Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Transl Psychiatry ; 14(1): 60, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38272876

ABSTRACT

The serotonin (5-HT) system is heavily implicated in the regulation of anxiety and trauma-related disorders such as panic disorder and post-traumatic stress disorder, respectively. However, the neural mechanisms of how serotonergic neurotransmission regulates innate panic and fear brain networks are poorly understood. Our earlier studies have identified that orexin (OX)/glutamate neurons within the perifornical hypothalamic area (PFA) play a critical role in adaptive and pathological panic and fear. While site-specific and electrophysiological studies have shown that intracranial injection and bath application of 5-HT inhibits PFA neurons via 5-HT1a receptors, they largely ignore circuit-specific neurotransmission and its physiological properties that occur in vivo. Here, we investigate the role of raphe nuclei 5-HT inputs into the PFA in panic and fear behaviors. We initially confirmed that photostimulation of glutamatergic neurons in the PFA of rats produces robust cardioexcitation and flight/aversive behaviors resembling panic-like responses. Using the retrograde tracer cholera toxin B, we determined that the PFA receives discrete innervation of serotonergic neurons clustered in the lateral wings of the dorsal (lwDRN) and in the median (MRN) raphe nuclei. Selective lesions of these serotonergic projections with saporin toxin resulted in similar panic-like responses during the suffocation-related CO2 challenge and increased freezing to fear-conditioning paradigm. Conversely, selective stimulation of serotonergic fibers in the PFA attenuated both flight/escape behaviors and cardioexcitation responses elicited by the CO2 challenge and induced conditioned place preference. The data here support the hypothesis that PFA projecting 5-HT neurons in the lwDRN/MRN represents a panic/fear-off circuit and may also play a role in reward behavior.


Subject(s)
Carbon Dioxide , Serotonin , Rats , Animals , Serotonin/physiology , Rats, Wistar , Fear/physiology , Panic/physiology , Serotonergic Neurons
2.
Neuroscience ; 491: 176-184, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35351573

ABSTRACT

The chemogenetic procedure DREADD (designer receptor exclusively activated by designer drugs) is an inventive way to selectively affect g-coupled protein receptors. In theory, DREADD receptors are only activated by administering inert compounds, primarily clozapine N-oxide (CNO). Research has shown that CNO does not cross the blood-brain barrier, and CNO is converted back to clozapine and N-desmethylclozapine (N-Des) in the brain. Clozapine and N-Des have many neurological effects including alterations in glutamate and dopamine (DA) levels in multiple brain regions. The current study examined the effects of peripheral administration of CNO on glutamate and DA levels in the medial prefrontal cortex (mPFC). Wistar rats were administered CNO, and microdialysis samples were collected from the mPFC. Administration of CNO significantly increased glutamate (31-87%) and DA (65-126%), CNO-induced increases in DA occurred for a longer duration than glutamate, and that for the two highest doses of CNO there was a significant correlation between the increase in glutamate and DA in the mPFC. In the mPFC, CNO-induced increases in DA occurred at 0.5 mg/kg, while increases in glutamate were observed at doses greater than 1.0 mg/kg. The source of the DA and glutamate could be caused by activation of projection neurons or local effects. The data replicate findings that CNO is not an inert compound and that interpretation of CNO-activated DREADD findings should be done with caution. The data indicate that low ('safe') doses of CNO still have neurochemical effects and that controlling for the actions of clozapine/N-Des in CNO-DREADD studies has many concerns.


Subject(s)
Clozapine , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Dopamine , Glutamic Acid , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
3.
Cell Rep ; 38(1): 110179, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986353

ABSTRACT

G protein-coupled receptors (GPCRs) in intestinal enteroendocrine cells (EECs) respond to nutritional, neural, and microbial cues and modulate the release of gut hormones. Here we show that Gpr17, an orphan GPCR, is co-expressed in glucagon-like peptide-1 (GLP-1)-expressing EECs in human and rodent intestinal epithelium. Acute genetic ablation of Gpr17 in intestinal epithelium improves glucose tolerance and glucose-stimulated insulin secretion (GSIS). Importantly, inducible knockout (iKO) mice and Gpr17 null intestinal organoids respond to glucose or lipid ingestion with increased secretion of GLP-1, but not the other incretin glucose-dependent insulinotropic polypeptide (GIP). In an in vitro EEC model, overexpression or agonism of Gpr17 reduces voltage-gated calcium currents and decreases cyclic AMP (cAMP) production, and these are two critical factors regulating GLP-1 secretion. Together, our work shows that intestinal Gpr17 signaling functions as an inhibitory pathway for GLP-1 secretion in EECs, suggesting intestinal GPR17 is a potential target for diabetes and obesity intervention.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Intestinal Mucosa/metabolism , Nerve Tissue Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Blood Glucose/analysis , Calcium/metabolism , Cell Line , Cyclic AMP/metabolism , Diabetes Mellitus/pathology , Female , Gastric Inhibitory Polypeptide/metabolism , Glucose Tolerance Test , HEK293 Cells , HeLa Cells , Humans , Incretins/metabolism , Insulin/metabolism , Insulin Secretion/physiology , Intestinal Mucosa/cytology , Male , Mice , Mice, Knockout , Obesity/pathology , Receptors, Gastrointestinal Hormone/metabolism
4.
Peptides ; 134: 170403, 2020 12.
Article in English | MEDLINE | ID: mdl-32882352

ABSTRACT

Atrial Naturietic Peptide (ANP) is a neuropeptide that regulates function of the hypothalamic-pituitary-adrenal (HPA) axis, immune and neuroimmune system, and epigenetic factors. Research has indicated that ANP may mediate alcohol intake, withdrawal, and craving like behaviors. ANP receptors are present in the mesocorticolimbic (MCL) reward pathway of the brain, which includes the nucleus accumbens (Acb) and the ventral tegmental area (VTA). The objectives of the present study were to examine the effects of ANP microinjected into Acb subregions (Shell (Sh), Core (Co), ventral to AcbSh) on operant ethanol (EtOH) self-administration and into posterior VTA (pVTA) on EtOH-seeking behavior of female alcohol-preferring (P) rats. In the first experiment, ANP (0, 10 µg, or 100 µg) was microinjected into subregions of the Acb to determine its effects on EtOH self-administration. In the second experiment, ANP was microinjected into pVTA to determine its effects on Pavlovian Spontaneous Recovery (PSR) of responding, a measure of context-induced EtOH-seeking behavior. Administration of ANP directly into the AcbSh significantly reduced EtOH self-administration compared to vehicle, whereas ANP into the AcbCo or areas directly ventral to the AcbSh did not alter responding for EtOH. Microinjection of ANP into the pVTA significantly reduced responding on the EtOH-associated lever during the PSR test. The data indicate that activation of ANP systems in the (a) AcbSh can inhibit EtOH intake, and (b) in the pVTA can inhibit EtOH-seeking behavior. The results suggest that manipulations of the ANP system could be a potential target for pharmacotherapeutic intervention to treat alcohol use disorder. Supported in part by AA07462, AA07611, AA10717, AA10721, AA013522, AA019366, AA020908, AA022287, and AA024612.


Subject(s)
Alcohol Drinking/prevention & control , Atrial Natriuretic Factor/pharmacology , Drug-Seeking Behavior/drug effects , Nucleus Accumbens/drug effects , Ventral Tegmental Area/drug effects , Alcohol Drinking/metabolism , Alcohol Drinking/pathology , Animals , Central Nervous System Depressants/toxicity , Disease Models, Animal , Female , Microinjections/methods , Nucleus Accumbens/metabolism , Rats , Self Administration/methods , Ventral Tegmental Area/metabolism
5.
J Pharmacol Exp Ther ; 374(3): 366-375, 2020 09.
Article in English | MEDLINE | ID: mdl-32527792

ABSTRACT

In humans, alcohol is consumed for its rewarding and anxiolytic effects. The central nucleus of the amygdala (CeA) is considered a neuronal nexus that regulates fear, anxiety, and drug self-administration. Manipulations of the CeA alter ethanol (EtOH) consumption under numerous EtOH self-administration models. The experiments determined whether EtOH is reinforcing/anxiolytic within the CeA, whether selective breeding for high alcohol consumption alters the rewarding properties of EtOH in the CeA, and whether the reinforcing/anxiolytic effects of EtOH in the CeA are mediated by the neuropeptides corticotropin-releasing factor (CRF) and nociceptin. The reinforcing properties of EtOH were determined by having male Wistar and Taconic alcohol-preferring (tP) rats self-administer EtOH directly into the CeA. The expression of anxiety-like behaviors was assessed through multiple behavioral models (social interaction, acoustic startle, and open field). Coadministration of EtOH and a CRF1 antagonist (NBI35965) or nociceptin on self-administration into the CeA and anxiety-like behaviors was determined. EtOH was self-administered directly into the lateral CeA, and tP rats self-administered a lower concentration of EtOH than Wistar rats. EtOH microinjected into the lateral CeA reduced the expression of anxiety-like behaviors, indicating an anxiolytic effect. Coadministration of NBI35965 failed to alter the rewarding/anxiolytic properties of EtOH in the CeA. In contrast, coadministration of the nociceptin enhanced both EtOH reward and anxiolysis in the CeA. Overall, the data indicate that the lateral CeA is a key anatomic location that mediates the rewarding and anxiolytic effects of EtOH, and local nociceptin receptors, but not local CRF1 receptors, are involved in these behaviors. SIGNIFICANCE STATEMENT: Alcohol is consumed for the stimulatory, rewarding, and anxiolytic properties of the drug of abuse. The current data are the first to establish that alcohol is reinforcing and anxiolytic within the lateral central nucleus of the amygdala (CeA) and that the nociceptin system regulates these effects of alcohol within the CeA.


Subject(s)
Anti-Anxiety Agents/pharmacology , Central Amygdaloid Nucleus/drug effects , Ethanol/pharmacology , Genetic Background , Opioid Peptides/metabolism , Reward , Animals , Behavior, Animal/drug effects , Central Amygdaloid Nucleus/physiology , Dose-Response Relationship, Drug , Male , Rats , Rats, Wistar , Social Behavior , Nociceptin
6.
J Psychopharmacol ; 34(4): 400-411, 2020 04.
Article in English | MEDLINE | ID: mdl-32153226

ABSTRACT

BACKGROUND: The central serotonergic system originating from the dorsal raphe nucleus (DR) plays a critical role in anxiety and trauma-related disorders such as posttraumatic stress disorder. Although many studies have investigated the role of serotonin (5-HT) within pro-fear brain regions such as the amygdala, the majority of these studies have utilized non-selective pharmacological approaches or poorly understood lesioning techniques which limit their interpretation. AIM: Here we investigated the role of amygdala-projecting 5-HT neurons in the DR in innate anxiety and conditioned fear behaviors. METHODS: To achieve this goal, we utilized (1) selective lesion of 5-HT neurons projecting to the amygdala with saporin toxin conjugated to anti-serotonin transporter (SERT) injected into the amygdala, and (2) optogenetic excitation of amygdala-projecting DR cell bodies with a combination of a retrogradely transported canine adenovirus-expressing Cre-recombinase injected into the amygdala and a Cre-dependent-channelrhodopsin injected into the DR. RESULTS: While saporin treatment lesioned both local amygdalar 5-HT fibers and neurons in the DR as well as reduced conditioned fear behavior, optical activation of amygdala-projecting DR neurons enhanced anxious behavior and conditioned fear response. CONCLUSION: Collectively, these studies support the hypothesis that amygdala-projecting 5-HT neurons in the DR represent an anxiety and fear-on network.


Subject(s)
Amygdala/physiology , Anxiety/psychology , Dorsal Raphe Nucleus/physiology , Fear/psychology , Gain of Function Mutation , Serotonergic Neurons , Animals , Conditioning, Classical , Male , Nerve Net/drug effects , Neural Pathways/drug effects , Optogenetics , Photic Stimulation , Rats , Rats, Wistar , Saporins/pharmacology , Social Interaction
7.
J Biol Chem ; 295(11): 3614-3634, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31953327

ABSTRACT

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.


Subject(s)
Brain/metabolism , Extinction, Psychological/drug effects , Fear/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/drug effects , Small Molecule Libraries/pharmacology , Allosteric Regulation/drug effects , Amygdala/metabolism , Animals , Behavior, Animal/drug effects , Binding Sites , Cognition/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , HEK293 Cells , Heart Atria/diagnostic imaging , Humans , Ligands , Mice, Inbred C57BL , Motor Activity/drug effects , Mutation/genetics , Myocardium/metabolism , Organ Specificity , Phenylurea Compounds/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation/drug effects , Protein Structure, Secondary , Protein Subunits/metabolism , Pyrazoles/pharmacology , Xenopus
8.
Brain Res ; 1731: 145942, 2020 03 15.
Article in English | MEDLINE | ID: mdl-30205108

ABSTRACT

Orexin has been implicated in a number of physiological functions, including arousal, regulation of sleep, energy metabolism, appetitive behaviors, stress, anxiety, fear, panic, and cardiovascular control. In this review, we will highlight research focused on orexin system in the medial hypothalamic regions of perifornical (PeF) and dorsomedial hypothalamus (DMH), and describe the role of this hypothalamic neuropeptide in the behavioral expression of panic and consequent fear and avoidance responses, as well as sympathetic regulation and possible development of chronic hypertension. We will also outline recent data highlighting the clinical potential of single and dual orexin receptor antagonists for neuropsychiatric conditions including panic, phobia, and cardiovascular conditions, such as in hypertension.


Subject(s)
Hypertension/physiopathology , Hypothalamus, Middle/physiology , Orexins/physiology , Panic/physiology , Phobic Disorders/physiopathology , Animals , Brain/drug effects , Brain/physiology , Humans , Hypertension/prevention & control , Neural Pathways/drug effects , Neural Pathways/physiology , Orexin Receptor Antagonists/administration & dosage , Panic/drug effects , Phobic Disorders/prevention & control , Stress, Psychological/physiopathology
9.
Transl Psychiatry ; 9(1): 33, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670681

ABSTRACT

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.


Subject(s)
Amygdala/metabolism , Anxiety Disorders/metabolism , Anxiety/metabolism , RNA-Binding Proteins/metabolism , Animals , Basolateral Nuclear Complex/metabolism , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Fear/physiology , Male , RNA-Binding Proteins/genetics , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism
10.
Prog Brain Res ; 241: 113-158, 2018.
Article in English | MEDLINE | ID: mdl-30447753

ABSTRACT

Autism spectrum disorders (ASD) represent a heterogeneous group of disorders defined by deficits in social interaction/communication and restricted interests, behaviors, or activities. Models of ASD, developed based on clinical data and observations, are used in basic science, the "bench," to better understand the pathophysiology of ASD and provide therapeutic options for patients in the clinic, the "bedside." Translational medicine creates a bridge between the bench and bedside that allows for clinical and basic science discoveries to challenge one another to improve the opportunities to bring novel therapies to patients. From the clinical side, biomarker work is expanding our understanding of possible mechanisms of ASD through measures of behavior, genetics, imaging modalities, and serum markers. These biomarkers could help to subclassify patients with ASD in order to better target treatments to a more homogeneous groups of patients most likely to respond to a candidate therapy. In turn, basic science has been responding to developments in clinical evaluation by improving bench models to mechanistically and phenotypically recapitulate the ASD phenotypes observed in clinic. While genetic models are identifying novel therapeutics targets at the bench, the clinical efforts are making progress by defining better outcome measures that are most representative of meaningful patient responses. In this review, we discuss some of these challenges in translational research in ASD and strategies for the bench and bedside to bridge the gap to achieve better benefits to patients.


Subject(s)
Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/drug therapy , Biomarkers , Disease Models, Animal , Translational Research, Biomedical , Animals , Humans
11.
Prog Brain Res ; 241: 37-62, 2018.
Article in English | MEDLINE | ID: mdl-30447756

ABSTRACT

Neurofibromatosis type 1 (NF1) is monogenic neurodevelopmental disorder caused by mutation of NF1 gene, which leads to increased susceptibility to various tumors formations. Additionally, majority of patients with NF1 are experience high incidence of cognitive deficits. Particularly, we review the growing number of reports demonstrated a higher incidence of autism spectrum disorder (ASD) in individuals with NF1. In this review we also discuss face validity of preclinical Nf1 mouse models. Then we describe discoveries from these animal models that have uncovered the deficiencies in the regulation of Ras and other intracellular pathways as critical mechanisms underlying the Nf1 cognitive problems. We also summarize and interpret recent preclinical and clinical studies that point toward potential pharmacological therapies for NF1 patients.


Subject(s)
Autism Spectrum Disorder , Cognitive Dysfunction , Disease Models, Animal , GABA Antagonists/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Neurofibromatosis 1 , Protein Kinase Inhibitors/pharmacology , Animals , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/physiopathology , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Humans , Mice , Neurofibromatosis 1/drug therapy , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/physiopathology
12.
Front Neurosci ; 12: 934, 2018.
Article in English | MEDLINE | ID: mdl-30618563

ABSTRACT

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.

14.
Psychoneuroendocrinology ; 65: 127-37, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26765933

ABSTRACT

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.


Subject(s)
Anti-Anxiety Agents/pharmacology , Body Temperature Regulation/drug effects , Body Temperature Regulation/physiology , Menopause/drug effects , Orexins/physiology , Animals , Estrogens/pharmacology , Female , Hypothalamus/drug effects , Menopause/physiology , Models, Animal , Neurons/drug effects , Orexin Receptor Antagonists/pharmacology , Ovariectomy , Rats , Rats, Sprague-Dawley , Vasodilation/drug effects
15.
Pharmacol Biochem Behav ; 138: 174-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26476009

ABSTRACT

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.


Subject(s)
Amygdala/metabolism , Anxiety/drug therapy , Anxiety/psychology , Conditioning, Psychological/drug effects , Fear/drug effects , Fear/psychology , Serotonin Antagonists/pharmacology , Serotonin/metabolism , 5,7-Dihydroxytryptamine/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Amygdala/drug effects , Animals , Cues , Desipramine/pharmacology , Electroshock , Interpersonal Relations , Male , Microinjections , Rats , Rats, Wistar , Serotonin Agents/pharmacology
16.
Nat Neurosci ; 17(11): 1583-90, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25242307

ABSTRACT

Children with neurofibromatosis type 1 (NF1) are increasingly recognized as having a high prevalence of social difficulties and autism spectrum disorders (ASDs). We demonstrated a selective social learning deficit in mice with deletion of a single Nf1 allele (Nf1(+/-)), along with greater activation of the mitogen-activated protein kinase pathway in neurons from the amygdala and frontal cortex, structures that are relevant to social behaviors. The Nf1(+/-) mice showed aberrant amygdala glutamate and GABA neurotransmission, deficits in long-term potentiation and specific disruptions in the expression of two proteins that are associated with glutamate and GABA neurotransmission: a disintegrin and metalloprotease domain 22 (Adam22) and heat shock protein 70 (Hsp70), respectively. All of these amygdala disruptions were normalized by the additional deletion of the p21 protein-activated kinase (Pak1) gene. We also rescued the social behavior deficits in Nf1(+/-) mice with pharmacological blockade of Pak1 directly in the amygdala. These findings provide insights and therapeutic targets for patients with NF1 and ASDs.


Subject(s)
Amygdala/metabolism , Hippocampus/physiology , Learning/physiology , Neurofibromin 1/metabolism , Social Behavior , p21-Activated Kinases/metabolism , Animals , Behavior, Animal , Child Development Disorders, Pervasive/metabolism , Child Development Disorders, Pervasive/physiopathology , Disease Models, Animal , Long-Term Potentiation/physiology , Male , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Neurofibromin 1/deficiency
17.
Nature ; 500(7461): 217-21, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23842490

ABSTRACT

The inner ear contains sensory epithelia that detect head movements, gravity and sound. It is unclear how to develop these sensory epithelia from pluripotent stem cells, a process that will be critical for modelling inner ear disorders or developing cell-based therapies for profound hearing loss and balance disorders. So far, attempts to derive inner ear mechanosensitive hair cells and sensory neurons have resulted in inefficient or incomplete phenotypic conversion of stem cells into inner-ear-like cells. A key insight lacking from these previous studies is the importance of the non-neural and preplacodal ectoderm, two critical precursors during inner ear development. Here we report the stepwise differentiation of inner ear sensory epithelia from mouse embryonic stem cells (ESCs) in three-dimensional culture. We show that by recapitulating in vivo development with precise temporal control of signalling pathways, ESC aggregates transform sequentially into non-neural, preplacodal and otic-placode-like epithelia. Notably, in a self-organized process that mimics normal development, vesicles containing prosensory cells emerge from the presumptive otic placodes and give rise to hair cells bearing stereocilia bundles and a kinocilium. Moreover, these stem-cell-derived hair cells exhibit functional properties of native mechanosensitive hair cells and form specialized synapses with sensory neurons that have also arisen from ESCs in the culture. Finally, we demonstrate how these vesicles are structurally and biochemically comparable to developing vestibular end organs. Our data thus establish a new in vitro model of inner ear differentiation that can be used to gain deeper insight into inner ear development and disorder.


Subject(s)
Cell Differentiation , Ear, Inner/cytology , Hair Cells, Auditory, Inner/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Ear, Inner/embryology , Electrical Synapses/physiology , Gene Expression Regulation, Developmental , Humans , Mice , Myosin VIIa , Myosins/genetics , Myosins/metabolism , Neurogenesis , Recombinant Proteins/metabolism
18.
Neuropsychopharmacology ; 38(7): 1352-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23358240

ABSTRACT

Neuropeptide Y (NPY) administration into the basolateral amygdala (BLA) decreases anxiety-like behavior, mediated in part through the Y1 receptor (Y1R) isoform. Activation of Y1Rs results in G-protein-mediated reduction of cAMP levels, which results in reduced excitability of amygdala projection neurons. Understanding the mechanisms linking decreased cAMP levels to reduced excitability in amygdala neurons is important for identifying novel anxiolytic targets. We studied the intracellular mechanisms of activation of Y1Rs on synaptic transmission in the BLA. Activating Y1Rs by [Leu(31),Pro(34)]-NPY (L-P NPY) reduced the amplitude of evoked NMDA-mediated excitatory postsynaptic currents (eEPSCs), without affecting AMPA-mediated eEPSCs, but conversely increased the amplitude of GABAA-mediated evoked inhibitory postsynaptic currents (eIPSCs). Both effects were abolished by the Y1R antagonist, PD160170. Intracellular GDP-ß-S, or pre-treatment with either forskolin or 8Br-cAMP, eliminated the effects of L-P NPY on both NMDA- and GABAA-mediated currents. Thus, both the NMDA and GABAA effects of Y1R activation in the BLA are G-protein-mediated and cAMP-dependent. Pipette inclusion of protein kinase A (PKA) catalytic subunit blocked the effect of L-P NPY on GABAA-mediated eIPSCs, but not on NMDA-mediated eEPSCs. Conversely, activating the exchange protein activated by cAMP (Epac) with 8CPT-2Me-cAMP blocked the effect of L-P NPY on NMDA-mediated eEPSCs, but not on GABAA-mediated eIPSCs. Thus, NPY regulates amygdala excitability via two signal-transduction events, with reduced PKA activity enhancing GABAA-mediated eIPSCs and Epac deactivation reducing NMDA-mediated eEPSCs. This multipathway regulation of NMDA- and GABAA-mediated currents may be important for NPY plasticity and stress resilience in the amygdala.


Subject(s)
Amygdala/physiology , Neuropeptide Y/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Neuropeptide/physiology , Signal Transduction/physiology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Aminoquinolines/pharmacology , Amygdala/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Colforsin/pharmacology , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Guanine Nucleotide Exchange Factors/drug effects , Guanosine Diphosphate/analogs & derivatives , Guanosine Diphosphate/pharmacology , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Neurons/drug effects , Neurons/physiology , Neuropeptide Y/administration & dosage , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/antagonists & inhibitors , Neuropeptide Y/pharmacology , Rats , Receptors, AMPA/drug effects , Receptors, AMPA/physiology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, GABA-A/drug effects , Receptors, GABA-A/physiology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/physiology , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/antagonists & inhibitors , Signal Transduction/drug effects , Sulfones/pharmacology , Thionucleotides/pharmacology
19.
Prog Brain Res ; 198: 133-61, 2012.
Article in English | MEDLINE | ID: mdl-22813973

ABSTRACT

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.


Subject(s)
Anxiety/physiopathology , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Panic/physiology , Animals , Anxiety/metabolism , Humans , Hypothalamus/physiology , Orexins , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
20.
Physiol Behav ; 107(5): 726-32, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-22652097

ABSTRACT

The hypothalamic neuropeptide orexin (ORX) has been implicated in anxiety, and anxiety-like behaviors. The purpose of these studies was to determine the role of ORX, specifically orexin-A (ORX-A) in the bed nucleus of the stria terminalis (BNST) on anxiety-like behaviors in rats. Rats injected with ORX-A into the BNST displayed greater anxiety-like measures in the social interaction and elevated plus maze tests compared to vehicle treated controls. Such anxiety-like behaviors were not observed when the ORX-A injections were adjacent to the BNST, in the medial septum. The anxiety-inducing effects of direct infusions of ORX-A into the BNST may be a consequence of increased activation of BNST neurons. In BNST slice preparations using patch-clamp techniques, ORX-A induced membrane depolarization and generation of action potentials in a subset of BNST neurons. The anxiety-inducing effects of ORX-A in the BNST also appear to be dependent on NMDA-type glutamate receptor activity, as pre-injecting the NMDA antagonist AP5 into the BNST blocked anxiogenic effects of local ORX-A injections. Injections of AMPA-type receptor antagonists into the BNST prior to ORX-A resulted in only a partial attenuation of anxiety-like behaviors.


Subject(s)
Anxiety/chemically induced , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , Receptors, Glutamate/drug effects , Septal Nuclei/drug effects , Animals , Anxiety/physiopathology , Male , Maze Learning/drug effects , Orexins , Rats , Rats, Sprague-Dawley , Receptors, Glutamate/physiology , Septal Nuclei/physiology , Social Behavior
SELECTION OF CITATIONS
SEARCH DETAIL
...