Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 175
1.
Health Expect ; 25(4): 1904-1918, 2022 08.
Article En | MEDLINE | ID: mdl-35672924

INTRODUCTION: Long-term benzodiazepine receptor agonist (BZRA) use persists in healthcare settings worldwide and poses risks of patient harm. OBJECTIVE: This study aimed to develop an intervention to support discontinuation of long-term BZRA use among willing individuals. METHODS: The intervention development process aligned with the UK Medical Research Council's complex intervention framework. This involved a previous systematic review of brief interventions targeting long-term BZRA use in primary care and qualitative interviews based on the Theoretical Domains Framework that explored barriers and facilitators to discontinuing long-term BZRA use. A codesign approach was used involving an active partnership between experts by experience, researchers and clinicians. Intervention content was specified in terms of behaviour change techniques (BCTs). RESULTS: The SAFEGUARDING-BZRAs (Supporting sAFE and GradUAl ReDuctIon of loNG-term BenZodiazepine Receptor Agonist uSe) toolkit comprises 24 BCTs and includes recommendations targeted at primary care-based clinicians for operationalizing each BCT to support individuals with BZRA discontinuation. CONCLUSION: The SAFEGUARDING-BZRAs toolkit has been developed using a systematic and theory-based approach that addresses identified limitations of previous research. Further research is needed to assess its usability and acceptability by service users and clinicians, as well as its potential to effectively support safe and gradual reduction of long-term BZRA use. PATIENT OR PUBLIC CONTRIBUTION: The qualitative interview phase included patients as participants. The codesign process included 'experts by experience' with either current or previous experience of long-term BZRA use as collaborators.


GABA-A Receptor Agonists , Behavior Therapy , Benzodiazepines , GABA-A Receptor Agonists/administration & dosage , Humans , Receptors, GABA-A
2.
Health Expect ; 25(1): 355-365, 2022 02.
Article En | MEDLINE | ID: mdl-34862703

INTRODUCTION: Existing interventions to reduce long-term benzodiazepine receptor agonist (BZRA) use lack theoretical underpinning and detailed descriptions. This creates difficulties in understanding how interventions work and how to replicate them in practice. The Theoretical Domains Framework (TDF) can be used to identify behaviour change determinants to target during intervention development. OBJECTIVE: To explore barriers and facilitators to discontinuing BZRA use from the perspective of both current and previous long-term BZRA users. DESIGN/SETTING AND PARTICIPANTS: Semistructured TDF-based interviews were conducted with community-based individuals with current or previous experience of long-term BZRA use. Data were recorded, transcribed and analysed using the framework method. RESULTS: Twenty-eight individuals were interviewed. Despite commonalities in perceived barriers/facilitators to discontinuing BZRA use within individual TDF domains, individual participants had different experiences of identified determinants of BZRA discontinuation. For example, both similarities and differences existed within and between each participant group in terms of knowledge of the appropriate duration of BZRA use ('Knowledge' domain) and experience of withdrawal symptoms ('Reinforcement' domain). Compared to previous users, current users typically anticipated more barriers to discontinuing BZRA use and fewer positive consequences of discontinuation. CONCLUSION: This study reports on barriers and facilitators to discontinuing BZRA use from the perspectives of current and previous long-term users. The findings highlight the challenging nature of BZRA discontinuation and a multitude of barriers that impact participants' behaviour regarding BZRA use. Future work will involve developing a theory-based intervention to support BZRA discontinuation in primary care. PATIENT CONTRIBUTION: The study included patients as participants.


GABA-A Receptor Agonists , Medication Adherence , Qualitative Research , Receptors, GABA-A , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/therapeutic use , Humans , Medication Adherence/psychology , Medication Adherence/statistics & numerical data
3.
J Integr Neurosci ; 20(3): 613-622, 2021 Sep 30.
Article En | MEDLINE | ID: mdl-34645094

As a gamma-aminobutyric acid type A receptor agonist sevoflurane is a common general anesthetic used in anesthesia and affects the neural development in offspring. We hypothesized that sevoflurane could regulate interneurons via the neuregulin-1-epidermal growth factor receptor-4 (NRG1-ErbB4) pathway in the entorhinal cortex (ECT) of the middle pregnancy. Six female rats in middle pregnancy (14.5 days of pregnancy) were randomly and equally divided into sevoflurane (SeV) and control groups. The rats in the SeV group were exposed to 4% sevoflurane for 3 hours. The expression levels of NRG1 and ErbB4, parvalbumin (PV) and glutamic acid decarboxylase (GAD67), and N-methyl-D-aspartate receptor subunit 2A (NR2A) and subunit 2B (NR2B) in offspring were examined through immunohistochemistry. The pyramidal neurons in the ECT were examined via Golgi staining. The levels of NRG1 and ErbB4 were significantly decreased (P < 0.01) and the levels of PV and GAD67 (interneurons) were found to be decreased in the SeV group (P < 0.01). The level of NR2B was found to be increased while the level of NR2A being decreased in the SeV group (P < 0.01). The development of pyramidal neurons was abnormal in the SeV group (P < 0.05). Conclusively, prenatal sevoflurane exposure could lead to the disturbance of the interneurons by activating the NRG1-ErbB4 pathway and subsequently result in abnormal development of pyramidal neurons in middle pregnancy. Prenatal sevoflurane exposure in middle pregnancy could be potentially harmful to the neural development of rat offspring. This study may reveal a novel pathway in the influence mechanism of sevoflurane on rat offspring.


Entorhinal Cortex/drug effects , GABA-A Receptor Agonists/pharmacology , Interneurons/drug effects , Neuregulin-1/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Pyramidal Cells/drug effects , Receptor, ErbB-4/drug effects , Sevoflurane/pharmacology , Animals , Disease Models, Animal , Female , GABA-A Receptor Agonists/administration & dosage , Pregnancy , Rats , Sevoflurane/administration & dosage
4.
Mol Brain ; 14(1): 139, 2021 09 10.
Article En | MEDLINE | ID: mdl-34507588

Chronic pain easily leads to concomitant mood disorders, and the excitability of anterior cingulate cortex (ACC) pyramidal neurons (PNs) is involved in chronic pain-related anxiety. However, the mechanism by which PNs regulate pain-related anxiety is still unknown. The GABAergic system plays an important role in modulating neuronal activity. In this paper, we aimed to study how the GABAergic system participates in regulating the excitability of ACC PNs, consequently affecting chronic inflammatory pain-related anxiety. A rat model of CFA-induced chronic inflammatory pain displayed anxiety-like behaviors, increased the excitability of ACC PNs, and reduced inhibitory presynaptic transmission; however, the number of GAD65/67 was not altered. Interestingly, intra-ACC injection of the GABAAR agonist muscimol relieved anxiety-like behaviors but had no effect on chronic inflammatory pain. Intra-ACC injection of the GABAAR antagonist picrotoxin induced anxiety-like behaviors but had no effect on pain in normal rats. Notably, chemogenetic activation of GABAergic neurons in the ACC alleviated chronic inflammatory pain and pain-induced anxiety-like behaviors, enhanced inhibitory presynaptic transmission, and reduced the excitability of ACC PNs. Chemogenetic inhibition of GABAergic neurons in the ACC led to pain-induced anxiety-like behaviors, reduced inhibitory presynaptic transmission, and enhanced the excitability of ACC PNs but had no effect on pain in normal rats. We demonstrate that the GABAergic system mediates a reduction in inhibitory presynaptic transmission in the ACC, which leads to enhanced excitability of pyramidal neurons in the ACC and is associated with chronic inflammatory pain-related anxiety.


Anxiety/physiopathology , Chronic Pain/physiopathology , GABAergic Neurons/physiology , Gyrus Cinguli/physiopathology , Inflammation/psychology , Pyramidal Cells/physiology , Animals , Anti-Anxiety Agents/administration & dosage , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/etiology , Central Nervous System Sensitization/drug effects , Chronic Pain/psychology , Clozapine/therapeutic use , Freund's Adjuvant/toxicity , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Agonists/therapeutic use , GABA-A Receptor Antagonists/administration & dosage , GABA-A Receptor Antagonists/pharmacology , GABA-A Receptor Antagonists/toxicity , GABAergic Neurons/enzymology , Genetic Vectors/pharmacology , Inflammation/chemically induced , Inflammation/physiopathology , Injections , Interneurons/drug effects , Male , Muscimol/administration & dosage , Muscimol/pharmacology , Muscimol/therapeutic use , Open Field Test , Pain Threshold/drug effects , Patch-Clamp Techniques , Picrotoxin/toxicity , Presynaptic Terminals/drug effects , Presynaptic Terminals/physiology , Pyramidal Cells/enzymology , Rats , Rats, Sprague-Dawley
5.
J Clin Psychopharmacol ; 41(4): 414-420, 2021.
Article En | MEDLINE | ID: mdl-34181362

PURPOSE/BACKGROUND: This study was designed as an early assessment of the safety of the orexin receptor antagonist suvorexant, but also included exploratory assessments of balance and psychomotor performance that are the focus of this report. METHODS/PROCEDURES: This was a double-blind, randomized, 3-period, crossover, phase 1 study. Balance and psychomotor performance were evaluated during the night in 12 healthy elderly participants after bedtime administration of suvorexant 30 mg (a supratherapeutic dose), the GABAergic agonist zolpidem 5 mg (the recommended dose in the elderly), or placebo. Balance (body sway measured by platform stability) and psychomotor performance (measured by choice reaction time) were assessed predose and at 1.5, 4, and 8 hours postdose in each period. Memory (measured by word recall) was assessed predose and at 4 hours postdose. FINDINGS/RESULTS: At 1.5 hours after nighttime administration of each drug (the approximate time of their anticipated maximal plasma concentrations), both zolpidem and suvorexant increased body sway versus placebo, with a greater increase for zolpidem than suvorexant. Suvorexant increased choice reaction time compared with placebo or zolpidem at 1.5 hours. There were no treatment differences on body sway or choice reaction time at 4 or 8 hours, or on word recall at 4 hours. IMPLICATIONS/CONCLUSIONS: These exploratory data suggest that a 30-mg dose of suvorexant (supratherapeutic) and a 5-mg dose of zolpidem (recommended dose in the elderly) impaired balance at 1.5 hours in healthy elderly people, with potentially less impairment for suvorexant relative to zolpidem, but no treatment differences on body sway or psychomotor performance at 4 and 8 hours. Because of their exploratory nature, these findings and their clinical relevance, if any, require confirmation in a prospective study.


Azepines , Memory/drug effects , Postural Balance/drug effects , Psychomotor Performance/drug effects , Triazoles , Zolpidem , Aged , Azepines/administration & dosage , Azepines/adverse effects , Cross-Over Studies , Double-Blind Method , Drug Chronotherapy , Drug Monitoring/methods , Female , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/adverse effects , Healthy Volunteers , Humans , Male , Neuropsychological Tests , Orexin Receptor Antagonists/administration & dosage , Orexin Receptor Antagonists/adverse effects , Reaction Time/drug effects , Sleep Aids, Pharmaceutical/administration & dosage , Sleep Aids, Pharmaceutical/adverse effects , Triazoles/administration & dosage , Triazoles/adverse effects , Zolpidem/administration & dosage , Zolpidem/adverse effects
6.
J Neurophysiol ; 125(6): 2068-2083, 2021 06 01.
Article En | MEDLINE | ID: mdl-33826443

The caudal fastigial nuclei (cFN) are the output nuclei by which the medio-posterior cerebellum influences the production of saccades toward a visual target. On the basis of the organization of their efferences to the premotor burst neurons and the bilateral control of saccades, the hypothesis was proposed that the same unbalanced activity accounts for the dysmetria of all saccades during cFN unilateral inactivation, regardless of whether the saccade is horizontal, oblique, or vertical. We further tested this hypothesis by studying, in two head-restrained macaques, the effects of unilaterally inactivating the caudal fastigial nucleus on saccades toward a target moving vertically with a constant, increasing or decreasing speed. After local muscimol injection, vertical saccades were deviated horizontally toward the injected side with a magnitude that increased with saccade size. The ipsipulsion indeed depended on the tested target speed but not its instantaneous value because it did not increase (decrease) when the target accelerated (decelerated). By subtracting the effect on contralesional horizontal saccades from the effect on ipsilesional ones, we found that the net bilateral effect on horizontal saccades was strongly correlated with the effect on vertical saccades. We explain how this correlation corroborates the bilateral hypothesis and provide arguments against the suggestion that the instantaneous saccade velocity would somehow be "encoded" by the discharge of Purkinje cells in the oculomotor vermis.NEW & NOTEWORTHY Besides causing dysmetric horizontal saccades, unilateral inactivation of caudal fastigial nucleus causes an ipsipulsion of vertical saccades. This study is the first to quantitatively describe this ipsipulsion during saccades toward a moving target. By subtracting the effects on contralesional (hypometric) and ipsilesional (hypermetric) horizontal saccades, we find that this net bilateral effect is strongly correlated with the ipsipulsion of vertical saccades, corroborating the suggestion that a common disorder affects all saccades.


Cerebellar Nuclei/physiology , GABA-A Receptor Agonists/pharmacology , Motion Perception/physiology , Muscimol/pharmacology , Ocular Motility Disorders/physiopathology , Saccades/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cerebellar Nuclei/drug effects , Disease Models, Animal , Eye-Tracking Technology , GABA-A Receptor Agonists/administration & dosage , Macaca mulatta , Male , Motion Perception/drug effects , Muscimol/administration & dosage , Ocular Motility Disorders/chemically induced , Saccades/drug effects
7.
Brain Res ; 1762: 147440, 2021 07 01.
Article En | MEDLINE | ID: mdl-33745922

Dysfunctional modulation of brain circuits that regulate the emotional response to potentially threatening stimuli is associated to an inappropriate representation of the emotional salience. Reduced top-down control by cortical areas is assumed to underlie several behavioral abnormalities including aggression and anxiety related behaviors. Previous studies have identified disrupted GABA signaling in the anterior cingulate cortex (ACC) as a possible mechanism underlying the top-down regulation of aggression and anxiety. In this study, we investigate a role for GABA-A receptor in the ACC in the regulation of aggression and anxiety related behaviors in socially isolated mice. We evaluated the effects of site directed injections of the GABA-A receptor agonist, muscimol or the GABA-A receptor antagonist, bicuculline into the ACC on these behaviors. Results showed that hyper-aggressive behavior, the anxiety and avoidance behavior in socially isolated mice were increased by muscimol microinfusion into ACC, while the sociability was not affected. In contrast, hyper-aggressive behavior in socially isolated mice was inhibited following bicuculline microinfusion without affecting anxiety. Furthermore, microinfusion of bicuculline into ACC decreased avoidance intensity and significantly reinforced social behavior, suggesting that GABA-A receptor inhibition in ACC specifically regulated aggression and sociability. Together, our results confirm a role for GABA-A receptor signaling in the ACC in the regulation of aggressive, social and anxiety related behaviors in socially isolated mice.


Aggression/physiology , Anxiety/metabolism , Gyrus Cinguli/metabolism , Receptors, GABA-A/metabolism , Signal Transduction/physiology , Social Isolation , Aggression/drug effects , Aggression/psychology , Animals , Anxiety/psychology , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , Gyrus Cinguli/drug effects , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Microinjections , Signal Transduction/drug effects , Social Isolation/psychology
8.
Eur J Clin Pharmacol ; 77(6): 903-912, 2021 Jun.
Article En | MEDLINE | ID: mdl-33410969

PURPOSE: To examine the impact of new controlled drugs legislation introduced in May 2017 on benzodiazepine receptor agonist (BZRA) prescribing in Ireland. METHODS: A repeated cross-sectional analysis was conducted using publically available monthly pharmacy claims data from the General Medical Services (GMS) database. The study population comprised all GMS-eligible individuals aged ≥ 16 years from January 2016 to September 2019. Monthly prevalence rates of individuals receiving BZRA prescriptions per 10,000 eligible population were calculated and trends examined over time. Segmented linear regression of prevalence rates was used to examine changes before and after introduction of the legislation stratified by gender and age groups. Regression coefficients (ß) and 95% confidence intervals (CIs) for monthly change were calculated. RESULTS: Pre-legislation (January 2016 to April 2017), there was a significant monthly decline in benzodiazepine prevalence rate (ß = - 1.18; 95% CI - 1.84, - 0.51; p < 0.001) but no significant change in Z-drug prescribing. Post-legislation (May 2017 to September 2019), increases in prevalence rates were observed for benzodiazepines (ß = 1.04; 95% CI 0.17, 1.92; p = 0.021) and Z-drugs (ß = 1.04; 95% CI 0.26, 1.83; p = 0.010). Post-legislation trends showed increases in BZRA prevalence rates among the youngest subgroup (16-44 years), with variable changes in the middle-aged subgroup (45-64 years) and no changes in the oldest subgroup (≥ 65 years). CONCLUSIONS: This study indicates that introduction of new legislation had limited impact on BZRA prescribing on the main public health scheme in Ireland. Interventions targeting specific population subgroups may be required to achieve sustained reductions in prescribing.


Benzodiazepines/administration & dosage , Drug and Narcotic Control/statistics & numerical data , GABA-A Receptor Agonists/administration & dosage , Practice Patterns, Physicians'/statistics & numerical data , Adolescent , Adult , Age Factors , Aged , Cross-Sectional Studies , Drug Misuse/prevention & control , Female , Humans , Ireland , Male , Middle Aged , Sex Factors , Young Adult
9.
Exp Anim ; 70(1): 101-107, 2021 Feb 06.
Article En | MEDLINE | ID: mdl-33071272

Drug interactions are significant in anesthesiology because drug combinations can potentially possess novel properties. The pharmacological advantages of a new combination of the benzodiazepine receptor agonist JM-1232(-) and propofol were investigated in mice. Male adult mice were administered JM-1232(-) or propofol or combinations of the two drugs intravenously. Loss of the righting reflex was evaluated as achieving hypnosis, and the time until recovery of the reflex was measured as hypnosis time. After determining the ED50, doses double and triple the ED50 of propofol were injected with JM-1232(-) to compare hypnosis time. The injections were repeated four times, and the hypnosis times were compared. Flumazenil was administered separately immediately after the last dose was injected. The ED50 values ([95% confidence interval]) for hypnosis were 3.76 [3.36-4.10] for JM-1232(-) and 9.88 [8.03-11.58] mg kg-1 for propofol. Co-administration of 0.5 and 1 mg kg-1 JM-1232(-) reduced the ED50 values of propofol to 1.76 [1.21-2.51] and 1.00 [0.46-1.86] mg kg-1, respectively. The drug combination for hypnosis produced a supra-additive interaction. Hypnosis time was significantly shorter in the groups given the mixtures compared to each hypnotic administered alone. After repeated injections, hypnosis time with the mixtures showed smaller prolongation than that with the hypnotic alone. Flumazenil completely restored the recovery time after anesthesia. The combination of JM-1232(-) and propofol showed a supra-additive interaction, and the reduced hypnotic dose contributed to a faster recovery even after multiple injections.


GABA-A Receptor Agonists , Hypnotics and Sedatives/administration & dosage , Isoindoles/administration & dosage , Piperazines/administration & dosage , Propofol/administration & dosage , Anesthesia Recovery Period , Animals , Dose-Response Relationship, Drug , Drug Combinations , Drug Interactions , Flumazenil/pharmacology , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacology , Hypnotics and Sedatives/pharmacology , Infusions, Intravenous , Isoindoles/pharmacology , Male , Mice, Inbred Strains , Piperazines/pharmacology , Propofol/pharmacology
10.
Neuropsychopharmacol Rep ; 41(1): 14-25, 2021 03.
Article En | MEDLINE | ID: mdl-33259705

AIMS: Benzodiazepine receptor agonists (BZ-RAs) are frequently prescribed to treat insomnia; however, their long-term use is not recommended. To introduce an appropriate pharmaco-therapy, the current state and background factors of BZ-RAs' dependence must be elucidated. In this study, we developed a Japanese version of the Benzodiazepine Dependence Self-Report Questionnaire (Bendep-SRQ-J) and conducted a study of BZ-RAs' use disorder. METHODS: The Bendep-SRQ-J was created with permission from the original developer. Subjects were inpatients and outpatients receiving BZ-RAs between 2012 and 2013. Clinical data collected were Bendep-SRQ-J scores, sleep disorders for which BZ-RAs were prescribed, physical comorbidities, psychotropic drugs, and lifestyle factors. Logistic analysis was performed to extract factors associated with severe symptoms. RESULTS: Of the 707 patients prescribed BZ-RAs, 324 had voluntarily tapered or discontinued their drugs. Logistic analysis showed that the total number of drugs administered in the last 6 months correlated with both worsening of symptoms or conditions. This was more notable among younger patients, and the proportion of patients with severe symptoms or conditions increased with the increasing number of drugs. CONCLUSION: Using the Bendep-SRQ-J, we elucidated the current state of BZ-RA dependence. Nearly half of the patients were non-compliant. The proportion of patients with severe symptoms or disease conditions increased with the increase in the number of drugs administered. These findings highlight the need for clinicians to be aware of the likelihood of benzodiazepine dependence, especially in young patients and patients prescribed multiple hypnotics.


Anti-Anxiety Agents/administration & dosage , Benzodiazepines/administration & dosage , Drug Tapering , GABA-A Receptor Agonists/administration & dosage , Hypnotics and Sedatives/administration & dosage , Mental Disorders/drug therapy , Patient Compliance , Polypharmacy , Psychometrics/instrumentation , Substance-Related Disorders/diagnosis , Adult , Aged , Aged, 80 and over , Cross-Sectional Studies , Drug Tapering/statistics & numerical data , Female , Health Surveys , Humans , Japan/epidemiology , Male , Mental Disorders/epidemiology , Middle Aged , Patient Compliance/statistics & numerical data , Self Report , Severity of Illness Index , Substance Withdrawal Syndrome/diagnosis , Substance Withdrawal Syndrome/epidemiology , Substance-Related Disorders/epidemiology , Young Adult
11.
Laryngoscope ; 131(10): 2187-2198, 2021 10.
Article En | MEDLINE | ID: mdl-33146426

OBJECTIVES: We aimed to examine the effect of unilateral inhibition of the medullary dorsal swallowing networks on the activities of swallowing-related cranial motor nerves and swallowing interneurons. METHODS: In 25 juvenile rats, we recorded bilateral vagal nerve activity (VNA) as well as unilateral phrenic and hypoglossal activity (HNA) during fictive swallowing elicited by electrical stimulation of the superior laryngeal nerve during control and following microinjection of the GABA agonist muscimol into the caudal dorsal medulla oblongata in a perfused brainstem preparation. In 20 animals, swallowing interneurons contralateral to the muscimol injection side were simultaneously recorded extracellularly and their firing rates were analyzed during swallowing. RESULTS: Integrated VNA and HNA to the injection side decreased to 49.0 ± 16.6% and 32.3 ± 17.9%, respectively. However, the VNA on the uninjected side showed little change after muscimol injection. Following local inhibition, 11 out of 20 contralateral swallowing interneurons showed either increased or decreased of their respective firing discharge during evoked-swallowing, while no significant changes in activity were observed in the remaining nine neurons. CONCLUSION: The neuronal networks underlying the swallowing pattern generation in the dorsal medulla mediate the ipsilateral motor outputs and modulate the contralateral activity of swallowing interneurons, suggesting that the bilateral coordination of the swallowing central pattern generator regulates the spatiotemporal organization of pharyngeal swallowing movements. LEVEL OF EVIDENCE: NA Laryngoscope, 131:2187-2198, 2021.


Deglutition/physiology , GABA-A Receptor Agonists/administration & dosage , Medulla Oblongata/physiology , Pharynx/physiology , Vagus Nerve/physiology , Animals , Deglutition/drug effects , Electric Stimulation , Hypoglossal Nerve/drug effects , Hypoglossal Nerve/physiology , Male , Medulla Oblongata/drug effects , Microinjections , Models, Animal , Muscimol/administration & dosage , Nerve Net/drug effects , Nerve Net/physiology , Neurons/physiology , Pharynx/innervation , Rats , Spatio-Temporal Analysis , Vagus Nerve/drug effects
12.
Behav Brain Res ; 401: 113077, 2021 03 05.
Article En | MEDLINE | ID: mdl-33345825

Food neophobia is a behavior observed in rodents involving reduced consumption of a novel food or drink. In the absence of negative post-ingestive consequences, consumption increases with exposure (attenuation of neophobia), which is seen as an associative safe memory. Olfaction and gustation are sensory modalities essential for the development of a food preference. However, little is known about the neural mechanisms underlying neophobia to a food-related odor stimulus. In the present study, we examined the effect of pharmacological inactivation of the ventral hippocampus (vHPC) on neophobia to orally consumed solutions in rats using muscimol, a gamma aminobutyric acid type A receptor agonist. Two different types of solutions, almond odor (benzaldehyde) and sweet taste (saccharin), were prepared. In the results, microinjections of muscimol into the bilateral vHPC before the first odor and taste exposures did not alter the neophobic reactions of the rats to each stimulus. However, in the second odor, but not taste, exposure, the muscimol-injected rats showed higher consumption in comparison to that observed in the control rats, suggesting that the vHPC inactivation facilitates the attenuation of odor neophobia. On the other hand, intra-vHPC muscimol microinjections after the first odor and taste exposures did not facilitate consumption at the second exposures. These results indicate that neural activations within vHPC during orally consuming a novel odor, but not taste, solution play an inhibitory role in the subsequent attenuation of neophobia.


Behavior, Animal/physiology , Food Preferences/physiology , GABA-A Receptor Agonists/pharmacology , Hippocampus/drug effects , Muscimol/pharmacology , Olfactory Perception/physiology , Taste Perception/physiology , Animals , Behavior, Animal/drug effects , Food Preferences/drug effects , GABA-A Receptor Agonists/administration & dosage , Male , Muscimol/administration & dosage , Olfactory Perception/drug effects , Rats , Rats, Wistar , Taste Perception/drug effects
13.
Behav Brain Res ; 403: 113094, 2021 04 09.
Article En | MEDLINE | ID: mdl-33359845

Autism spectrum disorder (ASD) is an immensely challenging developmental disorder characterized primarily by two core behavioral symptoms of social communication deficits and restricted/repetitive behaviors. Investigating the etiological process and identifying an appropriate therapeutic target remain as formidable challenges to overcome ASD due to numerous risk factors and complex symptoms associated with the disorder. Among the various mechanisms that contribute to ASD, the maintenance of excitation and inhibition balance emerged as a key factor to regulate proper functioning of neuronal circuitry. In this study, we employed prenatally exposed to valproic acid (VPA) to establish a validated ASD mouse model and found impaired inhibitory gamma-aminobutyric acid (GABAergic) neurotransmission through a presynaptic mechanism in these model mice, which was accompanied with decreased GABA release and GABA-A and GABA-B receptor subunits expression. And acute administration of individual GABA-A or GABA-B receptor agonists partially reversed autistic-like behaviors in the model mice. Furthermore, acute administration of the combined GABA-A and GABA-B receptor agonists palliated sociability deficits, anxiety and repetitive behaviors in the animal model of autistic-like behaviors, demonstrating the therapeutic potential of above cocktail in the treatment of ASD.


Autism Spectrum Disorder/drug therapy , Behavior, Animal/drug effects , GABA-A Receptor Agonists/pharmacology , GABA-B Receptor Agonists/pharmacology , Prefrontal Cortex/drug effects , Prenatal Exposure Delayed Effects/drug therapy , Synaptic Potentials/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/drug effects , Animals , Anticonvulsants/pharmacology , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/physiopathology , Disease Models, Animal , Drug Therapy, Combination , Female , GABA-A Receptor Agonists/administration & dosage , GABA-B Receptor Agonists/administration & dosage , Male , Mice , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/physiopathology , Valproic Acid/pharmacology
14.
Behav Brain Res ; 398: 112967, 2021 02 01.
Article En | MEDLINE | ID: mdl-33075397

The postpartum period is commonly accompanied by emotional changes, which for many new mothers includes a reduction in anxiety. Previous research in rodents has shown that the postpartum attenuation in anxiety is dependent on offspring contact and has further implicated enhanced GABAergic neurotransmission as an underlying mechanism. However, the specific brain regions where GABA acts to regulate the offspring-induced reduction in postpartum anxiety requires further investigation. Here, we test the hypothesis that offspring interactions suppress anxiety-like behavior in postpartum female rats via GABA signaling in the medial prefrontal cortex (mPFC). Our results show a postpartum reduction in anxiety-like behavior, an effect which was abolished by localized infusion of the GABAA receptor antagonist bicuculline in the mPFC. We also show that activation of GABAA receptors in the mPFC by the agonist muscimol was effective in restoring anxiolyisis in mothers separated from their pups. Lastly, we show that heightened anxiety-like behavior in pup-separated mothers was accompanied by a lower number and percentage of activated GABAergic neurons within the mPFC. Together, these results suggest that mother-offspring interactions reduce anxiety-like behavior in postpartum females via GABAA neurotransmission in the mPFC and in doing so provide insight into mechanisms that may become dysfunctional in mothers who experience high postpartum anxiety.


Anxiety/metabolism , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/pharmacology , GABAergic Neurons/physiology , Maternal Deprivation , Prefrontal Cortex/physiology , Puerperal Disorders/metabolism , gamma-Aminobutyric Acid/physiology , Animals , Anxiety/drug therapy , Behavior, Animal/drug effects , Behavior, Animal/physiology , Bicuculline/pharmacology , Disease Models, Animal , Female , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , Humans , Male , Muscimol/pharmacology , Prefrontal Cortex/metabolism , Puerperal Disorders/drug therapy , Rats, Sprague-Dawley
15.
Nat Commun ; 11(1): 4929, 2020 10 01.
Article En | MEDLINE | ID: mdl-33004789

Non-invasive, molecularly-specific, focal modulation of brain circuits with low off-target effects can lead to breakthroughs in treatments of brain disorders. We systemically inject engineered ultrasound-controllable drug carriers and subsequently apply a novel two-component Aggregation and Uncaging Focused Ultrasound Sequence (AU-FUS) at the desired targets inside the brain. The first sequence aggregates drug carriers with millimeter-precision by orders of magnitude. The second sequence uncages the carrier's cargo locally to achieve high target specificity without compromising the blood-brain barrier (BBB). Upon release from the carriers, drugs locally cross the intact BBB. We show circuit-specific manipulation of sensory signaling in motor cortex in rats by locally concentrating and releasing a GABAA receptor agonist from ultrasound-controlled carriers. Our approach uses orders of magnitude (1300x) less drug than is otherwise required by systemic injection and requires very low ultrasound pressures (20-fold below FDA safety limits for diagnostic imaging). We show that the BBB remains intact using passive cavitation detection (PCD), MRI-contrast agents and, importantly, also by sensitive fluorescent dye extravasation and immunohistochemistry.


Blood-Brain Barrier/metabolism , Brain Diseases/drug therapy , Drug Carriers/radiation effects , GABA-A Receptor Agonists/administration & dosage , Ultrasonography, Interventional/methods , Animals , Blood-Brain Barrier/diagnostic imaging , Blood-Brain Barrier/radiation effects , Dose-Response Relationship, Radiation , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Female , GABA-A Receptor Agonists/pharmacokinetics , Humans , Magnetic Resonance Imaging , Models, Animal , Muscimol/administration & dosage , Muscimol/pharmacokinetics , Rats , Stereotaxic Techniques , Ultrasonic Waves
16.
Psychopharmacology (Berl) ; 237(12): 3759-3771, 2020 Dec.
Article En | MEDLINE | ID: mdl-32875348

RATIONALE: Compulsive cocaine use, defined as the continued use despite the dire consequences, is a hallmark of cocaine addiction. Thus, understanding the brain mechanism regulating the compulsive cocaine-seeking and cocaine-taking behaviors is essential to understand cocaine addiction and the key to identification of the molecular targets for the development of medications against this condition. OBJECTIVE: This study aimed to determine how the GABAa and GABAb receptors of the central nucleus of the amygdala (CeA) regulate the compulsive cocaine-seeking behavior. METHODS: Male Wistar outbred rats were trained to self-administer intravenous cocaine (0.4 mg/kg/infusion) under a chained schedule. The compulsive cocaine-seeking behavior was measured as the cocaine-seeking behavior in the face of footshock punishment. The role of the GABA receptors of CeA in the regulation of such behavior was determined by measuring the dose-dependent effects of the GABAa agonist muscimol or the GABAb agonist baclofen bilaterally microinjected into the CeA on the punished cocaine-seeking behavior. RESULTS: The cocaine-seeking behavior was inhibited by footshock punishment in an intensity-dependent manner. Both muscimol and baclofen dose-dependently increased the punished cocaine-seeking behavior. However, the potency of muscimol but not baclofen was negatively correlated with the effects of punishment. CONCLUSION: These data indicate that the CeA GABAa receptors play a key role in the regulation of the compulsive cocaine-seeking behavior and suggest that an increase in the function of the GABAa receptors possibly induced by cocaine or genetic factors may be an important mechanism involved in the development of or vulnerability to the compulsive cocaine use and addiction.


Behavior, Addictive/psychology , Central Amygdaloid Nucleus/physiology , Cocaine/administration & dosage , Compulsive Behavior/psychology , Receptors, GABA-A/physiology , Receptors, GABA-B/physiology , Animals , Baclofen/administration & dosage , Behavior, Addictive/drug therapy , Central Amygdaloid Nucleus/drug effects , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/psychology , Compulsive Behavior/drug therapy , GABA-A Receptor Agonists/administration & dosage , GABA-B Receptor Agonists/administration & dosage , Male , Microinjections , Muscimol/administration & dosage , Rats , Rats, Wistar , Self Administration
17.
Neuropharmacology ; 179: 108275, 2020 11 15.
Article En | MEDLINE | ID: mdl-32835765

The amygdala is a key component of the neural circuits mediating the processing and response to emotionally salient stimuli. Amygdala lesions dysregulate social interactions, responses to fearful stimuli, and autonomic functions. In rodents, the basolateral and central nuclei of the amygdala have divergent roles in behavioral control. However, few studies have selectively examined these nuclei in the primate brain. Moreover, the majority of non-human primate studies have employed lesions, which only allow for unidirectional manipulation of amygdala activity. Thus, the effects of amygdala disinhibition on behavior in the primate are unknown. To address this gap, we pharmacologically inhibited by muscimol or disinhibited by bicuculline methiodide the basolateral complex of the amygdala (BLA; lateral, basal, and accessory basal) in nine awake, behaving male rhesus macaques (Macaca mulatta). We examined the effects of amygdala manipulation on: (1) behavioral responses to taxidermy snakes and social stimuli, (2) food competition and social interaction in dyads, (3) autonomic arousal as measured by cardiovascular response, and (4) prepulse inhibition of the acoustic startle (PPI) response. All modalities were impacted by pharmacological inhibition and/or disinhibition. Amygdala inhibition decreased fear responses to snake stimuli, increased examination of social stimuli, reduced competitive reward-seeking in dominant animals, decreased heart rate, and increased PPI response. Amygdala disinhibition restored fearful response after habituation to snakes, reduced competitive reward-seeking behavior in dominant animals, and lowered heart rate. Thus, both hypoactivity and hyperactivity of the basolateral amygdala can lead to dysregulated behavior, suggesting that a narrow range of activity is necessary for normal functions.


Amygdala/drug effects , Emotions/drug effects , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Antagonists/administration & dosage , Heart Rate/drug effects , Social Interaction/drug effects , Acoustic Stimulation/methods , Amygdala/diagnostic imaging , Amygdala/physiology , Animals , Emotions/physiology , Fear/drug effects , Fear/physiology , Fear/psychology , Heart Rate/physiology , Injections, Intraventricular , Macaca mulatta , Male , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Snakes
18.
Sci Rep ; 10(1): 11401, 2020 07 09.
Article En | MEDLINE | ID: mdl-32647241

Urodynamic studies, used to understand bladder function, diagnose bladder disease, and develop treatments for dysfunctions, are ideally performed with awake subjects. However, in small and medium-sized animal models, anesthesia is often required for these procedures and can be a research confounder. This study compared the effects of select survival agents (dexmedetomidine, alfaxalone, and propofol) on urodynamic (Δpressure, bladder capacity, bladder compliance, non-voiding contractions, bladder pressure slopes) and anesthetic (change in heart rate [∆HR], average heart rate [HR], reflexes, induction/recovery times) parameters in repeated cystometrograms across five adult male cats. The urodynamic parameters under isoflurane and α-chloralose were also examined in terminal procedures for four cats. Δpressure was greatest with propofol, bladder capacity was highest with α-chloralose, non-voiding contractions were greatest with α-chloralose. Propofol and dexmedetomidine had the highest bladder pressure slopes during the initial and final portions of the cystometrograms respectively. Cats progressed to a deeper plane of anesthesia (lower HR, smaller ΔHR, decreased reflexes) under dexmedetomidine, compared to propofol and alfaxalone. Time to induction was shortest with propofol, and time to recovery was shortest with dexmedetomidine. These agent-specific differences in urodynamic and anesthetic parameters in cats will facilitate appropriate study-specific anesthetic choices.


Anesthetics/pharmacology , Cats/physiology , Urodynamics/drug effects , Anesthesia Recovery Period , Anesthetics/administration & dosage , Animals , Chloralose/pharmacology , Dexmedetomidine/administration & dosage , Dexmedetomidine/pharmacology , Dose-Response Relationship, Drug , GABA-A Receptor Agonists/administration & dosage , GABA-A Receptor Agonists/pharmacology , Heart Rate/drug effects , Isoflurane/pharmacology , Male , Models, Animal , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Pregnanediones/administration & dosage , Pregnanediones/pharmacology , Pressure , Propofol/administration & dosage , Propofol/pharmacology , Urinary Bladder/drug effects , Urinary Bladder/physiology
19.
Psychopharmacology (Berl) ; 237(11): 3237-3247, 2020 Nov.
Article En | MEDLINE | ID: mdl-32666257

RATIONALE: The development of effective anxiety treatments has been hindered by limited understanding of the neurobiological mechanisms involved in anxiety regulation. Whilst gamma-aminobutyric acid (GABA) neurotransmission in the prefrontal cortex (PFC) is one mechanism consistently implicated in anxiety regulation, PFC subregions may contribute uniquely. OBJECTIVES: The present study examined the effects of inactivating the PFC subregions of the prelimbic cortex (PrL) or orbitofrontal cortex (OFC) through GABAA receptor (GABAAR) activation, on anxiety behaviours in male Wistar rats. METHODS: Sixty-six male Wistar rats were surgically implanted with bilateral cannulae into the PrL (n = 33) or the OFC (n = 33). Rats then received a microinjection of either the GABAA receptor agonist muscimol or vehicle prior to each experiment, conducted 1 week apart. Measures of anxiety were examined using the elevated plus maze (EPM) and the emergence test (ET). The effect on locomotor activity (baseline or methamphetamine-induced) was also tested. RESULTS: Differential effects of brain region inactivation on anxiety-like behaviour were shown by measures in the EPM and ET; muscimol infused into the PrL-reduced anxiety-like behaviour, yet had no significant effect when infused into the OFC, compared with control treated rats. No effects on locomotor activity at baseline or following methamphetamine treatment were found. CONCLUSIONS: This study highlights that activation of GABAARs specifically within the PrL, but not OFC, reduces anxiety behaviours in male rats. This suggests that activity of the PrL plays a more important role than the OFC in the neurobiological mechanisms of unconditioned anxiety and should be targeted for future therapies.


Anxiety/drug therapy , Anxiety/metabolism , GABA-A Receptor Agonists/administration & dosage , Prefrontal Cortex/metabolism , Receptors, GABA-A/metabolism , Animals , Anxiety/psychology , Male , Microinjections/methods , Muscimol/administration & dosage , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Treatment Outcome , gamma-Aminobutyric Acid/administration & dosage
20.
J Neurosci ; 40(33): 6379-6388, 2020 08 12.
Article En | MEDLINE | ID: mdl-32493711

The perception of time is critical to adaptive behavior. While prefrontal cortex and basal ganglia have been implicated in interval timing in the seconds to minutes range, little is known about the role of the mediodorsal thalamus (MD), which is a key component of the limbic cortico-basal ganglia-thalamocortical loop. In this study, we tested the role of the MD in timing, using an operant temporal production task in male mice. In this task, that the expected timing of available rewards is indicated by lever pressing. Inactivation of the MD with muscimol produced rightward shifts in peak pressing on probe trials as well as increases in peak spread, thus significantly altering both temporal accuracy and precision. Optogenetic inhibition of glutamatergic projection neurons in the MD also resulted in similar changes in timing. The observed effects were found to be independent of significant changes in movement. Our findings suggest that the MD is a critical component of the neural circuit for interval timing, without playing a direct role in regulating ongoing performance.SIGNIFICANCE STATEMENT The mediodorsal nucleus (MD) of the thalamus is strongly connected with the prefrontal cortex and basal ganglia, areas which have been implicated in interval timing. Previous work has shown that the MD contributes to working memory and learning of action-outcome contingencies, but its role in behavioral timing is poorly understood. Using an operant temporal production task, we showed that inactivation of the MD significantly impaired timing behavior.


Conditioning, Operant/physiology , Mediodorsal Thalamic Nucleus/physiology , Psychomotor Performance/physiology , Time Perception/physiology , Animals , Conditioning, Operant/drug effects , GABA-A Receptor Agonists/administration & dosage , Male , Mediodorsal Thalamic Nucleus/drug effects , Mice, Inbred C57BL , Muscimol/administration & dosage , Optogenetics , Psychomotor Performance/drug effects , Reward , Time Perception/drug effects
...