Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 295(11): 3614-3634, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-31953327

RESUMEN

G-protein-gated inwardly-rectifying K+ (GIRK) channels are targets of Gi/o-protein-signaling systems that inhibit cell excitability. GIRK channels exist as homotetramers (GIRK2 and GIRK4) or heterotetramers with nonfunctional homomeric subunits (GIRK1 and GIRK3). Although they have been implicated in multiple conditions, the lack of selective GIRK drugs that discriminate among the different GIRK channel subtypes has hampered investigations into their precise physiological relevance and therapeutic potential. Here, we report on a highly-specific, potent, and efficacious activator of brain GIRK1/2 channels. Using a chemical screen and electrophysiological assays, we found that this activator, the bromothiophene-substituted small molecule GAT1508, is specific for brain-expressed GIRK1/2 channels rather than for cardiac GIRK1/4 channels. Computational models predicted a GAT1508-binding site validated by experimental mutagenesis experiments, providing insights into how urea-based compounds engage distant GIRK1 residues required for channel activation. Furthermore, we provide computational and experimental evidence that GAT1508 is an allosteric modulator of channel-phosphatidylinositol 4,5-bisphosphate interactions. Through brain-slice electrophysiology, we show that subthreshold GAT1508 concentrations directly stimulate GIRK currents in the basolateral amygdala (BLA) and potentiate baclofen-induced currents. Of note, GAT1508 effectively extinguished conditioned fear in rodents and lacked cardiac and behavioral side effects, suggesting its potential for use in pharmacotherapy for post-traumatic stress disorder. In summary, our findings indicate that the small molecule GAT1508 has high specificity for brain GIRK1/2 channel subunits, directly or allosterically activates GIRK1/2 channels in the BLA, and facilitates fear extinction in a rodent model.


Asunto(s)
Encéfalo/metabolismo , Extinción Psicológica/efectos de los fármacos , Miedo/efectos de los fármacos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Activación del Canal Iónico/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Regulación Alostérica/efectos de los fármacos , Amígdala del Cerebelo/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Sitios de Unión , Cognición/efectos de los fármacos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/agonistas , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/química , Células HEK293 , Atrios Cardíacos/diagnóstico por imagen , Humanos , Ligandos , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Mutación/genética , Miocardio/metabolismo , Especificidad de Órganos , Compuestos de Fenilurea/farmacología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosforilación/efectos de los fármacos , Estructura Secundaria de Proteína , Subunidades de Proteína/metabolismo , Pirazoles/farmacología , Xenopus
2.
Artículo en Inglés | MEDLINE | ID: mdl-31415826

RESUMEN

The bed nucleus of the stria terminalis (BNST) is a nodal structure in neural circuits controlling anxiety-related defensive behavioral responses. It contains neurons expressing the stress- and anxiety-related neuropeptide corticotropin-releasing hormone (Crh) as well as Crh receptors. Repeated daily subthreshold activation of Crh receptors in the BNST is known to induce a chronic anxiety-like state, but how this affects neurotransmitter-relevant gene expression in target regions of the BNST is still unclear. Since the BNST projects heavily to the dorsal raphe nucleus (DR), the main source of brain serotonin, we here tested the hypothesis that such repeated, anxiety-inducing activation of Crh receptors in the BNST alters the expression of serotonergic genes in the DR, including tph2, the gene encoding the rate-limiting enzyme for brain serotonin synthesis, and slc6a4, the gene encoding the serotonin transporter (SERT). For 5 days, adult male Wistar rats received daily, bilateral, intra-BNST microinjections of vehicle (1% bovine serum albumin in 0.9% saline, n = 11) or behaviorally subthreshold doses of urocortin 1 (Ucn1, n = 11), a potent Crh receptor agonist. Priming with Ucn1 increased tph2 mRNA expression selectively within the anxiety-related dorsal part of the DR (DRD) and decreased social interaction (SI) time, a measure of anxiety-related defensive behavioral responses in rodents. Decreased social interaction was strongly correlated with increased tph2 mRNA expression in the DRD. Together with previous studies, our data are consistent with the hypothesis that Crh-mediated control of the BNST/DRD-serotonergic system plays a key role in the development of chronic anxiety states, possibly also contributing to stress-induced relapses in drug abuse and addiction behavior.


Asunto(s)
Ansiedad/metabolismo , Ansiedad/psicología , Núcleo Dorsal del Rafe/metabolismo , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Núcleos Septales/metabolismo , Triptófano Hidroxilasa/biosíntesis , Animales , Ansiedad/genética , Enfermedad Crónica , Núcleo Dorsal del Rafe/efectos de los fármacos , Expresión Génica , Masculino , Ratas , Ratas Wistar , Receptores de Hormona Liberadora de Corticotropina/agonistas , Receptores de Hormona Liberadora de Corticotropina/genética , Núcleos Septales/efectos de los fármacos , Triptófano Hidroxilasa/genética , Urocortinas/metabolismo , Urocortinas/farmacología
3.
Transl Psychiatry ; 9(1): 33, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30670681

RESUMEN

Genetic variation in serotonin transporter (SERT) that reduces transcriptional efficiency is associated with higher anxiety and fear traits and a greater incidence of post traumatic stress disorder (PTSD). Although previous studies have shown that rats with no expression of SERT (SERT-/-) have increased baseline anxiety behaviors, SERT+/- rats with low SERT expression (and more relevant to the clinical condition with low SERT expression) do not. Yet, no systematic studies of fear acquisition/extinction or their underlying neural mechanisms have been conducted in this preclinical genetic SERT+/- model. Here we sought to determine if SERT+/- or SERT-/-, compared to wildtype, rats would show exacerbated panic responses and/or persistent conditioned fear responses that may be associated with PTSD or phobia vulnerability. Results: Only SERT-/- rats showed increased baseline anxiety-like behaviors with heightened panic respiratory responses. However SERT+/- (also SERT-/-) rats showed enhanced acquisition of fear and delayed extinction of fear that was associated with changes in serotonergic-related genes (e.g., reduced 5-HT1A receptor) and disrupted inhibition within the basolateral amygdala (BLA). Furthermore, the disrupted fear responses in SERT+/- rats were normalized with 5HT1A antagonist infusions into the BLA. Enhanced acquisition and failure to extinguish fear memories displayed by both SERT-/- and SERT+/- rats are cardinal symptoms of disabling anxiety disorders such as phobias and PTSD. The data here support the hypothesis that reduced SERT function is a genetic risk that disrupts select gene expression and network properties in the amygdala that could result in vulnerability to these syndromes.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Trastornos de Ansiedad/metabolismo , Ansiedad/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Complejo Nuclear Basolateral/metabolismo , Conducta Animal/fisiología , Condicionamiento Psicológico/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Masculino , Proteínas de Unión al ARN/genética , Ratas , Ratas Wistar , Receptor de Serotonina 5-HT1A/metabolismo
4.
Behav Pharmacol ; 30(4): 376-382, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30480550

RESUMEN

Panic attacks (PAs) are episodes of intense fear or discomfort that are accompanied by a variety of both psychological and somatic symptoms. Panic induction in preclinical models (e.g. rats) has largely been assayed through flight and avoidance behavioral tests and cardiorespiratory activity. Yet, the literature pertaining to PAs shows that thermal sensations (hot flushes/heat sensations and chills) are also a common symptom during PAs in humans. Considering that temperature alterations are objectively measurable in rodents, we hypothesized that select panicogenic drugs and stimuli induce consistent changes in thermoregulation related to hot flushes and chills. Specifically, we challenged male rats with intraperitoneal injections of the GABAergic inverse agonist FG-7142; the α2 adrenoceptor antagonist yohimbine; the serotonin agonist D-fenfluramine, and 20% CO2 (an interoceptive homeostatic challenge). We assayed core body temperature and tail skin temperature using implanted radiotelemetry probes and tail thermistors/thermal imaging camera, respectively, and found that all challenges elicited rapid, high-amplitude (~7-9°C) increase in tail skin temperature and delayed decreases (~1-3°C) in core body temperature. We propose that thermal sensations such as these may be an additional indicator of a panic response in rodents and humans, as these panicogenic compounds or stimuli are known to precipitate PAs in persons with panic disorder.


Asunto(s)
Regulación de la Temperatura Corporal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Trastorno de Pánico/fisiopatología , Animales , Temperatura Corporal/fisiología , Carbolinas/farmacología , Fenfluramina/farmacología , Masculino , Modelos Animales , Pánico/fisiología , Ratas , Ratas Sprague-Dawley , Temperatura Cutánea/efectos de los fármacos , Temperatura Cutánea/fisiología , Yohimbina/farmacología
5.
Front Neurosci ; 12: 934, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30618563

RESUMEN

Orexins (OX), also known as hypocretins, are excitatory neuropeptides with well-described roles in regulation of wakefulness, arousal, energy homeostasis, and anxiety. An additional and recently recognized role of OX is modulation of fear responses. We studied the OX neurons of the perifornical hypothalamus (PeF) which send projections to the amygdala, a region critical in fear learning and fear expression. Within the amygdala, the highest density of OX-positive fibers was detected in the central nucleus (CeA). The specific mechanisms underlying OX neurotransmission within the CeA were explored utilizing rat brain slice electrophysiology, pharmacology, and chemogenetic stimulation. We show that OX induces postsynaptic depolarization of medial CeA neurons that is mediated by OX receptor 1 (OXR1) but not OX receptor 2 (OXR2). We further characterized the mechanism of CeA depolarization by OX as phospholipase C (PLC)- and sodium-calcium exchanger (NCX)- dependent. Selective chemogenetic stimulation of OX PeF fibers recapitulated OXR1 dependent depolarization of CeA neurons. We also observed that OXR1 activity modified presynaptic release of glutamate within the CeA. Finally, either systemic or intra-CeA perfusion of OXR1 antagonist reduced the expression of conditioned fear. Together, these data suggest the PeF-CeA orexinergic pathway can modulate conditioned fear through a signal transduction mechanism involving PLC and NCX activity and that selective OXR1 antagonism may be a putative treatment for fear-related disorders.

6.
Front Pharmacol ; 8: 357, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28649201

RESUMEN

Orexin neurons originating in the perifornical and lateral hypothalamic area are highly reactive to anxiogenic stimuli and have strong projections to anxiety and panic-associated circuitry. Recent studies support a role for the orexin system and in particular the orexin 1 receptor (OX1R) in coordinating an integrative stress response. However, no selective OX1R antagonist has been systematically tested in two preclinical models of using panicogenic stimuli that induce panic attack in the majority of people with panic disorder, namely an acute hypercapnia-panic provocation model and a model involving chronic inhibition of GABA synthesis in the perifornical hypothalamic area followed by intravenous sodium lactate infusion. Here we report on a novel brain penetrant, selective and high affinity OX1R antagonist JNJ-54717793 (1S,2R,4R)-7-([(3-fluoro-2-pyrimidin-2-ylphenyl)carbonyl]-N-[5-(trifluoromethyl)pyrazin-2-yl]-7-azabicyclo[2.2.1]heptan-2-amine). JNJ-54717793 is a high affinity/potent OX1R antagonist and has an excellent selectivity profile including 50 fold versus the OX2R. Ex vivo receptor binding studies demonstrated that after oral administration JNJ-54717793 crossed the blood brain barrier and occupied OX1Rs in the rat brain. While JNJ-54717793 had minimal effect on spontaneous sleep in rats and in wild-type mice, its administration in OX2R knockout mice, selectively promoted rapid eye movement sleep, demonstrating target engagement and specific OX1R blockade. JNJ-54717793 attenuated CO2 and sodium lactate induced panic-like behaviors and cardiovascular responses without altering baseline locomotor or autonomic activity. These data confirm that selective OX1R antagonism may represent a novel approach of treating anxiety disorders, with no apparent sedative effects.

7.
Stem Cells ; 35(5): 1303-1315, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28299842

RESUMEN

Electroacupuncture (EA) performed in rats and humans using limb acupuncture sites, LI-4 and LI-11, and GV-14 and GV-20 (humans) and Bai-hui (rats) increased functional connectivity between the anterior hypothalamus and the amygdala and mobilized mesenchymal stem cells (MSCs) into the systemic circulation. In human subjects, the source of the MSC was found to be primarily adipose tissue, whereas in rodents the tissue sources were considered more heterogeneous. Pharmacological disinhibition of rat hypothalamus enhanced sympathetic nervous system (SNS) activation and similarly resulted in a release of MSC into the circulation. EA-mediated SNS activation was further supported by browning of white adipose tissue in rats. EA treatment of rats undergoing partial rupture of the Achilles tendon resulted in reduced mechanical hyperalgesia, increased serum interleukin-10 levels and tendon remodeling, effects blocked in propranolol-treated rodents. To distinguish the afferent role of the peripheral nervous system, phosphoinositide-interacting regulator of transient receptor potential channels (Pirt)-GCaMP3 (genetically encoded calcium sensor) mice were treated with EA acupuncture points, ST-36 and LIV-3, and GV-14 and Bai-hui and resulted in a rapid activation of primary sensory neurons. EA activated sensory ganglia and SNS centers to mediate the release of MSC that can enhance tissue repair, increase anti-inflammatory cytokine production and provide pronounced analgesic relief. Stem Cells 2017;35:1303-1315.


Asunto(s)
Sistema Nervioso Central/citología , Electroacupuntura , Células Madre Mesenquimatosas/citología , Tendón Calcáneo/patología , Puntos de Acupuntura , Adipocitos/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Blanco/citología , Animales , Antígenos CD/metabolismo , Miembro Anterior/fisiología , Miembro Posterior/fisiología , Humanos , Hiperalgesia/terapia , Hipotálamo/citología , Interleucina-10/sangre , Macrófagos/citología , Ratones , Red Nerviosa/fisiología , Ratas , Rotura , Células Receptoras Sensoriales/metabolismo , Proteína Desacopladora 1/metabolismo
9.
Pain ; 157(9): 2124-2140, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27537210

RESUMEN

Uncoupling the protein-protein interaction between collapsin response mediator protein 2 (CRMP2) and N-type voltage-gated calcium channel (CaV2.2) with an allosteric CRMP2-derived peptide (CBD3) is antinociceptive in rodent models of inflammatory and neuropathic pain. We investigated the efficacy, duration of action, abuse potential, and neurobehavioral toxicity of an improved mutant CRMP2 peptide. A homopolyarginine (R9)-conjugated CBD3-A6K (R9-CBD3-A6K) peptide inhibited the CaV2.2-CRMP2 interaction in a concentration-dependent fashion and diminished surface expression of CaV2.2 and depolarization-evoked Ca influx in rat dorsal root ganglia neurons. In vitro studies demonstrated suppression of excitability of small-to-medium diameter dorsal root ganglion and inhibition of subtypes of voltage-gated Ca channels. Sprague-Dawley rats with tibial nerve injury had profound and long-lasting tactile allodynia and ongoing pain. Immediate administration of R9-CBD3-A6K produced enhanced dopamine release from the nucleus accumbens shell selectively in injured animals, consistent with relief of ongoing pain. R9-CBD3-A6K, when administered repeatedly into the central nervous system ventricles of naive rats, did not result in a positive conditioned place preference demonstrating a lack of abusive liability. Continuous subcutaneous infusion of R9-CBD3-A6K over a 24- to 72-hour period reversed tactile allodynia and ongoing pain, demonstrating a lack of tolerance over this time course. Importantly, continuous infusion of R9-CBD3-A6K did not affect motor activity, anxiety, depression, or memory and learning. Collectively, these results validate the potential therapeutic significance of targeting the CaV-CRMP2 axis for treatment of neuropathic pain.


Asunto(s)
Aptámeros de Péptidos/uso terapéutico , Péptidos y Proteínas de Señalización Intercelular/química , Proteínas del Tejido Nervioso/química , Neuralgia/tratamiento farmacológico , Potenciales de Acción/efectos de los fármacos , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Aptámeros de Péptidos/farmacología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Estimulación Eléctrica , Conducta Exploratoria/efectos de los fármacos , Femenino , Ganglios Espinales/citología , Suspensión Trasera , Hiperalgesia/tratamiento farmacológico , Aprendizaje por Laberinto/efectos de los fármacos , Ratones Endogámicos C57BL , Neuralgia/patología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Umbral del Dolor/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos
10.
Animals (Basel) ; 6(8)2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27490573

RESUMEN

Current recommendations for the use of CO 2 as a euthanasia agent for rats require the use of gradual fill protocols (such as 10% to 30% volume displacement per minute) in order to render the animal insensible prior to exposure to levels of CO 2 that are associated with pain. However, exposing rats to CO 2 , concentrations as low as 7% CO 2 are reported to cause distress and 10%-20% CO 2 induces panic-associated behavior and physiology, but loss of consciousness does not occur until CO 2 concentrations are at least 40%. This suggests that the use of the currently recommended low flow volume per minute displacement rates create a situation where rats are exposed to concentrations of CO 2 that induce anxiety, panic, and distress for prolonged periods of time. This study first characterized the response of male rats exposed to normoxic 20% CO 2 for a prolonged period of time as compared to room air controls. It demonstrated that rats exposed to this experimental condition displayed clinical signs consistent with significantly increased panic-associated behavior and physiology during CO 2 exposure. When atmospheric air was then again delivered, there was a robust increase in respiration rate that coincided with rats moving to the air intake. The rats exposed to CO 2 also displayed behaviors consistent with increased anxiety in the behavioral testing that followed the exposure. Next, this study assessed the behavioral and physiologic responses of rats that were euthanized with 100% CO 2 infused at 10%, 30%, or 100% volume per minute displacement rates. Analysis of the concentrations of CO 2 and oxygen in the euthanasia chamber and the behavioral responses of the rats suggest that the use of the very low flow volume per minute displacement rate (10%) may prolong the duration of panicogenic ranges of ambient CO 2 , while the use of the higher flow volume per minute displacement rate (100%) increases agitation. Therefore, of the volume displacement per minute rates evaluated, this study suggests that 30% minimizes the potential pain and distress experienced by the animal.

11.
Menopause ; 23(11): 1257-1266, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27465717

RESUMEN

OBJECTIVE: As longitudinal studies determined that anxiety is a strong risk factor for hot flashes, we hypothesized that an anxiogenic stimulus that signals air hunger (hypercapnic, normoxic gas) would trigger an exacerbated hot flash-associated increase in tail skin temperature (TST) in a rat ovariectomy (OVEX) model of surgical menopause and hot flashes in symptomatic postmenopausal women. We also assessed TST responses in OVEX serotonin transporter (SERT) rats that models a common polymorphism that is associated with increased climacteric symptoms in postmenopausal women and increases in anxiety traits. METHODS: OVEX and sham-OVEX rats (initial experiment) and wildtype and SERT OVEX rats (subsequent experiment) were exposed to a 5-minute infusion of 20% carbon dioxide (CO2) normoxic gas while measuring TST. Postmenopausal women were given brief 20% and 35% CO2 challenges, and hot flashes were self-reported and objectively verified. RESULTS: Compared to controls, OVEX rats had exacerbated increases in TST, and SERT OVEX rats had prolonged TST increases following CO2. Most women reported mild/moderate hot flashes after CO2 challenges, and the hot flash severity to CO2 was positively correlated with daily hot flash frequency. CONCLUSIONS: The studies demonstrate that this anxiogenic stimulus is capable of inducing cutaneous vasomotor responses in OVEX rats, and eliciting hot flashes in postmenopausal women. In rats, the severity of the response was mediated by loss of ovarian function and increased anxiety traits (SERT), and, in women, by daily hot flash frequency. These findings may provide insights into anxiety-related triggers and genetic risk factors for hot flashes in thermoneutral environments.


Asunto(s)
Ansiedad/fisiopatología , Dióxido de Carbono/administración & dosificación , Sofocos/fisiopatología , Sofocos/psicología , Posmenopausia/fisiología , Animales , Ansiedad/etiología , Femenino , Sofocos/genética , Humanos , Mutación con Pérdida de Función , Persona de Mediana Edad , Modelos Animales , Ovariectomía , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética
12.
Psychoneuroendocrinology ; 65: 127-37, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26765933

RESUMEN

Distressing symptoms such as hot flashes and sleep disturbances affect over 70% of women approaching menopause for an average of 4-7 years, and recent large cohort studies have shown that anxiety and stress are strongly associated with more severe and persistent hot flashes and can induce hot flashes. Although high estrogen doses alleviate symptoms, extended use increases health risks, and current non-hormonal therapies are marginally better than placebo. The lack of effective non-hormonal treatments is largely due to the limited understanding of the mechanisms that underlie menopausal symptoms. One mechanistic pathway that has not been explored is the wake-promoting orexin neuropeptide system. Orexin is exclusively synthesized in the estrogen receptor rich perifornical hypothalamic region, and has an emerging role in anxiety and thermoregulation. In female rodents, estrogens tonically inhibit expression of orexin, and estrogen replacement normalizes severely elevated central orexin levels in postmenopausal women. Using an ovariectomy menopause model, we demonstrated that an anxiogenic compound elicited exacerbated hot flash-associated increases in tail skin temperature (TST, that is blocked with estrogen), and cellular responses in orexin neurons and efferent targets. Furthermore, systemic administration of centrally active, selective orexin 1 or 2 and dual receptor antagonists attenuated or blocked TST responses, respectively. This included the reformulated Suvorexant, which was recently FDA-approved for treating insomnia. Collectively, our data support the hypothesis that dramatic loss of estrogen tone during menopausal states leads to a hyperactive orexin system that contributes to symptoms such as anxiety, insomnia, and more severe hot flashes. Additionally, orexin receptor antagonists may represent a novel non-hormonal therapy for treating menopausal symptoms, with minimal side effects.


Asunto(s)
Ansiolíticos/farmacología , Regulación de la Temperatura Corporal/efectos de los fármacos , Regulación de la Temperatura Corporal/fisiología , Menopausia/efectos de los fármacos , Orexinas/fisiología , Animales , Estrógenos/farmacología , Femenino , Hipotálamo/efectos de los fármacos , Menopausia/fisiología , Modelos Animales , Neuronas/efectos de los fármacos , Antagonistas de los Receptores de Orexina/farmacología , Ovariectomía , Ratas , Ratas Sprague-Dawley , Vasodilatación/efectos de los fármacos
13.
Pharmacol Biochem Behav ; 138: 174-9, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26476009

RESUMEN

The basolateral and lateral amygdala nuclei complex (BLC) is implicated in a number of emotional responses including conditioned fear and social anxiety. Based on previous studies demonstrating that enhanced serotonin release in the BLC leads to increased anxiety and fear responses, we hypothesized that pharmacologically depleting serotonin in the BLC using 5,7-dihydroxytryptamine (5,7-DHT) injections would lead to diminished anxiety and disrupted fear conditioning. To test this hypothesis, 5,7-DHT(a serotonin-depleting agent) was bilaterally injected into the BLC. Desipramine (a norepinephrine reuptake inhibitor) was systemically administered to prevent non-selective effects on norepinephrine. After 5days, 5-7-DHT-treated rats showed increases in the duration of social interaction (SI) time, suggestive of reduced anxiety-like behavior. We then used a cue-induced fear conditioning protocol with shock as the unconditioned stimulus and tone as the conditioned stimulus for rats pretreated with bilateral 5,7-DHT, or vehicle, injections into the BLC. Compared to vehicle-treated rats, 5,7-DHT rats had reduced acquisition of fear during conditioning (measured by freezing time during tone), also had reduced fear retrieval/recall on subsequent testing days. Ex vivo analyses revealed that 5,7-DHT reduced local 5-HT concentrations in the BLC by ~40% without altering local norepinephrine or dopamine concentrations. These data provide additional support for 5-HT playing a critical role in modulating anxiety-like behavior and fear-associated memories through its actions within the BLC.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Ansiedad/tratamiento farmacológico , Ansiedad/psicología , Condicionamiento Psicológico/efectos de los fármacos , Miedo/efectos de los fármacos , Miedo/psicología , Antagonistas de la Serotonina/farmacología , Serotonina/metabolismo , 5,7-Dihidroxitriptamina/farmacología , Inhibidores de Captación Adrenérgica/farmacología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Señales (Psicología) , Desipramina/farmacología , Electrochoque , Relaciones Interpersonales , Masculino , Microinyecciones , Ratas , Ratas Wistar , Serotoninérgicos/farmacología
14.
Depress Anxiety ; 32(9): 671-83, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26332431

RESUMEN

BACKGROUND: The neuropeptides orexin A and B play a role in reward and feeding and are critical for arousal. However, it was not initially appreciated that most prepro-orexin synthesizing neurons are almost exclusively concentrated in the perifornical hypothalamus, which when stimulated elicits panic-associated behavior and cardiovascular responses in rodents and self-reported "panic attacks" and "fear of dying" in humans. More recent studies support a role for the orexin system in coordinating an integrative stress response. For instance, orexin neurons are highly reactive to anxiogenic stimuli, are hyperactive in anxiety pathology, and have strong projections to anxiety and panic-associated circuitry. Although the two cognate orexin receptors are colocalized in many brain regions, the orexin 2 receptor (OX2R) most robustly maps to the histaminergic wake-promoting region, while the orexin 1 receptor (OX1R) distribution is more exclusive and dense in anxiety and panic circuitry regions, such as the locus ceruleus. Overall, this suggests that OX1Rs play a critical role in mobilizing anxiety and panic responses. METHODS: Here, we used a CO2 -panic provocation model to screen a dual OX1/2R antagonist (DORA-12) to globally inhibit orexin activity, then a highly selective OX1R antagonist (SORA1, Compound 56) or OX2R antagonist (SORA2, JnJ10397049) to assess OX1R and OX2R involvement. RESULTS: All compounds except the SORA2 attenuated CO2 -induced anxiety-like behaviors, and all but the SORA2 and DORA attenuated CO2 -induced cardiovascular responses. CONCLUSIONS: SORA1s may represent a novel method of treating anxiety disorders, with no apparent sedative effects that were present with a benzodiazepine.


Asunto(s)
Sistema Nervioso Autónomo/fisiopatología , Encéfalo/metabolismo , Neuronas , Receptores de Orexina/metabolismo , Trastorno de Pánico/metabolismo , Trastorno de Pánico/psicología , Ansiedad/metabolismo , Ansiedad/fisiopatología , Ansiedad/psicología , Nivel de Alerta , Dióxido de Carbono , Sistema Cardiovascular/efectos de los fármacos , Humanos , Neuronas/metabolismo , Pánico , Trastorno de Pánico/inducido químicamente , Trastorno de Pánico/fisiopatología , Recompensa
15.
J Pharmacol Exp Ther ; 352(3): 590-601, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25583879

RESUMEN

Orexins (OXs) are peptides produced by perifornical (PeF) and lateral hypothalamic neurons that exert a prominent role in arousal-related processes, including stress. A critical role for the orexin-1 receptor (OX1R) in complex emotional behavior is emerging, such as overactivation of the OX1R pathway being associated with panic or anxiety states. Here we characterize a brain-penetrant, selective, and high-affinity OX1R antagonist, compound 56 [N-({3-[(3-ethoxy-6-methylpyridin-2-yl)carbonyl]-3-azabicyclo[4.1.0]hept-4-yl}methyl)-5-(trifluoromethyl)pyrimidin-2-amine]. Ex vivo receptor binding studies demonstrated that, after subcutaneous administration, compound 56 crossed the blood-brain barrier and occupied OX1Rs in the rat brain at lower doses than standard OX1R antagonists GSK-1059865 [5-bromo-N-({1-[(3-fluoro-2-methoxyphenyl)carbonyl]-5-methylpiperidin-2-yl}methyl)pyridin-2-amine], SB-334867 [1-(2-methyl-1,3-benzoxazol-6-yl)-3-(1,5-naphthyridin-4-yl)urea], and SB-408124 [1-(6,8-difluoro-2-methylquinolin-4-yl)-3-[4-(dimethylamino)phenyl]urea]. Although compound 56 did not alter spontaneous sleep in rats and in wild-type mice, its administration in orexin-2 receptor knockout mice selectively promoted rapid eye movement sleep, demonstrating target engagement and specific OX1R blockade. In a rat model of psychological stress induced by cage exchange, the OX1R antagonist prevented the prolongation of sleep onset without affecting sleep duration. In a rat model of panic vulnerability (involving disinhibition of the PeF OX region) to threatening internal state changes (i.e., intravenous sodium lactate infusion), compound 56 attenuated sodium lactate-induced panic-like behaviors and cardiovascular responses without altering baseline locomotor or autonomic activity. In conclusion, OX1R antagonism represents a novel therapeutic strategy for the treatment of various psychiatric disorders associated with stress or hyperarousal states.


Asunto(s)
Aminopiridinas/uso terapéutico , Nivel de Alerta/fisiología , Antagonistas de los Receptores de Orexina , Receptores de Orexina/metabolismo , Piperidinas/uso terapéutico , Estrés Psicológico/metabolismo , Estrés Psicológico/prevención & control , Aminopiridinas/metabolismo , Aminopiridinas/farmacología , Animales , Nivel de Alerta/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Células HEK293 , Humanos , Hipnóticos y Sedantes , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Piperidinas/metabolismo , Piperidinas/farmacología , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley
16.
Alcohol Clin Exp Res ; 38(5): 1275-83, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24611993

RESUMEN

BACKGROUND: Corticotropin-releasing hormone (CRH) and urocortins (UCNs) bind to corticotropin-releasing hormone type 2 receptor (CRF2 receptor ), a Gs protein-coupled receptor that plays an important role in modulation of anxiety and stress responses. The Crhr2 gene maps to a quantitative trait locus (QTL) for alcohol preference on chromosome 4 previously identified in inbred alcohol-preferring (iP) and-nonpreferring (iNP) F2 rats. METHODS: Real-time polymerase chain reaction was utilized to screen for differences in Crhr2 mRNA expression in the central nervous system (CNS) of male iP and iNP rats. DNA sequence analysis was then performed to screen for polymorphism in Crhr2 in order to identify genetic variation, and luciferase reporter assays were then applied to test their functional significance. Next, binding assays were used to determine whether this polymorphism affected CRF2 receptor binding affinity as well as CRF2 receptor density in the CNS. Finally, social interaction and corticosterone levels were measured in the P and NP rats before and after 30-minute restraint stress. RESULTS: Crhr2 mRNA expression studies found lower levels of Crhr2 mRNA in iP rats compared to iNP rats. In addition, DNA sequencing identified polymorphisms in the promoter region, coding region, and 3'-untranslated region between the iP and iNP rats. A 7 bp insertion in the Crhr2 promoter of iP rats altered expression in vitro as measured by reporter assays, and we found that CRF2 receptor density was lower in the amygdala of iP as compared to iNP rats. Male P rats displayed decreased social interaction and significantly higher corticosterone levels directly following 30-minute restraint when compared to male NP rats. CONCLUSIONS: This study identified Crhr2 as a candidate gene of interest underlying the chromosome 4 QTL for alcohol consumption that was previously identified in the P and NP model. Crhr2 promoter polymorphism is associated with reduced mRNA expression in certain brain regions, particularly the amygdala, and lowered the density of CRF2 receptor in the amygdala of iP compared to iNP rats. Together, these differences between the animals may contribute to the drinking disparity as well as the anxiety differences of the P and NP rats.


Asunto(s)
Alcoholismo/genética , Sistema Hipotálamo-Hipofisario/fisiopatología , Sistema Hipófiso-Suprarrenal/fisiopatología , Receptores de Hormona Liberadora de Corticotropina/genética , Alcoholismo/fisiopatología , Animales , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Corticosterona/sangre , Masculino , Polimorfismo de Nucleótido Simple/genética , Sitios de Carácter Cuantitativo , Ratas , Ratas Endogámicas , Receptores de Hormona Liberadora de Corticotropina/análisis , Receptores de Hormona Liberadora de Corticotropina/fisiología , Estrés Psicológico/fisiopatología
17.
Artículo en Inglés | MEDLINE | ID: mdl-23523745

RESUMEN

Rats treated with three daily urocortin 1 (UCN) injections into the basolateral amygdala (BLA; i.e., UCN/BLA-primed rats) develop prolonged anxiety-associated behavior and vulnerability to panic-like physiological responses (i.e., tachycardia, hypertension and tachypnea) following intravenous infusions of 0.5 M sodium lactate (NaLac, an ordinarily mild interoceptive stressor). In these UCN-primed rats, the osmosensitive subfornical organ (SFO) may be a potential site that detects increases in plasma NaLac and mobilizes panic pathways since inhibiting the SFO blocks panic following NaLac in this model. Furthermore, since SFO neurons synthesize angiotensin II (A-II), we hypothesized that the SFO projects to the BLA and releases A-II to mobilizing panic responses in UCN/BLA-primed rats following NaLac infusions. To test this hypothesis, rats received daily bilateral injections of UCN or vehicle into the BLA daily for 3 days. Five to seven days following the intra-BLA injections, we microinjected either the nonspecific A-II type 1 (AT1r) and 2 (AT2r) receptor antagonist saralasin, or the AT2r-selective antagonist PD123319 into the BLA prior to the NaLac challenge. The UCN/BLA-primed rats pre-injected with saralasin, but not PD123319 or vehicle, had reduced NaLac-induced anxiety-associated behavior and panic-associated tachycardia and tachypnea responses. We then confirmed the presence of AT1rs in the BLA using immunohistochemistry which, combined with the previous data, suggest that A-II's panicogenic effects in the BLA is AT1r dependent. Surprisingly, the SFO had almost no neurons that directly innervate the BLA, which suggests an indirect pathway for relaying the NaLac signal. Overall these results are the first to implicate A-II and AT1rs as putative neurotransmitter-receptors in NaLac induced panic-like responses in UCN/BLA-primed rats.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Angiotensina II/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Receptores de Angiotensina/metabolismo , Urocortinas/administración & dosificación , Amígdala del Cerebelo/fisiología , Análisis de Varianza , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Toxina del Cólera/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Frecuencia Cardíaca/efectos de los fármacos , Imidazoles/farmacología , Masculino , Microinyecciones , Vías Nerviosas/fisiología , Dolor/inducido químicamente , Proteínas Proto-Oncogénicas c-fos/metabolismo , Piridinas/farmacología , Ratas , Ratas Wistar , Lactato de Sodio/toxicidad
18.
J Psychopharmacol ; 27(2): 152-61, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22914798

RESUMEN

Rats with chronic inhibition of GABA synthesis by infusion of l-allyglycine, a glutamic acid decarboxylase inhibitor, into their dorsomedial/perifornical hypothalamus are anxious and exhibit panic-like cardio-respiratory responses to treatment with intravenous (i.v.) sodium lactate (NaLac) infusions, in a manner similar to what occurs in patients with panic disorder. We previously showed that either NMDA receptor antagonists or metabotropic glutamate receptor type 2/3 receptor agonists can block such a NaLac response, suggesting that a glutamate mechanism is contributing to this panic-like state. Using this animal model of panic, we tested the efficacy of CBiPES and THIIC, which are selective group II metabotropic glutamate type 2 receptor allosteric potentiators (at 10-30 mg/kg i.p.), in preventing NaLac-induced panic-like behavioral and cardiovascular responses. The positive control was alprazolam (3mg/kg i.p.), a clinically effective anti-panic benzodiazepine. As predicted, panic-prone rats given a NaLac challenge displayed NaLac-induced panic-like cardiovascular (i.e. tachycardia and hypertensive) responses and "anxiety" (i.e. decreased social interaction time) and "flight" (i.e. increased locomotion) -associated behaviors; however, systemic injection of the panic-prone rats with CBiPES, THIIC or alprazolam prior to the NaLac dose blocked all NaLac-induced panic-like behaviors and cardiovascular responses. These data suggested that in a rat animal model, selective group II metabotropic glutamate type 2 receptor allosteric potentiators show an anti-panic efficacy similar to alprazolam.


Asunto(s)
Ansiolíticos/farmacología , Conducta Animal/efectos de los fármacos , Trastorno de Pánico/tratamiento farmacológico , Pánico/efectos de los fármacos , Receptores de Glutamato Metabotrópico/metabolismo , Alprazolam/farmacología , Animales , Ansiedad/tratamiento farmacológico , Sistema Cardiovascular/efectos de los fármacos , Ácido Glutámico/metabolismo , Masculino , Trastorno de Pánico/inducido químicamente , Ratas , Ratas Sprague-Dawley , Lactato de Sodio/efectos adversos , Ácido gamma-Aminobutírico/farmacología
19.
Prog Brain Res ; 198: 133-61, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22813973

RESUMEN

A panic response is an adaptive response to deal with an imminent threat and consists of an integrated pattern of behavioral (aggression, fleeing, or freezing) and increased cardiorespiratory and endocrine responses that are highly conserved across vertebrate species. In the 1920s and 1940s, Philip Bard and Walter Hess, respectively, determined that the posterior regions of the hypothalamus are critical for a "fight-or-flight" reaction to deal with an imminent threat. Since the 1940s it was determined that the posterior hypothalamic panic area was located dorsal (perifornical hypothalamus: PeF) and dorsomedial (dorsomedial hypothalamus: DMH) to the fornix. This area is also critical for regulating circadian rhythms and in 1998, a novel wake-promoting neuropeptide called orexin (ORX)/hypocretin was discovered and determined to be almost exclusively synthesized in the DMH/PeF perifornical hypothalamus and adjacent lateral hypothalamus. The most proximally emergent role of ORX is in regulation of wakefulness through interactions with efferent systems that mediate arousal and energy homeostasis. A hypoactive ORX system is also linked to narcolepsy. However, ORX role in more complex emotional responses is emerging in more recent studies where ORX is linked to depression and anxiety states. Here, we review data that demonstrates ORX ability to mobilize a coordinated adaptive panic/defense response (anxiety, cardiorespiratory, and endocrine components), and summarize the evidence that supports a hyperactive ORX system being linked to pathological panic and anxiety states.


Asunto(s)
Ansiedad/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuropéptidos/fisiología , Pánico/fisiología , Animales , Ansiedad/metabolismo , Humanos , Hipotálamo/fisiología , Orexinas , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
20.
Physiol Behav ; 107(5): 733-42, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-22554617

RESUMEN

BACKGROUND: Although the hypothalamic orexin system is known to regulate appetitive behaviors and promote wakefulness and arousal (Sakurai, 2007 [56]), this system may also be important in adaptive and pathological anxiety/stress responses (Suzuki et al., 2005 [4]). In a recent study, we demonstrated that CSF orexin levels were significantly higher in patients experiencing panic attacks compared to non-panicking depressed subjects (Johnson et al., 2010 [9]). Furthermore, genetically silencing orexin synthesis or blocking orexin 1 receptors attenuated lactate-induced panic in an animal model of panic disorder. Therefore, in the present study, we tested if orexin (ORX) modulates panic responses and brain pathways activated by two different panicogenic drugs. METHODS: We conducted a series of pharmacological, behavioral, physiological and immunohistochemical experiments to study the modulation by the orexinergic inputs of anxiety behaviors, autonomic responses, and activation of brain pathways elicited by systemic injections of anxiogenic/panicogenic drugs in rats. RESULTS: We show that systemic injections of two different anxiogenic/panicogenic drugs (FG-7142, an inverse agonist at the benzodiazepine site of the GABA(A) receptor, and caffeine, a nonselective competitive adenosine receptor antagonist) increased c-Fos induction in a specific subset of orexin neurons located in the dorsomedial/perifornical (DMH/PeF) but not the lateral hypothalamus. Pretreating rats with an orexin 1 receptor antagonist attenuated the FG-7142-induced anxiety-like behaviors, increased heart rate, and neuronal activation in key panic pathways, including subregions of the central nucleus of the amygdala, bed nucleus of the stria terminalis, periaqueductal gray and in the rostroventrolateral medulla. CONCLUSION: Overall, the data here suggest that the ORX neurons in the DMH/PeF region are critical to eliciting coordinated panic responses and that ORX1 receptor antagonists constitute a potential novel treatment strategy for panic and related anxiety disorders. The neural pathways through which ORX1 receptor antagonists attenuate panic responses involve the extended amygdala, periaqueductal gray, and medullary autonomic centers.


Asunto(s)
Encéfalo/efectos de los fármacos , Pánico/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de Neuropéptido/efectos de los fármacos , Amígdala del Cerebelo/efectos de los fármacos , Amígdala del Cerebelo/fisiología , Animales , Encéfalo/fisiología , Cafeína/farmacología , Carbolinas/farmacología , Estimulantes del Sistema Nervioso Central/farmacología , Antagonistas del GABA/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/fisiología , Masculino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiología , Receptores de Orexina , Pánico/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Receptores Acoplados a Proteínas G/fisiología , Receptores de Neuropéptido/fisiología , Núcleos Septales/efectos de los fármacos , Núcleos Septales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...