understanding of the molecular events that contribute to MDD pathophysiology. Recent evidence points toward aberrant activity of synaptic proteins as a critical contributing factor. In the present studies, we used viral-mediated gene transfer to target a key mediator of activity-dependent synaptic protein synthesis downstream of mechanistic target of rapamycin complex 1 (mTORC1) known as p70 S6 kinase 1 (S6K1). Targeted delivery of two mutants of S6K1, constitutively active or dominant-negative, to the medial prefrontal cortex (mPFC) of rats allowed control of the mTORC1/S6K1 translational pathway. Our results demonstrate that increased expression of S6K1 in the mPFC produces antidepressant effects in the forced swim test without altering locomotor activity. Moreover, expression of active S6K1 in the mPFC blocked the anhedonia caused by chronic stress, resulting in a state of stress resilience. This antidepressant response was associated with increased neuronal complexity caused by enhanced S6K1 activity. Conversely, expression of dominant-negative S6K1 in the mPFC resulted in prodepressive behavior in the forced swim test and was sufficient to cause anhedonia in the absence of chronic stress exposure. Together, these data demonstrate a critical role for S6K1 activity in depressive behaviors, and suggest that pathways downstream of mTORC1 may underlie the pathophysiology and treatment of MDD.


Asunto(s)
Trastorno Depresivo Mayor/metabolismo , Regulación Enzimológica de la Expresión Génica , Corteza Prefrontal/metabolismo , Proteínas Quinasas S6 Ribosómicas/fisiología , Animales , Antidepresivos/uso terapéutico , Conducta Animal , Modelos Animales de Enfermedad , Ketamina/química , Masculino , Aprendizaje por Laberinto , Neuronas/metabolismo , Fenotipo , Fosforilación , Ratas , Ratas Sprague-Dawley , Proteínas Quinasas S6 Ribosómicas/genética , Transducción de Señal , Sirolimus/química , Natación
2.
Neurobiol Dis ; 82: 254-261, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26102021

RESUMEN

Clinical studies demonstrate that scopolamine, a non-selective muscarinic acetylcholine receptor (mAchR) antagonist, produces rapid therapeutic effects in depressed patients, and preclinical studies report that the actions of scopolamine require glutamate receptor activation and the mechanistic target of rapamycin complex 1 (mTORC1). The present study extends these findings to determine the role of the medial prefrontal cortex (mPFC) and specific muscarinic acetylcholine receptor (M-AchR) subtypes in the actions of scopolamine. The administration of scopolamine increases the activity marker Fos in the mPFC, including the infralimbic (IL) and prelimbic (PrL) subregions. Microinfusions of scopolamine into either the IL or the PrL produced significant antidepressant responses in the forced swim test, and neuronal silencing of IL or PrL blocked the antidepressant effects of systemic scopolamine. The results also demonstrate that the systemic administration of a selective M1-AChR antagonist, VU0255035, produced an antidepressant response and stimulated mTORC1 signaling in the PFC, similar to the actions of scopolamine. Finally, we used a chronic unpredictable stress model as a more rigorous test of rapid antidepressant actions and found that a single dose of scopolamine or VU0255035 blocked the anhedonic response caused by CUS, an effect that requires the chronic administration of typical antidepressants. Taken together, these findings indicate that mPFC is a critical mediator of the behavioral actions of scopolamine and identify the M1-AChR as a therapeutic target for the development of novel and selective rapid-acting antidepressants.


Asunto(s)
Antidepresivos/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptor Muscarínico M1/metabolismo , Escopolamina/farmacología , Anhedonia/efectos de los fármacos , Anhedonia/fisiología , Animales , Enfermedad Crónica , Sacarosa en la Dieta , Modelos Animales de Enfermedad , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Microinyecciones , Complejos Multiproteicos/metabolismo , Antagonistas Muscarínicos/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas Sprague-Dawley , Receptor Muscarínico M1/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Sulfonamidas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Tiadiazoles/farmacología , Factores de Tiempo , Técnicas de Cultivo de Tejidos
3.
Int J Neuropsychopharmacol ; 18(1)2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25539510

RESUMEN

BACKGROUND: Recent studies demonstrate that the rapid antidepressant ketamine increases spine number and function in the medial prefrontal cortex (mPFC), and that these effects are dependent on activation of glutamate α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptors and brain-derived neurotrophic factor (BDNF). In vitro studies also show that activation of AMPA receptors stimulates BNDF release via activation of L-type voltage-dependent calcium channels (VDCC). METHODS: Based on this evidence, we examined the role of BDNF release and the impact of L-type VDCCs on the behavioral actions of ketamine. RESULTS: The results demonstrate that infusion of a neutralizing BDNF antibody into the mPFC blocks the behavioral effects of ketamine in the forced swim test (FST). In addition, we show that pretreatment with nifedipine or verapamil, two structurally-different L-type calcium channel antagonists, blocks the behavioral effects of ketamine in the FST. Finally, we show that ketamine treatment stimulates BDNF release in primary cortical neurons and that this effect is blocked by inhibition of AMPA receptors or L-type VDCCs. CONCLUSIONS: Taken together, these results indicate that the antidepressant effects of ketamine are mediated by activation of L-type VDCCs and the release of BDNF. They further elucidate the cellular mechanisms underlying this novel rapid-acting antidepressant.


Asunto(s)
Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ketamina/farmacología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/fisiopatología , Masculino , Neuronas/efectos de los fármacos , Neuronas/fisiología , Nifedipino/farmacología , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiopatología , Ratas Sprague-Dawley , Receptores AMPA/antagonistas & inhibidores , Receptores AMPA/metabolismo , Verapamilo/farmacología
4.
Proc Natl Acad Sci U S A ; 107(18): 8457-62, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20404172

RESUMEN

Phosphodiesterase 11A (PDE11A) is the most recently identified family of phosphodiesterases (PDEs), the only known enzymes to break down cyclic nucleotides. The tissue expression profile of this dual specificity PDE is controversial, and little is understood of its biological function, particularly in the brain. We seek here to determine if PDE11A is expressed in the brain and to understand its function, using PDE11A(-/-) knockout (KO) mice. We show that PDE11A mRNA and protein are largely restricted to hippocampus CA1, subiculum, and the amygdalohippocampal area, with a two- to threefold enrichment in the ventral vs. dorsal hippocampus, equal distribution between cytosolic and membrane fractions, and increasing levels of protein expression from postnatal day 7 through adulthood. Interestingly, PDE11A KO mice show subtle psychiatric-disease-related deficits, including hyperactivity in an open field, increased sensitivity to the glutamate N-methyl-D-aspartate receptor antagonist MK-801, as well as deficits in social behaviors (social odor recognition memory and social avoidance). In addition, PDE11A KO mice show enlarged lateral ventricles and increased activity in CA1 (as per increased Arc mRNA), phenotypes associated with psychiatric disease. The increased sensitivity to MK-801 exhibited by PDE11A KO mice may be explained by the biochemical dysregulation observed around the glutamate alpha-amino-3-hydroxy-5-methyl-4-isozazolepropionic (AMPA) receptor, including decreased levels of phosphorylated-GluR1 at Ser845 and the prototypical transmembrane AMPA-receptor-associated proteins stargazin (gamma2) and gamma8. Together, our data provide convincing evidence that PDE11A expression is restricted in the brain but plays a significant role in regulating brain function.


Asunto(s)
3',5'-GMP Cíclico Fosfodiesterasas/metabolismo , Hipocampo/enzimología , Trastornos Mentales/enzimología , 3',5'-GMP Cíclico Fosfodiesterasas/deficiencia , 3',5'-GMP Cíclico Fosfodiesterasas/genética , Animales , Conducta Animal , Femenino , Regulación Enzimológica de la Expresión Génica , Glutamina/metabolismo , Hipocampo/patología , Masculino , Trastornos Mentales/genética , Trastornos Mentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal , Conducta Social
5.
Int J Neuropsychopharmacol ; 15(4): 429-34, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22114864

RESUMEN

Recent studies demonstrate that ketamine, a fast-acting antidepressant, rapidly activates the mammalian target of rapamycin (mTOR) and increases synaptogenesis in the prefrontal cortex. Because of the side-effect and abuse potential of ketamine we are investigating alternative agents that produce similar effects. Here, we demonstrate that a single dose of LY 341495, an mGluR2/3 antagonist, produces ketamine-like biochemical and behavioural actions. LY 341495 administration rapidly (1 h) activates the mTOR pathway (mTOR, p70S6K, 4E-BP1) and subsequently (24 h later) increases levels of synaptic proteins (PSD-95, GluR1 and Synapsin I), similar to the effects of ketamine. Finally, the antidepressant effects of LY 341495 in the rat forced swim test are completely blocked by the mTOR inhibitor, rapamycin. The results indicate that the antidepressant actions of LY 341495 are mediated by activation of mTOR and suggest that this and other mGluR2/3 antagonists could produce rapid antidepressant effects in depressed patients.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Receptores de Glutamato Metabotrópico/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Aminoácidos/farmacología , Animales , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ketamina/farmacología , Masculino , Proteínas de la Membrana/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/ultraestructura , Ratas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Natación/psicología , Sinapsinas/metabolismo , Sinaptosomas/efectos de los fármacos , Sinaptosomas/metabolismo , Xantenos/farmacología
6.
J Pharmacol Exp Ther ; 338(1): 345-52, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21508084

RESUMEN

Metabotropic glutamate receptor 7 (mGluR7) remains the most elusive of the eight known mGluRs primarily because of the limited availability of tool compounds to interrogate its potential therapeutic utility. The discovery of N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082) as the first orally active, brain-penetrable, mGluR7-selective allosteric agonist by Mitsukawa and colleagues (Proc Natl Acad Sci USA 102:18712-18717, 2005) provides a means to investigate this receptor system directly. AMN082 demonstrates mGluR7 agonist activity in vitro and interestingly has a behavioral profile that supports utility across a broad spectrum of psychiatric disorders including anxiety and depression. The present studies were conducted to extend the in vitro and in vivo characterization of AMN082 by evaluating its pharmacokinetic and metabolite profile. Profiling of AMN082 in rat liver microsomes revealed rapid metabolism (t(1/2) < 1 min) to a major metabolite, N-benzhydrylethane-1,2-diamine (Met-1). In vitro selectivity profiling of Met-1 demonstrated physiologically relevant transporter binding affinity at serotonin transporter (SERT), dopamine transporter (DAT), and norepinephrine transporter (NET) (323, 3020, and 3410 nM, respectively); whereas the parent compound AMN082 had appreciable affinity at NET (1385 nM). AMN082 produced antidepressant-like activity and receptor occupancy at SERT up to 4 h postdose, a time point at which AMN082 is significantly reduced in brain and plasma while the concentration of Met-1 continues to increase in brain. Acute Met-1 administration produced antidepressant-like activity as would be expected from its in vitro profile as a mixed SERT, NET, DAT inhibitor. Taken together, these data suggest that the reported in vivo actions of AMN082 should be interpreted with caution, because they may involve other mechanisms in addition to mGluR7.


Asunto(s)
Compuestos de Bencidrilo/farmacología , Monoaminas Biogénicas/farmacología , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/fisiología , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Compuestos de Bencidrilo/metabolismo , Monoaminas Biogénicas/fisiología , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Masculino , Ratones , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley
7.
Bioorg Med Chem Lett ; 21(1): 195-9, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21126874

RESUMEN

A series of 3-(pyridin-2-yl-ethynyl)benzamide negative allosteric modulators of the metabotropic glutamate receptor 5 (mGluR5 NAMs) have been prepared. Starting from HTS hit 1 (IC(50): 926 nM), potent mGluR5 NAMs showing excellent potencies (IC(50)s<50 nM) and good physicochemical profiles were prepared by monitoring LipE values. One compound 26 showed excellent mGluR5 binding (K(i): 21 nM) and antagonism (IC(50): 8 nM), an excellent rat PK profile (CL: 12 mL/min/kg, %F: 85) and showed oral activity in a mouse 4-Plate Behavioral model of anxiety (MED: 30 mpk) and a mouse Stress Induced Hyperthermia model of anxiety (MED 17.8 mpk).


Asunto(s)
Benzamidas/química , Piridinas/química , Receptores de Glutamato Metabotrópico/química , Regulación Alostérica , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Benzamidas/farmacocinética , Benzamidas/uso terapéutico , Modelos Animales de Enfermedad , Ensayos Analíticos de Alto Rendimiento , Ratones , Piridinas/farmacocinética , Piridinas/uso terapéutico , Ratas , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/metabolismo
8.
Neurobiol Dis ; 39(2): 148-55, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20381618

RESUMEN

Rimonabant was the first clinically marketed cannabinoid (CB)(1) receptor antagonist developed to treat obesity. Unfortunately, CB(1) receptor antagonism produced adverse psychiatric events in patients. To determine whether this occurs pre-clinically, we investigated the effects of rimonabant in rodent models of mood disorders. Chronic treatment with rimonabant increased immobility time in the rat forced swim test and reduced the consumption of sucrose-sweetened water in an assay postulated to model anhedonia. These responses were similar to the effects elicited by chronic mild stress in these behavioral models, which, taken together, are indicative of a depression-like phenotype. Additionally, chronic treatment with rimonabant produced decreases in frontal cortex serotonin levels, marked reductions in hippocampal cell proliferation, survival, and BDNF levels, and elevations in the concentrations of pro-inflammatory cytokines including interferon gamma and TNF alpha. These preclinical findings mimic clinical reports and implicate possible mechanisms responsible for the unfavorable psychiatric events reported following chronic rimonabant use.


Asunto(s)
Fenotipo , Piperidinas/efectos adversos , Pirazoles/efectos adversos , Receptor Cannabinoide CB1/antagonistas & inhibidores , Estrés Psicológico/inducido químicamente , Estrés Psicológico/fisiopatología , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo/métodos , Preferencias Alimentarias/efectos de los fármacos , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Pérdida de Tono Postural/efectos de los fármacos , Masculino , Microdiálisis/métodos , Ratas , Ratas Sprague-Dawley , Rimonabant , Estrés Psicológico/patología , Sacarosa/administración & dosificación , Edulcorantes/administración & dosificación , Natación/psicología
9.
Int J Neuropsychopharmacol ; 13(9): 1193-205, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20047711

RESUMEN

Biogenic amines such as norepinephrine, dopamine, and serotonin play a well-described role in the treatment of mood disorders and some types of pain. As alpha2A-adrenoceptors regulate the release of these neurotransmitters, we examined the therapeutic potential of BRL 44408, a potent (Ki=8.5 nM) and selective (>50-fold) alpha2A-adrenoceptor antagonist (K(B)=7.9 nM). In rats, BRL 44408 penetrated the central nervous system resulting in peak brain and plasma concentrations of 586 ng/g and 1124 ng/ml, respectively. In a pharmacodynamic assay, pretreatment with BRL 44408 to rats responding under a fixed-ratio 30 operant response paradigm resulted in a rightward shift of the clonidine dose-response curve, an effect indicative of alpha2-adrenoceptor antagonism in vivo. Consistent with presynaptic autoreceptor antagonism and tonic regulation of neurotransmitter release, acute administration of BRL 44408 elevated extracellular concentrations of norepinephrine and dopamine, but not serotonin, in the medial prefrontal cortex. Additionally, BRL 44408, probably by inhibiting alpha2A heteroceptors, produced a significant increase in cortical levels of acetylcholine. In the forced swim test and schedule-induced polydipsia assay, BRL 44408 produced an antidepressant-like response by dose-dependently decreasing immobility time and adjunctive water intake, respectively, while in a model of visceral pain, BRL 44408 exhibited analgesic activity by decreasing para-phenylquinone (PPQ)-induced abdominal stretching. Finally, BRL 44408 did not produce deficits in overall motor coordination nor alter general locomotor activity. This preclinical characterization of the neurochemical and behavioural profile of BRL 44408 suggests that selective antagonism of alpha2A-adrenoceptors may represent an effective treatment strategy for mood disorders and visceral pain.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Analgésicos/farmacología , Antidepresivos/farmacología , Depresión/tratamiento farmacológico , Imidazoles/farmacología , Isoindoles/farmacología , Receptores Adrenérgicos alfa 2/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Analgésicos/farmacocinética , Animales , Antidepresivos/farmacocinética , Monoaminas Biogénicas/metabolismo , Encéfalo/metabolismo , Células CHO , Cricetinae , Cricetulus , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Imidazoles/farmacocinética , Isoindoles/farmacocinética , Masculino , Ratones , Microdiálisis , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Natación , Sed/efectos de los fármacos
10.
Psychopharmacology (Berl) ; 203(1): 41-52, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18949460

RESUMEN

RATIONALE: Acid sensing ion channels (ASICs) are proton-gated ion channels located in the central and peripheral nervous systems. Of particular interest is ASIC1a, which is located in areas associated with fear and anxiety behaviors. Recent reports suggest a role for ASIC1a in preclinical models of fear conditioning and anxiety. OBJECTIVES: The present experiments evaluated various ASIC inhibitors in preclinical models of autonomic and behavioral parameters of anxiety. In addition, neurochemical studies evaluated the effects of an ASIC inhibitor (A-317567) on neurotransmitter levels in the amygdala. RESULTS: In electrophysiological studies using hippocampal primary neuronal cultures, three ASIC inhibitors (PcTX-1, A-317567, and amiloride) produced concentration-dependent inhibition of acid-evoked currents. In the stress-induced hyperthermia model, acute administration of psalmotoxin 1 (PcTX-1; 10-56 ng, i.c.v.), A-317567 (0.1-1.0 mg/kg, i.p.), and amiloride (10-100 mg/kg, i.p.) prevented stress-induced elevations in core body temperature. In the four-plate test, acute treatment with PcTX-1 (10-56 ng, i.c.v.) and A-317567 (0.01-0.1 mg/kg, i.p.), but not amiloride (3-100 mg/kg, i.p.), produced dose-dependent and significant increases in the number of punished crossings relative to vehicle-treated animals. Additionally, PcTX-1 (56-178 ng, i.c.v.), A-317567 (0.1-10 mg/kg, i.p.), and amiloride (10-100 mg/kg, i.p.) lacked significant anxiolytic-like activity in the elevated zero maze. In neurochemical studies, an infusion of A-317567 (100 microM) into the amygdala significantly elevated the extracellular levels of GABA, but not glutamate, in this brain region. CONCLUSIONS: These findings demonstrate that ASIC inhibition produces anxiolytic-like effects in some behavioral models and indicate a potential role for GABAergic mechanisms to underlie these anxiolytic-like effects.


Asunto(s)
Ansiolíticos/farmacología , Ansiedad/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Bloqueadores de los Canales de Sodio/farmacología , Canales Iónicos Sensibles al Ácido , Amilorida/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/metabolismo , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos/métodos , Conducta Exploratoria/efectos de los fármacos , Fiebre/metabolismo , Fiebre/prevención & control , Fiebre/psicología , Ácido Glutámico/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/embriología , Hipocampo/metabolismo , Isoquinolinas/farmacología , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microdiálisis , Naftalenos/farmacología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Péptidos , Ratas , Ratas Sprague-Dawley , Canales de Sodio/metabolismo , Venenos de Araña/farmacología , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Ácido gamma-Aminobutírico/metabolismo
11.
Am J Psychiatry ; 176(5): 388-400, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30606046

RESUMEN

OBJECTIVE: The N-methyl-d-aspartate receptor antagonist ketamine produces rapid and sustained antidepressant actions even in patients with treatment-resistant depression. Vascular endothelial growth factor (VEGF) has been implicated in the effects of conventional monoamine-based antidepressants, but the role of VEGF in the rapid antidepressant actions of ketamine remains unclear. The authors examined whether neuronal VEGF signaling in the medial prefrontal cortex (mPFC) mediates the rapid antidepressant actions of ketamine. METHODS: The authors used a combination of approaches, including conditional, neuron-specific knockout of VEGF or its receptor, Flk-1; antibody neutralization; viral-mediated knockdown of Flk-1; and pharmacological inhibitors. Further in vitro and in vivo experiments were performed to examine whether neuronal VEGF signaling was required for the neurotrophic and synaptogenic actions of ketamine that underlie its behavioral actions. RESULTS: The behavioral actions of systemic ketamine are blocked by forebrain excitatory neuron-specific deletion of either VEGF or Flk-1 or by intra-mPFC infusion of a VEGF neutralizing antibody. Moreover, intra-mPFC infusions of VEGF are sufficient to produce rapid ketamine-like behavioral actions, and these effects are blocked by neuron-specific Flk-1 deletion. The results also show that local knockdown of Flk-1 in mPFC excitatory neurons in adulthood blocks the behavioral effects of systemic ketamine. Moreover, inhibition of neuronal VEGF signaling blocks the neurotrophic and synaptogenic effects of ketamine. CONCLUSIONS: Together, these findings indicate that neuronal VEGF-Flk-1 signaling in the mPFC plays an essential role in the antidepressant actions of ketamine.


Asunto(s)
Antagonistas de Aminoácidos Excitadores/farmacología , Ketamina/farmacología , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptor 2 de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Animales , Anticuerpos Neutralizantes/farmacología , Conducta Animal/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Técnicas de Inactivación de Genes , Técnicas In Vitro , Ratones , Neuronas/metabolismo , Neuronas/patología , Corteza Prefrontal/metabolismo , Quinazolinas/farmacología , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
Neurobiol Dis ; 31(3): 334-41, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18606547

RESUMEN

The diuretic amiloride has recently proven neuroprotective in models of cerebral ischemia, a property attributable to the drug's inhibition of central acid-sensing ion channels (ASICs). Given that Parkinson's disease (PD), like ischemia, is associated with cerebral lactic acidosis, we tested amiloride in the MPTP-treated mouse, a model of PD also manifesting lactic acidosis. Amiloride was found to protect substantia nigra (SNc) neurons from MPTP-induced degeneration, as determined by attenuated reductions in striatal tyrosine hydroxylase (TH) and dopamine transporter (DAT) immunohistochemistry, as well as smaller declines in striatal DAT radioligand binding and dopamine levels. More significantly, amiloride also preserved dopaminergic cell bodies in the SNc. Administration of psalmotoxin venom (PcTX), an ASIC1a blocker, resulted in a much more modest effect, attenuating only the deficits in striatal DAT binding and dopamine. These findings represent the first experimental evidence of a potential role for ASICs in the pathogenesis of Parkinson's disease.


Asunto(s)
Acidosis Láctica/tratamiento farmacológico , Amilorida/farmacología , Fármacos Neuroprotectores/farmacología , Trastornos Parkinsonianos/tratamiento farmacológico , Sustancia Negra/efectos de los fármacos , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina , Canales Iónicos Sensibles al Ácido , Acidosis Láctica/etiología , Acidosis Láctica/fisiopatología , Animales , Antiparkinsonianos/farmacología , Unión Competitiva/efectos de los fármacos , Unión Competitiva/fisiología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/metabolismo , Péptidos , Ensayo de Unión Radioligante , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio/metabolismo , Venenos de Araña/farmacología , Sustancia Negra/metabolismo , Sustancia Negra/fisiopatología , Tirosina 3-Monooxigenasa/metabolismo
13.
Neuropharmacology ; 54(7): 1136-42, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18423777

RESUMEN

Recent studies have reported that estrogen has antidepressant-like effects in animal models. In this study we used the highly selective ER beta agonist, WAY-200070, to examine the role of ER beta activation on brain neurochemistry and activity in antidepressant and anxiolytic models in male mice. Within 15 min of administration, WAY-200070 (30 mg/kg s.c.) caused the nuclear translocation of striatal ER beta receptors from the cytosol. WAY-200070 also increased c-fos activation 4h, but not 15 min after administration. Both nuclear translocation and c-fos induction effects of WAY-200070 demonstrate that WAY-200070 has bound to estrogen receptors and triggered downstream events. The absence of these effects in the ER beta KO mice confirms that WAY-200070 was targeting ER beta. Administration of WAY-200070 (30 mg/kg s.c.) produced a delayed approximately 50% increase in dopamine in the striatum of wild type mice. The effect was significant and maintained from 90 to 240 min. This increase was absent in ER beta KO mice. In wild type mice, WAY-200070 (30 mg/kg s.c.) also produced a delayed and transient approximately 100% increase in 5-HT. To further investigate the role of ER beta receptors on serotonergic function, 5-HTP accumulation was measured. ER beta KO mice were found to have reduced frontal cortex levels of 5-HTP, indicating reduced tryptophan hydroxylase activity. WAY-200070 (3-30 mg/kg s.c.) was also tested in behavioural models. WAY-200070 (30 mg/kg s.c.) reduced immobility time in the mouse tail suspension test indicating an antidepressant-like effect. WAY-200070 (30 mg/kg) showed anxiolytic-like effects in the four-plate test (increased punished crossings) and stress-induced hyperthermia (attenuation of hyperthermic response). The effects of the selective ER beta agonist, WAY-200070, on dopamine and serotonin, the anxiolytic-like and antidepressant-like effects as well as the genotype specific effects on neurochemistry support that positive modulation of ER beta function may provide a novel treatment for affective disorders.


Asunto(s)
Ansiolíticos/uso terapéutico , Antidepresivos/uso terapéutico , Ansiedad/tratamiento farmacológico , Depresión/tratamiento farmacológico , Receptor beta de Estrógeno/agonistas , Oxazoles/uso terapéutico , Fenoles/uso terapéutico , 5-Hidroxitriptófano/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Receptor alfa de Estrógeno/deficiencia , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/deficiencia , Receptor beta de Estrógeno/metabolismo , Suspensión Trasera/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microdiálisis , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Factores de Tiempo
14.
Psychopharmacology (Berl) ; 197(4): 601-11, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18311561

RESUMEN

RATIONALE: Neuropeptide S (NPS) and its receptor (NPSR) comprise a recently deorphaned G protein-coupled receptor system. Recent reports implicate NPS in the mediation of anxiolytic-like activity in rodents. OBJECTIVES: To extend the characterization of NPS, the present studies examined the in vitro pharmacology of mouse NPSR and the in vivo pharmacology of NPS in three preclinical mouse models predictive of anxiolytic action: the four-plate test (FPT), elevated zero maze (EZM), and stress-induced hyperthermia (SIH). The ability of NPS to produce antidepressant-like effects in the tail suspension test (TST) was also investigated. RESULTS: In vitro, mouse NPS 1-20 (mNPS 1-20) and the C-terminal glutamine-truncated mouse NPS 1-19 bound mNPSR with high affinity (Ki = 0.203 +/- 0.060, 0.635 +/- 0.141 nM, respectively) and potently activated intracellular calcium release (EC50 = 3.73 +/- 1.08, 4.10 +/- 1.25 nM). NPS produced effects in vivo consistent with anxiolytic-like activity. In FPT, NPS increased punished crossings (minimal effective dose [MED]: mNPS 1-20 = 0.2 microg, mNPS(1-19) = 0.02 microg), similar to the reference anxiolytic, alprazolam (MED 0.5 microg). NPS increased the percentage of time spent in the open quadrants of EZM (MED: mNPS 1-20 = 0.1 microg, mNPS 1-19 = 1.0 microg), like the reference anxiolytic, chlordiazepoxide (MED 56 microg). In SIH, NPS attenuated stress-induced increases in body temperature similar to alprazolam but with a large potency difference between the NPS peptides (MED: mNPS 1-20 = 2.0 microg, mNPS 1-19 = 0.0002 microg) and mNPS 1-20 increased baseline temperature. Unlike fluoxetine, NPS did not effect immobility time in TST, indicating a lack of antidepressant-like activity. CONCLUSIONS: These data provide an important confirmation and expansion of the anxiolytic-like effects of NPS and implicate the NPS system as a novel target for anxiolytic drug discovery.


Asunto(s)
Ansiolíticos/farmacología , Ansiolíticos/uso terapéutico , Trastornos de Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Modelos Animales de Enfermedad , Neuropéptidos/farmacología , Neuropéptidos/uso terapéutico , Alprazolam/farmacología , Alprazolam/uso terapéutico , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Trastornos de Ansiedad/psicología , Nivel de Alerta/efectos de los fármacos , Regulación de la Temperatura Corporal/efectos de los fármacos , Calcio/metabolismo , Línea Celular , Clordiazepóxido/farmacología , Clordiazepóxido/uso terapéutico , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Miedo/efectos de los fármacos , Fluoxetina/farmacología , Fluoxetina/uso terapéutico , Humanos , Técnicas In Vitro , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Actividad Motora/efectos de los fármacos , Oxitocina , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/uso terapéutico , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/fisiología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/fisiología , Relación Estructura-Actividad
15.
Neuropharmacology ; 111: 242-252, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27634096

RESUMEN

Recent preclinical and clinical studies demonstrate that three functionally different compounds, the NMDA receptor channel blocker ketamine, mGlu2/3 receptor antagonist LY341495, and NMDA receptor glycine site agent GLYX-13 produce rapid and long lasting antidepressant effects. Furthermore, these agents are reported to stimulate ERK and mTORC1 signaling in brain. Here we used rat primary cortical culture neurons to further examine the cellular actions of these agents. The results demonstrate that low concentrations of all three compounds rapidly increase levels of the phosphorylated and activated forms of ERK and a downstream target of mTORC1, p70S6 kinase, in a concentration and time dependent manner. In addition, each compound rapidly increases BDNF release into the culture media. Further studies demonstrate that induction of BDNF release, as well as stimulation of phospho-ERK is blocked by incubation with an AMPA receptor antagonist. The requirement for AMPA receptor stimulation suggests that the effects of these rapid agents are activity dependent. This possibility is supported by studies demonstrating that neuronal silencing, via incubation with the GABAA receptor agonist muscimol, completely blocks phospho-ERK and BDNF release by each agent. Finally, incubation with each drug for 24 h increases the number and length of neuronal branches. Together, the results demonstrate that these three different rapid acting antidepressant agents increase ERK signaling and BDNF release in an activity dependent manner that leads to increased neuronal complexity. Further studies will be required to determine the exact mechanisms underlying these effects in cultured neurons and in rodent models.


Asunto(s)
Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Aminoácidos/farmacología , Animales , Células Cultivadas , Corteza Cerebral/citología , Dendritas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Ketamina/farmacología , Oligopéptidos/farmacología , Cultivo Primario de Células , Ratas , Receptor trkB/metabolismo , Receptores AMPA/metabolismo , Xantenos/farmacología
16.
Biol Psychiatry ; 73(12): 1189-98, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23295207

RESUMEN

Antidepressants that produce rapid and robust effects, particularly for severely ill patients, represent one of the largest unmet medical needs for the treatment of depression. Currently available drugs that modulate monoamine neurotransmission provide relief for only a subset of patients, and this minimal efficacy requires several weeks of chronic treatment. The recent discovery that the glutamatergic agent ketamine produces rapid antidepressant responses within hours has opened a new area of research to explore the molecular mechanisms through which ketamine produces these surprising responses. Clinical and preclinical findings have exposed some of the unique actions of ketamine and identified a cell-signaling pathway known as the mammalian target of rapamycin. Activation of mammalian target of rapamycin and increased synaptogenesis in the prefrontal cortex are crucial in mediating the antidepressant effects of ketamine. Importantly, the synaptic actions of ketamine allow rapid recovery from the insults produced by exposure to repeated stress that cause neuronal atrophy and loss of synaptic connections. In the following review, we explore some of the clinical and preclinical findings that have thrust ketamine to the forefront of rapid antidepressant research and unveiled some of its unique molecular and cellular actions.


Asunto(s)
Antidepresivos/farmacología , Neurogénesis/efectos de los fármacos , Sirolimus/metabolismo , Sinapsis/efectos de los fármacos , Animales , Antidepresivos/uso terapéutico , Humanos , Mamíferos
17.
J Mol Psychiatry ; 1(1): 15, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25408908

RESUMEN

BACKGROUND: Depression is a prevalent neuropsychiatric disorder that affects an estimated 350 million people worldwide. Currently available treatments for depression are lacking in both speed of onset and efficacy. Recent pharmacological efforts have targeted the glutamatergic neurotransmitter system using the N-methyl-D-aspartate (NMDA) receptor antagonist ketamine to produce rapid and robust antidepressant effects, however the widespread clinical use of ketamine is limited due to side effects and abuse liability. More recently, work evaluating metabotropic mGluR2/3 receptor antagonists has demonstrated many similarities with ketamine. METHODS: Male, Sprague-Dawley rats were exposed to a chronic unpredictable stress paradigm, which produces decreased sucrose preference, a measure of anhedonia. Rats were then treated with vehicle or a single injection of the mGluR2/3 antagonist LY341495 (3 mg/kg, i.p.) and tested at 24 hrs, 48 hrs or 10 days after a single treatment. RESULTS: We demonstrate that a single treatment with LY341495 produces a rapid (within 1-2 days) and long-lasting (10 days) reversal of anhedonia caused by chronic unpredictable stress in rats. This model provides a rigorous test of rapid-acting agents as typical antidepressants require several weeks of treatment to produce a response. CONCLUSIONS: These data suggest that LY341495 has the ability to produce rapid and robust antidepressant effects similar to ketamine. Together, the results highlight the potential for similar compounds to produce rapid and lasting efficacy for the treatment of depression.

18.
Neuropsychopharmacology ; 38(11): 2268-77, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23680942

RESUMEN

A single dose of the short-acting NMDA antagonist ketamine produces rapid and prolonged antidepressant effects in treatment-resistant patients with major depressive disorder (MDD), which are thought to occur via restoration of synaptic connectivity. However, acute dissociative side effects and eventual fading of antidepressant effects limit widespread clinical use of ketamine. Recent studies in medial prefrontal cortex (mPFC) show that the synaptogenic and antidepressant-like effects of a single standard dose of ketamine in rodents are dependent upon activation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) signaling pathway together with inhibitory phosphorylation of glycogen synthase kinase-3 (GSK-3), which relieves its inhibitory in influence on mTOR. Here, we found that the synaptogenic and antidepressant-like effects of a single otherwise subthreshold dose of ketamine were potentiated when given together with a single dose of lithium chloride (a nonselective GSK-3 inhibitor) or a preferential GSK-3ß inhibitor; these effects included rapid activation of the mTORC1 signaling pathway, increased inhibitory phosphorylation of GSK-3ß, increased synaptic spine density/diameter, increased excitatory postsynaptic currents in mPFC layer V pyramidal neurons, and antidepressant responses that persist for up to 1 week in the forced-swim test model of depression. The results demonstrate that low, subthreshold doses of ketamine combined with lithium or a selective GSK-3 inhibitor are equivalent to higher doses of ketamine, indicating the pivotal role of the GSK-3 pathway in modulating the synaptogenic and antidepressant responses to ketamine. The possible mitigation by GSK-3 inhibitors of the eventual fading of ketamine's antidepressant effects remains to be explored.


Asunto(s)
Antidepresivos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Ketamina/farmacología , Cloruro de Litio/farmacología , Sinapsis/efectos de los fármacos , Animales , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/ultraestructura , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Glucógeno Sintasa Quinasa 3/metabolismo , Pérdida de Tono Postural/efectos de los fármacos , Indoles/farmacología , Masculino , Maleimidas/farmacología , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Fosforilación , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/fisiología , Ratas , Transducción de Señal/efectos de los fármacos , Sinapsis/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
19.
Curr Opin Cell Biol ; 23(6): 730-7, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21996102

RESUMEN

Depression is associated with structural alterations in limbic brain regions that control emotion and mood. Studies of chronic stress in animal models and postmortem tissue from depressed subjects demonstrate that these structural alterations result from atrophy and loss of neurons and glial cells. These findings indicate that depression and stress-related mood disorders can be considered mild neurodegenerative disorders. Importantly, there is evidence that these structural alterations can be blocked or even reversed by elimination of stress and by antidepressant treatments. A major focus of current investigations is to characterize the molecular signaling pathways and factors that underlie these effects of stress, depression, and antidepressant treatment. Recent advances in this research area are discussed and potential novel targets for antidepressant development are highlighted.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Depresión/patología , Animales , Atrofia/tratamiento farmacológico , Encéfalo/patología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/patología , Humanos , Trastornos del Humor/tratamiento farmacológico , Trastornos del Humor/patología , Neuroglía/patología , Neuronas/patología
20.
Biol Psychiatry ; 69(8): 754-61, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21292242

RESUMEN

BACKGROUND: Despite widely reported clinical and preclinical studies of rapid antidepressant actions of glutamate N-methyl-D-aspartate (NMDA) receptor antagonists, there has been very little work examining the effects of these drugs in stress models of depression that require chronic administration of antidepressants or the molecular mechanisms that could account for the rapid responses. METHODS: We used a rat 21-day chronic unpredictable stress (CUS) model to test the rapid actions of NMDA receptor antagonists on depressant-like behavior, neurochemistry, and spine density and synaptic function of prefrontal cortex neurons. RESULTS: The results demonstrate that acute treatment with the noncompetitive NMDA channel blocker ketamine or the selective NMDA receptor 2B antagonist Ro 25-6981 rapidly ameliorates CUS-induced anhedonic and anxiogenic behaviors. We also found that CUS exposure decreases the expression levels of synaptic proteins and spine number and the frequency/amplitude of synaptic currents (excitatory postsynaptic currents) in layer V pyramidal neurons in the prefrontal cortex and that these deficits are rapidly reversed by ketamine. Blockade of the mammalian target of rapamycin protein synthesis cascade abolishes both the behavioral and biochemical effects of ketamine. CONCLUSIONS: The results indicate that the structural and functional deficits resulting from long-term stress exposure, which could contribute to the pathophysiology of depression, are rapidly reversed by NMDA receptor antagonists in a mammalian target of rapamycin dependent manner.


Asunto(s)
Conducta Animal/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Neuronas/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología , Sinapsis/efectos de los fármacos , Animales , Conducta Animal/fisiología , Western Blotting , Conducta de Elección/efectos de los fármacos , Conducta de Elección/fisiología , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/fisiología , Electrofisiología , Ketamina/farmacología , Neuronas/fisiología , Fenoles/farmacología , Piperidinas/farmacología , Corteza Prefrontal/fisiopatología , Ratas , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Sirolimus/farmacología , Sinapsis/fisiología , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA